You are on page 1of 242

MEDICAL INTELLIGENCE UNIT 23

Friedhelm Beyersdorf

Ischemia-Reperfusion Injury
in Cardiac Surgery
MEDICAL
INTELLIGENCE
UNIT 23

Ischemia-Reperfusion
Injury in Cardiac Surgery

Friedhelm Beyersdorf, M.D.


Department of Cardiovascular Surgery
Albert-Ludwigs-University
Freiburg, Germany

LANDES BIOSCIENCE EUREKAH.COM


GEORGETOWN, TEXAS AUSTIN, TEXAS
U.S.A. U.S.A.
ISCHEMIA-REPERFUSION INJURY IN CARDIAC SURGERY
Medical Intelligence Unit

EUREKAH.COM
LANDES BIOSCIENCE
Designed by Judith Kemper
Copyright ©2001 Eurekah.com
All rights reserved.
No part of this book may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopy, recording, or any information storage and retrieval system,
without permission in writing from the publisher.
Printed in the U.S.A.
Please address all inquiries to the Publishers:
Eurekah.com / Landes Bioscience
810 South Church Street, Georgetown, Texas, U.S.A. 78626
Phone: 512/ 863 7762; FAX: 512/ 863 0081
www.landesbioscience.com
www.Eurekah.com

ISBN 1-58706-002-7 hard cover version


ISBN 1-58706-085-X soft cover version

While the authors, editors and publisher believe that drug selection and dosage and the specifications
and usage of equipment and devices, as set forth in this book, are in accord with current recommend-
ations and practice at the time of publication, they make no warranty, expressed or implied, with
respect to material described in this book. In view of the ongoing research, equipment development,
changes in governmental regulations and the rapid accumulation of information relating to the
biomedical sciences, the reader is urged to carefully review and evaluate the information provided
herein.
Library of Congress Cataloging-in-Publication Data
Ischemia-reperfusion injury in cardiac surgery / [edited by] Friedhelm Beyersdorf.
p. cm. --- (Medical Intelligence unit)
Includes bibliographical references and index.
ISBN 1-58706-002-7 (alk. paper)
1. Heart--Surgery. 2. Myocardial reperfusion. 3. Reperfusion injury. 4. Ischemia.
I. Beyersdorf, Friedhelm. II. Series.
[DNLM: 1. Myocardial Reperfusion Injury--prevention & control. 2. Cardiac
Surgical Procedures--adverse effects. 3. Myocardial Ischemia--prevention & control.
4. Myocardial Reperfusion Injury--physiopathology. WG 280 I774 1999]
RD598.I78 1999
617.4'12--dc21
DNLM/DLC
for Library of Congress 99022059
CIP
CONTENTS
Foreword ............................................................................................... xi
Preface ................................................................................................ xiv

I. Basic Science in Ischemia-Reperfusion Injury


1. Harnessing the Cardioprotective Potential of Nitric Oxide
in Nonsurgical and Surgical Ischemic-Reperfusion Injury ...................... 2
Jakob Vinten-Johansen and Russell S. Ronson
Chemistry of NO• Generation ............................................................... 3
Physiological Actions of NO• Relevant to Ischemia-Reperfusion ........... 5
Decreased NO• Release After Endothelial Damage
from Ischemic-Reperfusion Injury ..................................................... 6
Time Course of Endothelial Injury ........................................................ 7
Endothelial Dysfunction and Neutrophils ........................................... 10
NO•-Related Therapeutic Strategies .................................................... 11
Protection by Nitric Oxide Donors ..................................................... 12
Peroxynitrite: The Potential Dark Side of Nitric Oxide Therapy ......... 15
Nitric Oxide in Hypoxia-Reoxygenation Injury ................................... 19
Nitric Oxide and Myocardial Apoptosis .............................................. 21
Summary ............................................................................................. 21
2. The Cellular Basis of Immediate Lethal Reperfusion Injury ................. 28
H. M. Piper and D. García-Dorado
Lethal Reperfusion Injury: Definition of Terms .................................. 28
A Short Note on the Role of Oxygen Radicals ..................................... 29
Three Initial Causes of Immediate Lethal Reperfusion Injury .............. 30
Following Initial Causes: Spreading of Necrosis .................................. 36
The Problem of Delay of Necrosis and Initiation of Apoptosis ............ 37
Conclusions ......................................................................................... 37

3. Apoptosis in Ischemia—Reperfusion Injury ......................................... 41


Harald Darius, Waltraud Ibe and Michael Buerke
Characteristics of Apoptosis ................................................................. 41
Detection of Apoptosis ........................................................................ 42
Apoptosis in Cardiac Disease ............................................................... 42
Experimental and Clinical Evidence of Apoptosis
in Acute Myocardial Infarction and Reperfusion ............................. 43
Prevention of Apoptosis During Ischemia and Reperfusion ................. 46
4. Changes in Myocardial Gene Expression Following Ischemia
and Reperfusion ................................................................................... 49
John W. C. Entwistle III and Andrew S. Wechsler
Basics of Myocardial Gene Expression ................................................. 50
Mechanisms of Gene Changes in Stunning ......................................... 51
Calcium as a Regulator of Gene Expression ......................................... 51
Alpha-Adrenergic Receptor Stimulation as a Regulator
of Gene Expression .......................................................................... 51
Myocellular Stretchas a Regulator of Gene Expression ......................... 52
Changes in Gene Expression in Ischemia and Stunning ....................... 52
Conclusions ......................................................................................... 56

II. Methods to Prevent Ischemia-Reperfusion Injury


5. The NO-Donor L-Arginine Reduces the Reperfusion Injury
after Heart Transplantation ................................................................. 61
Gábór Szabó
Effects of Nitric Oxide ........................................................................ 61
Reduction of Reperfusion Injury by Nitric Oxide:
Potential Mechanisms ..................................................................... 62

6. Sodium-Proton Exchange Inhibition as a Novel Strategy


for Myocardial Protection .................................................................... 70
Willem Flameng and Wolfgang Scholz
Sodium-Proton Exchange Inhibition in Cardiac
Ischemia-Reperfusion ...................................................................... 70
Effects on Intracellular Na+ ................................................................. 71
Effects on Intracellular Ca++ ............................................................... 72
Effects on Intracellular pH and Ischemic Metabolism ......................... 72
Cardioprotective Effects ...................................................................... 73
Preliminary Clinical Experience with Sodium-Proton Exchange
Inhibition ........................................................................................ 74
7. Metabolic Support for the Heart During Ischemia and Reperfusion:
Role of Amino Acids ............................................................................ 79
Heinrich Taegtmeyer and Torsten Doenst
Energy for Contraction: Cycles Improve Efficiency ............................. 79
Amino Acids: Cycling In and Out of Proteins ..................................... 80
Amino Acid Metabolism in Normal Heart .......................................... 80
Amino Acid Metabolism in Oxygen-Deprived Heart .......................... 83
Amino Acids as Substrates for Metabolic Support
During Reperfusion ......................................................................... 84
Substrate Enhancement of Cardioplegic Solutions
by Amino Acids ............................................................................... 85
Conclusions and Outlook .................................................................... 87
8. Metabolic and Antioxidant Support with Amino Acids ........................ 90
Oleg I. Pisarenko
Glutamate and Aspartate ..................................................................... 90
Other Amino Acids ............................................................................. 93
Conclusion .......................................................................................... 95
9. Methods to Reduce Ischemia/Reperfusion Injury—PICSO ................. 99
Günter Steurer, Katharina Palisek and Werner Mohl
Ischemia/Reperfusion Injury ............................................................... 99
Coronary Sinus Interventions .............................................................. 99
Pressure-Controlled Intermittent Coronary/Sinus
Occlusion (PICSO) ....................................................................... 100
Experimental Studies ......................................................................... 102
Clinical Studies ................................................................................. 102
Future Directions .............................................................................. 104

10. Ischemic Preconditioning—from Bench to Bedside ........................... 106


Torsten Doenst and Heinrich Taegtmeyer
Discovery, Definition and Capabilities .............................................. 106
Limitations of Ischemic Preconditioning ........................................... 106
Effects of Ischemic Preconditioning on Cardiac Metabolism ............. 109
Delivery of Preconditioning—How Is Protection Mediated? ............. 112
Preconditioning Without Preconditioning? ....................................... 116
Clinical Aspects—Implications for the Cardiac Surgeon .................... 117
Conclusions ....................................................................................... 117

11. Away from Ischemic Preconditioning and Towards


Pharmacological Preconditioning ....................................................... 125
Louis P. Perrault and Philippe Menasché
Abstract ............................................................................................. 125
Clinical Trials: An Apparent Discrepancy .......................................... 126

Section III. Intravenous Metabolic Support


12. Intravenous Metabolic Support with GIK
(Glucose-Insulin-Potassium) and Amino Acids in Cardiac Surgery .... 135
Rolf Svedjeholm
Myocardial Substrate Metabolism ..................................................... 136
Metabolic Intervention ...................................................................... 137
GIK (Glucose-Insulin-Potassium) ..................................................... 137
Amino Acids ...................................................................................... 140
Clinical Experience with Metabolic Support in Cardiac Surgery ........ 144
Summary ........................................................................................... 151

Section IV. Surgical Techniques


A. Temperature of Blood Cardioplegia
13. Cold/Tepid Cardioplegia ................................................................... 158
Hendrick B. Barner and Andrew C. Fiore
Goals of Cardioplegia ........................................................................ 159
Current Research ............................................................................... 159
Future Applications ........................................................................... 162
New Research .................................................................................... 162
Anticipated Developments ................................................................ 162

14. Surgical Techniques for Warm Blood Cardioplegia ............................ 165


Syed T. Raza and Tomas A. Salerno
Conceptual Framework ..................................................................... 165
Techniques ........................................................................................ 165
Retrograde Continuous Warm Blood Cardioplegia (RCWBC) ......... 166
Alternate Retrograde/Antegrade Cardioplegia .................................... 166
Simultaneous Antegrade/Retrograde Cardioplegia ............................. 166
Warm Cardioplegia Techniques for Valvular Surgery ........................ 167
Normothermic Perfusion ................................................................... 167
Intermittent Techniques of Warm Blood Cardioplegia ...................... 167
Myocardial Resuscitation ................................................................... 167
Monitoring of Cardioplegia Delivery ................................................. 168
Advantages and Disadvantages of Warm Blood Cardioplegia ............ 168
Technical Considerations in WBC .................................................... 169
Visual Enhancement During WBC ................................................... 169
Inadequate RV Protection with WBC ............................................... 169
Coronary Sinus Injuries ..................................................................... 170
Postoperative Neurological Deficits and Stroke Rates
after WBC and Normothermic Cardiopulmonary Bypass ............. 170
Tepid Cardioplegia ............................................................................ 170
Cold Agglutinins and Warm Blood Cardioplegia .............................. 171
Future Perspectives ............................................................................ 171

15. Interrupting Warm Blood Cardioplegia ............................................. 174


Harold L. Lazar
Experimental Studies ......................................................................... 175
Clinical Studies ................................................................................. 177
Summary and Conclusions ................................................................ 179
B. Protection Strategies

16. Myocardial Protection Strategies in Routine Coronary and Valve


Operations ......................................................................................... 183
Kiyozo Morita and Michio Yoshitake
Integrated Myocardial Management .................................................. 183
Background and Logic of Integrated Myocardial Management .......... 184
Method of Integrated Myocardial Management
and Surgical Techniques ................................................................ 185
Standard Protocol for Integrated Blood Cardioplegia ........................ 190
Special Consideration for the Type of Surgical Procedure ................. 193
17. Acute Myocardial Infarction and Cardiogenic Shock .......................... 196
Francesco Donatelli, Marco Pocar and Adalberto Grossi
Indications for Operation, Preoperative Management
and Surgical Technique ................................................................. 197
Personal Experience and Conclusive Considerations .......................... 200
18. Protection Strategies in Reoperations ................................................. 203
Fumiyuki Okamoto and Keisuke Sakai
Coronary Bypass Reoperations .......................................................... 203
Valve Reoperations ............................................................................ 205
Thoracic Aortic Aneurysm Reoperations ........................................... 207
Summary ........................................................................................... 207
19. Protection Strategies for Heart Transplantation ................................. 210
Juergen Martin, Armin Geiger and Friedhelm Beyersdorf
Preharvesting Phase ........................................................................... 210
Cardioplegic Induction Phase ............................................................ 212
Organ Storage ................................................................................... 212
Reperfusion Phase ............................................................................. 213
Future Directions .............................................................................. 215
Preservation Solutions ....................................................................... 216
Celsior ............................................................................................... 219
Blood Cardioplegia ............................................................................ 220
Conclusions ....................................................................................... 220

Index .................................................................................................. 226


EDITORS
Friedhelm Beyersdorf, M.D.
Department of Cardiovascular Surgery
Albert-Ludwigs-University
Freiburg, Germany

CONTRIBUTORS
Hendrick B. Barner D. Garcia-Dorado
Division of Cardiothoracic Surgery Servicio de Cardiologia
Washington University School Hospital General Vall d’Hebron
of Medicine Barcelona, Spain
St. Louis, Missouri, U.S.A. Chapter 2
Chapter 13
John W.C. Entwistle
Gerald D. Buckberg Department of Cardiothoracic Surgery
UCLA Medical School Allegheny University
Division of Cardio-Thoracic Surgery of the Health Sciences
Los Angeles, California, U.S.A. Philadelphia, Pennsylvania, U.S.A.
Foreword Chapter 4

Michael Buerke Andrew C. Fiore


Department of Medicine II Division of Cardiothoracic Surgery
Johannes Gutenberg-University St. Louis University
Mainz, Germany Health Sciences Center
Chapter 3 St. Louis, Missouri, U.S.A.
Chapter 13
Harald Darius
Department of Medicine II Willem Flameng
Johannes Gutenberg-University Cardiac Surgery
Mainz, Germany Katholieke Universiteit Leuven
Chapter 3 Leuven, Belgium
Chapter 6
Torsten Doenst
Albert-Ludwigs Universitat Armin Geiger
Division of Cardiovascular Surgery Division of Cardiovascular Surgery
Freiburg, Germany Albert-Ludwigs-University
Chapters 7,10 Freiburg, Germany
Chapter 19
Francesco Donatelli
Cattedra e Divisione di Cardiochirurgia Adalberto Grossi
IRCCS, Ospedale Maggiore Policlinico Cattedra e Divisione di Cardiochirurgia
Università degli Studi di Milano IRCCS, Ospedale Maggiore Policlinico
Milano, Italy Università degli Studi di Milano
Chapter 17 Milano, Italy
Chapter 17
Siegfried Hagl Katharina Palisek
Department of Cardiac Surgery Clinic of Cardiology and Clinic
University of Heidelberg of Surgery
Heidelberg, Germany University of Vienna
Chapter 5 Vienna, Austria
Chapter 9
Waltraud Ibe
Department of Medicine II Louis P. Perrault
Johannes Gutenberg-University Montreal Heart Institute
Mainz, Germany Montreal, Quebec, Canada
Chapter 3 Chapter 11

Harold L. Lazar H.M. Piper


Boston University School of Medicine Physiologisches Institut
Boston Medical Center Kinikum der Justus-Liebig-Universitat
Boston, Massachusetts, U.S.A. Giessen, Germany
Chapter 15 Chapter 2

Juergen Martin Oleg I. Pisarenko


Division of Cardiovascular Surgery Institute of Experimental Cardiology
Albert-Ludwigs-University Cardiology Research Centre
Freiburg, Germany Moscow, Russia
Chapter 19 Chapter 8

Philippe Menasche Marco Pocar


Department of Cardiovascular Surgery Cattedra e Divisione di Cardiochirurgia
and INSERM Unite 127 IRCCS, Ospedale Maggiore Policlinico
Hopital Lariboisière Università degli Studi di Milano
Paris, France Milano, Italy
Chapter 11 Chapter 17

Werner Mohl Syed T. Raza


Clinic of Surgery Department of Surgery
University of Vienna Buffalo General Hospital
Vienna, Austria Buffalo, New York, U.S.A.
Chapter 9 Chapter 14

Kiyozo Morita Russell S. Ronson


Department of Cardiothoracic Surgery The Cardiothoracic Research Laboratory
Jikei University School of Medicine The Department of Surgery, Section
Tokyo, Japan of Cardiothoracic Surgery
Chapter 16 The Carlyle Fraser Heart Center
Crawford W. Long Hospital
Fumiyuki Okamoto of Emory University School
Department of Cardiovascular Surgery of Medicine
Teine Keijin-kai Hospital Atlanta, Georgia, U.S.A.
Teine-ku, Sapporo, Japan Chapter 1
Chapter 18
Keisuke Sakai Heinrich Taegtmeyer
Department of Cardiovascular Surgery University of Texas at Houston Medical
Teine Keijin-kai Hospital School
Teine-ku, Sapporo, Japan Division of Cardiology
Chapter 18 Houston, Texas, U.S.A.
Chapters 7, 10
Tomas A. Salerno
Division of Cardiothoracic Surgery Jakob Vinten-Johansen
Department of Surgery The Cardiothoracic Research Laboratory
Buffalo, New York, U.S.A. The Department of Surgery, Section
Chapter 14 of Cardiothoracic Surgery
The Carlyle Fraser Heart Center
Wolfgang Scholz Crawford W. Long Hospital of Emory
Disease Group Cardiovascular University School of Medicine
Hoechst Marion Roussel Atlanta, Georgia, U.S.A.
Frankfurt/Main, Germany Chapter 1
Chapter 6
Andrew S. Wechsler
Gunter Steurer Department of Cardiothoracic Surgery
Clinic of Cardiology Allegheny University of the Health
and Clinic of Surgery Sciences
University of Vienna Philadelphia, Pennsylvania, U.S.A.
Vienna, Austria Chapter 4
Chapter 9
Michio Yoshitake
Rolf Svedjeholm Department of Cardiothoracic Surgery
Department of Cardiothoracic Surgery Jikei University School of Medicine
Linkoping Heart Center Tokyo, Japan
University Hospital Chapter 16
Linkoping, Sweden
Chapter 12

Gábor Szabó
Department of Cardiac Surgery
University of Heidelberg
Heidelberg, Germany
Chapter 5
FOREWORD

I
t has become increasingly clear that ischemia during cardiac surgery causes a
variety of events that can cause, if unaltered by the control of reperfusion,
myocyte endothelial necrosis. This volume describes the injury and operative
procedures that can be employed to improve surgical results.
The metabolic changes associated with ischemia and reperfusion
can be mitigated before, during, and after aortic clamping. This book propounds
two fundamental principles; 1) protection against ischemic injury is related to
how the heart is managed metabolically during ischemia, rather than merely aortic
clamp time and 2) the reperfusion process, that can in part be controlled by alter-
ation of temperature, metabolic substrates, pressure, and substrate supplemen-
tation, can be adjusted to avoid the deleterious and potentially lethal consequences
of ischemia only when the normal blood supply is restored.
Three examples are selected to illustrate these principles. First, cardiac
recovery after 45 minutes of global normothermic ischemia is marginal, with only
30% return of function and high morbidity and mortality after normal blood
reperfusion without myocardial protection during ischemia. In contrast, 4 hours
of aortic clamping with blood cardioplegia can completely protect:
1. the normal heart from damage, and
2. more importantly, hearts that have previously undergone 45 minutes of
normothermic ischemia followed by 2 more hours of ischemia with warm
and cold blood cardioplegia, with associated interventions.
This confirms that the way the heart is managed during aortic clamping is a more
important variable than the rapidity of operation if no protection is provided, and
reliance is placed only upon the speed of the surgeon. Clearly, surgical skill can reduce
ischemic/reperfusion damage, but protective interventions can avoid it, despite a
longer period of aortic clamping.
Second, 2 hours of regional ischemia causes extensive myocardial infarction,
with only minimal salvage if regional tissue or normal blood supply is restored in
a beating working heart (i.e., PTCA after acute myocardial infarction). Under
these regional circumstances, there exists a condition where no control of the
ischemic process is possible so that only the reperfusate modification will define
subsequent improvement. Studies, both experimental and clinical, show that this
ischemic/reperfusion lesion can be limited remarkably by controlling only the
reperfusate conditions and composition.
Third, inadequate distribution of cardioprotective agents must be delivered
homogeneously to limit ischemic damage and/or reverse reperfusion damage. This
allows assessment of the adequacy of the benefits of surgical correction of lesions
requiring ischemia to facilitate technical excellence during operative repair.
These observations have major surgical implications, since:
1. some problems cannot be offset under global circumstances, if
temperature alone is used to reduce metabolic work;
2. responsible processes can be modified surgically to offset the lethal
metabolic and functional changes that cause reperfusion damage when
only normal blood supply is restored;
3. avoidance of reperfusion injury in the regional ischemic heart without
any form of protection during acute myocardial infarction demonstrates
the benefits of controlled reperfusion since this is the only method avail-
able to deal with the consequences of ischemia and reoxygenation. The
absence of myocardial protection indicates clearly the occurrence of
reperfusion injury and its modification by knowledge of its consequences.
In this book, alterations of myocytes and endothelium caused by ischemia
and reperfusion are described in order to provide a comprehensive basis for con-
sideration of the elements of effective cardioprotection. These elements include
(a) characterization of myocardial injury; (b) reduction of endothelial injury by
modifying the potential production of nitric oxide (NO) and augmenting NO
production with metabolic precursors such as L-arginine. Restoration of NO al-
lows more normal vasodilatation and decreases the adherence of neutrophils to
the endothelium and platelet accumulation with ensuing thrombosis. Futher
elements in the development of effective cardioprotection include: (c) manipulation
of the intracellular buffering system by a selective sodium/hydrogen ion exchange
inhibitor to limit postischemic sodium influx and subsequent calcium influx and
accumulation; (d) providing substrates for metabolic support of the mitochondria
by replacing amino acids depleted by ischemia and reperfusion; (e) defining cellu-
lar changes during ischemia and reoxygenation so that a reperfusate can be devel-
oped that decreases calcium entry and limits hypercontracture; (f ) limiting
reperfusion injury by reducing oxygen levels and adding antioxidants to limit
reoxygenation injury; (g) further understanding preconditioning at the cellular
level to limit post-ischemic stunning and thereby setting the stage for pharma-
cologic treatment to limit damage without enhancing ischemia; (h) supplement-
ing the cardioplegic solution with magnesium to further limit calcium influx;
and (i) changing the method of arrest by using repolarizing agents like pencidil to
alter calcium influx and adding a beta adrenergic blocker (esmolol) simultaneously
limit calcium influx and cardiac work after aortic unclamping. In any event, the
ideal mode of cardioplegic delivery, whether antegrade, retrograde, simultaneous
antegrade/retrograde or even pulsatile perfusion. must open vessels closed by is-
chemia to establish homogeneous postischemic distribution in steady state flow.
With prolonged ischemia without protection, as in acute myocardial infarc-
tion, myocardial damage occurs that cannot be altered by integrated modification
of reperfusion. For surgical ischemia with protection, the metabolic environment
can be changed by cardioplegic substrates delivered antegrade and/or retrograde.
Yet normal reflow establishes a sequence of metabolic and functional events that
may render the heart susceptible to extensive damage. This damage can be limited
by modifying the conditions of ischemia so that the initial reperfusate restores a
more normal cellular metabolism to allow subsequent ischemia to be better toler-
ated. Our goal in this book is to provide information on ischemia and reperfusion
so as to promote efforts in cardioprotection.
Gerald D. Buckberg, M.D.
UCLA Medical School
Division of Cardiothoracic Surgery
Los Angeles, California, U.S.A.
PREFACE

C
ardiac surgery was always associated with ischemia and consequently
reperfusion from its very beginning. After the recognition that the various
techniques used to perform open heart surgery resulted in some form of
ischemia, many different techniques evolved to protect the myocardium during
that period of interruption of blood supply. In recent years, convincing data evolved
showing that the reintroduction of normal blood to previously ischemic myocar-
dium results in further injury or reperfusion damage. Great efforts have been un-
dertaken to avoid the problems during more and more complex cardiac opera-
tions. These efforts included:
• Improved myocardial protection during ischemia
• Avoidance or at least reduction of the subsequent reperfusion injury
• Avoidance of ischemia at all (e.g., continuous delivery of oxygenated
cardioplegic solutions, beating heart operations with or without extracorporeal
circulation)
• Increased knowledge of the basic mechanisms underlying ischemia and
reperfusion processes
This book will present the current knowledge of world-renowned experts in
their field. They report their clinical and experimental data and give an overview
of their entire subspecialty.
A successful cardiac operation requires both a perfect anatomical repair and
conditions for myocardial protection that allow these repair techniques to occur.
Even though great improvements have occurred in cardiac surgery over the years
allowing more and more complex operations with better results for the patients to
be performed, we as cardiac surgeons have to further improve both our anatomi-
cal repair techniques as well as the methods with which these techniques can be
applied.
I thank all the contributors for the tremendous work they have done with
their chapters, and I do hope that this will be of benefit for our patients and our
surgical community.

Friedhelm Beyersdorf, M.D.


Freiburg, Germany
Section I:
Basic Science
in Ischemia-Reperfusion Injury
CHAPTER 1

Harnessing the Cardioprotective Potential


of Nitric Oxide in Nonsurgical and Surgical
Ischemic-Reperfusion Injury
Jakob Vinten-Johansen and Russell S. Ronson

I
n cardiac surgery, there are numerous cardioplegia solution through a newly
opportunities during the conduct of the revascularized segment, or initialization of
operation for both unplanned and hence blood flow through an internal mammary
unprotected ischemia with subsequent artery conduit, or 3) removal of the aortic cross
reperfusion. These periods of potential injury clamp. The targets of ischemic-reperfusion
include 1) antecedent ischemia (regional coro- injury are not restricted to myocytes alone in
nary occlusion, profound hypotension) occur- the form of necrosis or contractile dysfunc-
ring before institution of cardiopulmonary tion, but also include the vascular endothe-
bypass or other means of hemodynamic sup- lium and its ability to elaborate important
port, 2) “protected” ischemia encountered endogenous factors such as nitric oxide (NO•)
between infusions of cardioplegia solution, but and adenosine which are active in regulation
which may be complicated by maldistribution of blood flow, blood pressure and cell-cell (neu-
of cardioplegia solution by coronary obstruc- trophil-endothelial cell) interactions.5 Addi-
tions or inadequate delivery pressures, and tionally, extracorporeal circuits used in many
3) inadvertent ischemia occurring after cardiac surgical procedures contribute to the
reperfusion has been initiated.1,2 Each ischemic pathophysiology of ischemia and reperfusion
event not only carries the potential for pro- injuries by activating complement and cyto-
ducing damage in its own right, but may also kines which recruit neutrophils and other
interact with each other in a cumulative fash- inflammatory cells and thereby amplify the
ion. Each interval of ischemia also carries the inflammatory process.6 This extracorporeal
potential for subsequent reperfusion injury, inflammatory component makes surgical
defined as injury extending beyond that ischemic-reperfusion injury uniquely different
present during ischemia. Whether this is an from its nonsurgical counterpart.
active process of dynamic injury development Nitric oxide is a naturally occurring auta-
leading to new necrosis or dysfunction, or coid that has a plethora of physiological actions
whether reperfusion injury is simply a passive as diverse as producing vasodilation to acting
phenotypic expression of morphologic changes as a neuronal signaling molecule responsible
that have occurred during ischemia, and that for many of the biological actions of excita-
interventions to alter reperfusion injury are tory amines. However, an extremely impor-
altering the course of that expression are issues tant physiological effect is the inhibition of
that are fervently debated.3,4 Therefore addi- inflammatory processes, including endothelial
tional reperfusion injury may follow 1) resus- cell activation and expression of adhesion
citation or hemodynamic restabilization before molecules, and inhibition of neutrophil actions
cardiopulmonary bypass, 2) infusion of (Fig. 1.1). NO• is a radical species that has an

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Harnessing the Cardioprotective Potential of Nitric Oxide 3

Fig. 1.1. The plethora of physiological effects of nitric oxide (NO•) are summarized. NO• inhibits (-) neutrophil
activation and adherence to endothelial cells (VEC), as well as inhibits platelet (plt) and mast cell (Mast) aggre-
gation. NO• also directly inhibits the expression or upregulation of cellular adhesion molecules (CAMs) on VEC.
NO• has been implicated in promoting (+) the second window of preconditioning and is a substrate for the
extremely rapid biradical reaction with •O–2 to form peroxynitrite (ONOO-) which may have deleterious effects
in crystalloid solutions because of a lack of detoxifying reactions that prevent ONOO- accumulation, but which
may also have beneficial effects in blood media (i.e., blood cardioplegia) secondary to the presence of those
detoxifying pathways. NO• is most widely known for its effects on vasodilation thereby mediating in part auto-
regulation of blood flow and reactive hyperemia. Other abbreviations: L-Arg = L-arginine; eNOS = endothelial
nitric oxide synthase; VSMC = vascular smooth muscle cells.

extremely short half-life, and hence can dif- contrast, NO• has also been implicated in pro-
fuse short distances before being inactivated, mulgating injury because of its actions as a
but this molecule nonetheless exerts potent radical species or the generation of potentially
physiological actions. NO• formed by the vas- deleterious metabolites such as peroxynitrite
cular endothelium is released into the intra- (ONOO–) and subsequently hydroxyl radical
vascular compartment and the perivascular and (•OH). Therefore, a duality of opposing physi-
interstitial compartments. This close proxim- ological actions is associated with endogenous
ity to compartments which are important in and exogenous NO•, the mechanisms of which
the pathogenesis of ischemia-reperfusion in- are not fully known. This chapter summarizes
jury places NO• in a unique and powerful po- the role of NO• derived either endogenously
sition in modulating cell-cell interactions char- from the vascular endothelium or provided
acteristic of the inflammatory component of exogenously from its physiological precursor
postischemic processes. In recent years, experi- L-arginine or molecules that donate NO• spon-
mental research has shown that NO• exerts taneously in modulating nonsurgical and sur-
potent cardioprotection from ischemic- gical ischemic-reperfusion injury.
reperfusion injury in both surgical and non-
surgical settings by a number of mechanisms, Chemistry of NO• Generation
including inhibiting neutrophils and mast Nitric oxide is a free radical gas formed by
cells,7 and attenuating adhesion molecule the five electron oxidation of L-arginine to
expression on vascular endothelium.8-10 In the products NO• and L-citrulline by the
4 Ischemia-Reperfusion Injury in Cardiac Surgery

monoxygenase enzyme, nitric oxide synthase site, which then facilitates the conversion of
(NOS) (Fig. 1.2). The reaction requires molecular O2 and L-arginine to NO• and
molecular oxygen, NADPH as the electron L-citrulline. Tetrahydrobiopterin acts to facili-
donor, and tetrahydrobiopterin. There are tate the flow of electrons from NADPH to
three isoforms of NOS which are products of the heme moiety of eNOS. Therefore, the
three separate genes and which share 50-60% binding and dissociation of Ca2+-calmodulin
homology between them: 1) neuronal NOS acts as a molecular toggle switch turning the
(nNOS or NOS1) is constitutively present in enzyme on and off. Agonists stimulate an
neuronal tissue; 2) inducible NOS (iNOS or increase in intracellular Ca2+ through recep-
NOS2) is stimulated to generate NO• by se- tor-dependent mechanisms which transduce
lected cytokines and endotoxin over relatively the signal by the phosphoinositide second mes-
long periods of time (hours); 3) endothelial senger system (Fig. 1.2). This second messen-
NOS (eNOS or NOS3) which, like nNOS, is ger generates inositol 1,4,5-trisphosphate
constitutively expressed and is rapidly respon- (IP3), which, in turn, binds to receptors on
sive to selected physiological and pharmaco- the endoplasmic reticulum and subsequently,
logical stimuli (agonists acetylcholine, brady- stimulates the release of Ca2+ from intracellu-
kinin, Ca2+ ionophores). eNOS was initially lar stores. This receptor-dependent Ca2+ sig-
described as originating from endothelial cells nal then stimulates eNOS to increase NO•
but is now known to be expressed by both production and release. Therefore, acetylcho-
vascular and endocardial endothelium as well line and bradykinin stimulate eNOS in a con-
as by myocytes. eNOS will be the focus of this centration-dependent manner. Ca2+ iono-
chapter as the primary source of NO• relevant phores such as A23187 will also stimulate
to myocardial ischemic-reperfusion injury. eNOS to release NO• by receptor-independent
eNOS is a heme-containing enzyme that mechanisms (Fig. 1.2) which helps to differ-
is Ca 2+-calmodulin sensitive. In contrast, entiate receptor-related from nonreceptor-re-
iNOS is Ca2+ insensitive due to the very tight lated impaired NO • release. Vasodilatory
binding of calmodulin to the binding domain responses to receptor-dependent and receptor-
which is not regulated by physiological con- independent stimulators of eNOS forms the
centrations of Ca2+. iNOS is therefore not basis of bioassay systems interrogating the
regulated by physiological concentrations of functional capacity of vascular tissue to gen-
Ca2+, but rather by other slow-response mecha- erate NO•. In addition to intracellular Ca2+,
nisms. eNOS is tethered to the sarcolemmal eNOS is also regulated by vascular wall shear
membrane in microdomains known as stress created by the pressure-flow interactions
caveolea; these specialized areas of the cell plas- in the vessel11,12 and by deformation of the
malemma are involved generally in transcytosis vascular endothelium, both of which accom-
of large molecules and the uptake of smaller pany pulsatile perfusion of the vasculature. The
molecules. Interactions of agonists that stimu- potent vasodilator effects of NO•, coupled with
late eNOS, such as acetylcholine and brady- its physiological regulation by intracellular
kinin, dissociate eNOS from the membrane calcium put this autacoid in an effective posi-
with subsequent translocation into the cyto- tion to contribute to the regulation of blood
sol where it is phosphorylated, thereby acti- pressure and organ blood flow.12 Accordingly,
vating the enzyme. endothelium-derived NO• has been implicated
eNOS enzyme activity is regulated by in the regulation of normal cardiovascular
physiological concentrations of Ca2+ through homeostatic processes such as autoregula-
the binding of calmodulin to the binding tion13,14 and reactive hyperemia,15,16 and in the
domain of eNOS, thereby initiating electron pathophysiology of disease states including
flow involved in the production of the NO• shock17,18 atherosclerosis19,20 hypertension,21,22
radical. The enzyme has a calmodulin-bind- and hypercholesterolemia.20,23 Attenuated
ing domain that undergoes a conformational NO• release after cardiopulmonary bypass has
change when Ca2+ and calmodulin bind to this also been implicated in the etiology of pul-
Harnessing the Cardioprotective Potential of Nitric Oxide 5

Fig. 1.2. Nitric oxide (NO•) is produced by the oxidation of the guanidino moiety of L-arginine by endothelial
nitric oxide synthase (eNOS or NOS III). Basal release of nitric oxide is stimulated by vascular shear stress and
physiological agonists such as acetylcholine (ACl) and bradykinin (BK) through receptor interactions. Nitric oxide
rapidly diffuses ablumenally when it causes vascular smooth muscle relaxation and vasodilation. Diffusion
intralumenally inhibits platelet aggregation, attenuates neutrophil activation, and interacts with superoxide anion
• –
O 2 to form peroxynitrite (ONOO–). Under experimental conditions such as in organ chambers, pharmacologi-
cal agonists can stimulate eNOS by receptor-dependent (ACh, BK) or receptor-independent (Ca2+ ionophore
A23187) to produce degrees of vasorelaxation which are dependent on concentration of agonists, efficiency of the
receptor mechanism, or the function of eNOS.

monary vasoconstriction and pulmonary hy- released NO• is the iron center at the active
pertension after cardiopulmonary bypass.24 site of guanylate cyclase. NO• stimulates
guanylate cyclase by binding to iron in the
Physiological Actions of NO• heme center, which stimulates the enzyme to
Relevant to Ischemia-Reperfusion generate cGMP and induce vasorelaxation
through a number of mechanisms. The
vasorelaxation response to acetylcholine of iso-
Vasodilation lated coronary vessels with intact endothelium
The role of NO• (endothelium-derived re- in organ chambers is consistent with vasodila-
laxing factor, later identified as NO• or a tor and hypotensive responses observed in vivo.
nitrosylated intermediate) as a physiological The degree of vasorelaxation is related to the
regulator of vascular tone was first identified amount of NO• released and the sensitivity of
by the classic studies of Furchgott and guanylate cyclase. The implication of this
Zawadski25 in which the endothelium was vasodilator effect on macrovessels (conduit or
shown to play an obligatory role. NO• has sub- conductance vessels) and microvessels (resis-
sequently been identified as the endogenous tance vessels) in normal as well as in post-
nitrovasodilator, and the dilating component ischemic myocardium is that the loss of
in vasoactive agents such as nitroglycerin, regulation of local vascular resistance by NO•,
nitroprusside and other organic nitrates. NO• secondary to endothelial dysfunction and
released either through basal or agonist-medi- impaired release of NO•, may lead to defects
ated mechanisms diffuses both lumenally and in reactive hyperemic responses and in global
ablumenally. A key target of ablumenally
6 Ischemia-Reperfusion Injury in Cardiac Surgery

and regional postischemic blood flow.12,16,26 physiological concentrations of NO•.37,38 The


Impaired reactive hyperemia may also involve disparate observations may be related to the
the neutralization of NO• by superoxide radi- amount of NO• released, which may be higher
cals generated during even brief ischemia.27 (micromolar range) with cytokine stimulation
(i.e., of iNOS) compared to either endog-
Inhibition of Metabolism enously released NO• (0.1-1 nMol in coro-
and Contractile Function nary circulation)39 or therapeutic concentra-
NO• may decrease the rate of glycolysis by tions of NO• donor agents (approximately 500
mechanisms independent of guanylate cy- nMol). Higher concentrations of NO• released
clase.28 NO• stimulates the ribosylation of by endotoxin stimulation may mediate cardiac
ADP; stimulation of auto-ADP ribosylation depression during the later stages of septic
of glyceraldehyce-3-phosphate dehydrogenase shock. However, in surgically or nonsurgically
(GAPDH) inhibits the enzyme and in turn reperfused postischemic hearts, any direct
inhibits glycolysis. Ostensibly, the inhibition negative inotropic actions of NO• may be over-
of metabolism and ATP generation may ac- ridden by the net cardioprotective effects of
cordingly reduce contractile function during NO• secondary to a reduction of neutrophil-
early reperfusion. The negative metabolic ef- and oxidant-mediated injury, resulting in bet-
fect of NO• may therefore have implications ter postischemic function.40-44
in the genesis of myocardial stunning29 and
hibernation in which glycolysis plays a large Decreased NO• Release After
role. NO• may also have a negative modula- Endothelial Damage
tory effect on oxidative metabolism by inhib- from Ischemic-Reperfusion
iting mitochondrial oxidative phosphorylation
and hence oxygen consumption.30 However,
Injury
this effect on oxygen consumption is not uni- Under normal conditions, the endothelium
versally reported.31 tonically releases NO•45-47 in the neighborhood
of 0.1-1 nMol into the vascular and intersti-
The inhibition of glycolysis may have some tial compartments. This basal NO• release is
implication in the purported negative inotro- regulated by vascular shear stress,48-50 the tonic
pic effects of NO• in normal myocardium.32 release of physiological agonists and humoral
However, there is controversy over the effects agents, and intracellular calcium. During car-
of NO• on the inotropic state and contractile diopulmonary bypass, a basal release of NO•
function of the heart. Finkel et al32 first re- has been observed by Hattler et al.29 Basal
ported a negative inotropic effect of cytokines release of NO• is inhibited by a number of
(TNF-!) in papillary muscle preparations pharmacological agents and pathological
which they attributed to the effects of NO•. conditions as summarized in Figure 1.3.
This observation would have relevance to Guanidino-substituted L-arginine analogue
procedures using cardiopulmonary bypass inhibitors of eNOS such as NG-nitro-L-argi-
since cytokines, 33,34 as well as comple- nine (L-NAME) and N G -monomethyl-
ment,6,35,36 are elevated during bypass by ex- L-arginine (L-NMMA), as well as calmodulin
posure of blood to foreign surfaces and the antagonists,45 reduce directly measured NO•.
triggering of cytokine and complement frag- On the other hand, L-arginine but not D-argi-
ment release by ischemia-reperfusion. In sup- nine stimulates the basal release of NO•.45,51
port of stimulated release of NO• during by- Because of the near diffusion-limited biradical
pass, transmyocardial NO• levels as well as reaction rate of •O –2 with NO • to form
NOS activity have been observed to be el- ONOO–,52-55 the rapid degradation of •O–2
evated after release of the crossclamp during to H2O2 by superoxide dismutase also increases
cardiac surgery,29 a time when cardiac depres- the amperometrically measured NO• in vitro.45
sion is often observed. However, other studies The neutralization of basally released NO• and
have reported no negative inotropic effects of its vasodilator effects by •O–2 in conditions
Harnessing the Cardioprotective Potential of Nitric Oxide 7

Fig. 1.3. Inhibitors of basal nitric oxide production and release by the vascular endothelium. L-arginine (L-Arg)
is transported into the vascular endothelium and converted to NO• and L-citrulline by eNOS. This process requires
the presence of molecular oxygen. The generation of NO• can be inhibited by lack of oxygen (ischemia, anoxia).
Nonmetabolized analogues of L-arginine then inhibit enzyme activity (L-NAME, L-NMMA), and pathophysi-
ological conditions such as angiotensin II (Ang-II)-dependent hypertension (HyperT), ischemia-reperfusion (I-R)
and hypercholesterolemia.

such as angiotensin-related hypertension may cardioprotection in those same patients is not


be one of the pathophysiological mechanisms clear.
of high blood pressure. In addition, NO• itself
may have a negative feedback regulation on Time Course of Endothelial
eNOS activity. This was first reported by Injury
Rogers and Ignarro56 for constitutive NOS in
Ischemia and reperfusion cause injury to
rat cerebellum, and later by Buga et al57 for
the vascular endothelium, manifest as a reduc-
eNOS activity in cultured endothelial cells.
tion in basal and stimulated NO• release58-61
Therefore, exogenous NO• may regulate its
and hence attenuated responses to agonist
own synthase activity. As demonstrated by Ma
stimulators of eNOS. 59,62,63 In coronary
et al,31 exogenous NO• also regulates in vivo
occlusion models (regional ischemia), endot-
basal NO• release by a negative feedback
helial dysfunction is minimally expressed after
mechanism on its own endothelial synthase
the ischemic period (60-90 minutes), but is
activity. This inhibitory mechanism may in-
progressively expressed starting as early as 2.5
volve binding of NO• to the heme moiety of
minutes after the start of reperfusion, and persists
eNOS, thereby inhibiting its catalytic activ-
hours58,64 to days65 after reperfusion. Figure 1.4
ity. Clinically, this negative feedback inhibi-
shows coronary artery endothelial function
tion may help explain impaired NO• -depen-
(assayed as responsiveness to agonist stimula-
dent autoregulation in patients on long-term
tors of eNOS) 6, 24 and 48 hours after LAD
nitrovasodilator therapy. Whether this auto-
occlusion. Notice that responses to acetylcho-
inhibition has implications for less effective
line were less in the LAD compared to the
nonischemic left circumflex coronary artery
8 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 1.4. Responses of preconstricted (U46619) postischemic coronary arteries to endothelium-dependent acetyl-
choline (receptor-dependent) and smooth muscle dilator sodium nitroprusside. LCX = normally perfused left
circumflex coronary artery; LAD6H, LAD24H and LAD48H = ischemic (1 hour)-reperfused left anterior de-
scending coronary artery reperfused for 6, 24, or 48 hours. Top Panel. Responses to acetylcholine. Bottom Panel.
Responses to sodium nitroprusside. * p < 0.05 vs nonischemic LCX. Concentrations are final organ chamber
concentrations. Note the persistent depression of endothelial responses over 48 hours of reperfusion. (From Zhao
et al, unpublished results)

at any concentration. This impaired respon- normothermic ischemia with or without


siveness persisted for 48 hours. In models of unmodified blood reperfusion in cardiopul-
global ischemia, endothelial dysfunction has monary bypass. The left anterior descending
been shown to be present after reperfusion and left circumflex coronary arteries were
with little or no dysfunction expressed during excised, placed in organ chambers in a Krebs-
ischemia60,61 unless more prolonged periods Henseleit buffer medium, and agonist-stimu-
of ischemia are imposed.60 This endothelial lated endothelial function interrogated using
dysfunction is expressed as a reduction in basal acetylcholine for receptor-dependent vaso-
NO• release as well as stimulated NO• release, relaxation responses and the Ca2+ ionophore
with obtunded relaxation responses to acetyl- A23187 for receptor independent vaso-
choline or other agonist stimulators of NOS. relaxation responses. As shown in Fig. 1.5A,
In the study by Nakanishi et al,61 canine hearts maximal vascular relaxation responses to
were subjected to 45 minutes of global acetylcholine in hearts subjected to ischemia
Harnessing the Cardioprotective Potential of Nitric Oxide 9

Fig. 1.5. Relaxation responses of preconstricted (U46619) postcardioplegia epicardial coronary arteries to agonists.
PANEL A: Maximal relaxation responses to the endothelium-dependent, receptor-dependent agonist acetylcho-
line (ACl). PANEL B: Maximal relaxation responses to the direct (endothelium-independent) vascular smooth
muscle dilator acidified NaNO2. Cntl = normal control vessel; Isch = after 45 minutes global normothermic
ischemia (no reperfusion); Rep = ischemia followed by one hour unmodified blood reperfusion; Isch + BCP = 45
minutes global normothermic ischemia followed by one hour intermittent (every 20 minutes) 4˚C hyperkalemic
blood cardioplegia (4 parts blood:1 part crystalloid); BCP + Rep = global ischemia plus one hour cardioplegia
followed by one hour blood reperfusion. *p < 0.05 vs unstarred groups. Adapted from Nakanishi et al.61

without reperfusion were comparable to nor- endothelium was largely normal, with normal
mal control hearts. In contrast, maximal vaso- and confluent attachment to the subendothe-
relaxation responses in coronary arteries sub- lial matrix. Ischemic endothelial cells differed
jected to both global ischemia and reperfusion from normal tissue by the presence of vacu-
were reduced by approximately 35-40% rela- oles randomly distributed in the cytoplasm.
tive to control hearts and ischemia only hearts. In sharp contrast, tissue reperfused with un-
A modest rightward shift in the entire con- modified blood demonstrated detached
centration-relaxation response curve to acetyl- endothelium with exposed subendothelial
choline occurred in ischemia—only coronary matrix and endothelial cell fragmentation. In
arteries (EC50 of 4.0 ± 0.5 x 10–8 Mol vs attached endothelial cells, the cytoplasm was
control 2.2 ± 0.3 x 10–8 Mol, p < 0.05), while dramatically hypervessiculated. These mor-
a greater shift was seen in reperfused vessels phologic changes are in concordance with the
(6.0 ± 0.2 x 10–8 Mol) suggesting endothelial severe dysfunction to the enzyme system
dysfunction. Similar impaired maximal and described above. These data demonstrating
concentration-dependent responses to those in impaired vasorelaxation responses to agonist
Figure 1.5A were also observed for the Ca2+ stimulators of eNOS and morphologically-
ionophore A23187. Smooth muscle relaxation apparent damage are consistent with loss of
responses to the NO• donor acidified (pH 2.0) basal NO • release measured ampero-
NaNO2 were 100%, indicating no damage to metrically, reported by Engelman et al.47
the vascular smooth muscle (Fig. 1.5B). These In regional ischemia, vasodilator responses
data suggest that endothelial dysfunction in- to both receptor-dependent (acetylcholine)
volved not only damage to the receptor-trans- and receptor-independent (calcium ionophore
duction complex, but also to the more distal A23187 which stimulates eNOS distal to the
processes regulating eNOS activity or dam- receptor-G-protein complex) stimulators of
age to the enzyme itself.61 In this model of NOS are adversely effected, while in global
global ischemia61 endothelial dysfunction was models involving short (∀ 30 minutes) peri-
associated with morphological abnormalities ods of normothermic ischemia before car-
in endothelial structure. In ischemic-only (no dioplegia primarily the receptor-mediated
reperfusion) hearts, the coronary microvascular endothelial vasodilator responses are
10 Ischemia-Reperfusion Injury in Cardiac Surgery

effected.61,66 These data suggest that a dif- most likely the sialomucin P-selectin glyco-
ferent phenotype of injury may be expressed protein ligand-1 (PSGL-1).71,72 This initial
dependent on the type and duration of interaction is an obligatory step necessary for
ischemia (regional vs global) imposed. Short- distal firm adherence interactions and
term global ischemia (i.e., ∀ 30 minutes) rel- transendothelial migration into the myocar-
evant to cardiac surgery may selectively target dial parenchyma and their physiological se-
the receptor-G-protein complex,66 while more quelae (no-reflow, necrosis).73-76 After initial
severe regional ischemia-reperfusion (i.e., tethering of neutrophils by endothelial
inducing myocardial necrosis and infarction) p-selectin, a well orchestrated sequence of neu-
may target both the receptor-G-protein com- trophil-endothelial cell interactions evolves
plex and the distal stimulus coupling mecha- with the endothelial expression of adhesion
nism, or the enzyme itself. In both cases, the molecules, such as E-selectin and ICAM-1,
acute injury to the endothelium may be and expression of adhesion counterligands on
mediated by oxygen radicals formed by 1) the the neutrophils such as CD11/CD18, that
endothelium (via xanthine oxidase activity or allow firm adherence of the neutrophil to the
NAD(P)H oxidase activity), 2) by mitochon- endothelium. Nitric oxide also limits the
dria or 3) by neutrophils adherent to the vas- capability of the endothelium to express both
cular endothelium. Superoxide dismutase re- E-selectin and ICAM-1 after activation by
duces the injury to the endothelium following cytokines released during ischemia and
ischemia and reperfusion, while direct expo- reperfusion.77,78 The ability of nitric oxide to
sure of coronary artery endothelium to oxy- effect multiple points in the neutrophil-endo-
gen radicals produces endothelium-dependent thelial cell adhesion process not only makes it
injury to receptor-mediated dilation responses a powerful tool for minimizing reperfusion
without effect to the receptor-independent injury caused by neutrophils, but also leads to
dilator (smooth muscle) responses.67 The oxy- the possibility that NO• is acting on a proxi-
gen radicals may not only damage the trigger mal common step in the formation of several
mechanisms of NO• but may also directly neu- types of adhesion molecules. DeCaterina et al
tralize NO• through the rapid biradical reac- have shown significant decreases in activation
tion with superoxide anion. of NF-#B, a gene promoter for synthesis of
adhesion molecules, following endothelial
Endothelial Dysfunction activation with TNF-!. This same decrease in
NF- # B was not present in endothelium
and Neutrophils
exposed to c-GMP, glutathione, or nitrite. 77
Endothelial dysfunction plays a critical role
This important action of NO• has the poten-
in the pathogenesis of reperfusion injury in
tial to decrease the accumulation of neutro-
the myocardium.64,68-70 This key role is thrust
phils in the myocardium following ischemia
upon the endothelium because of its interac-
and reperfusion acutely as well as over longer
tion with polymorphonuclear leukocytes or
term time frames (requiring protein synthesis).
neutrophils and other inflammatory cell types
After initial p-selectin-mediated engage-
at the vascular interface in the early and later
ment with the endothelium, the neutrophils
phases of reperfusion. This interaction is
accumulate in the ischemic-reperfused myo-
mediated by a highly specific and temporally
cardium primarily after the onset of reper-
orchestrated sequence of interactions between
fusion.68,79,80 Dreyer et al79 have shown that
adhesion molecules on both the endothelium
the rate of neutrophil accumulation within the
and neutrophils. Thus, the initial interaction
reperfused myocardial area at risk is rapid
between endothelium and neutrophils is char-
within the first hour; Lefer et al64 have reported
acterized by rolling of the neutrophils along
a similar time course using tissue myelo-
the endothelial surface mediated by interac-
peroxidase activity as a marker of neutrophil
tion between p-selectin on the endothelium
accumulation. Furthermore, Lefer et al68 have
and sialylated glycoprotein on the neutrophil,
shown that this accumulation of neutrophils
Harnessing the Cardioprotective Potential of Nitric Oxide 11

in ischemic tissue is preceded by a more rapid 600 ∝M, a direct scavenger of NO•, reversed
rate of adherence of neutrophils to the coro- the effects of L-arginine.51,84 Moreover, in a
nary vascular endothelium. This adherence is nonsurgical model of regional ischemia,
associated closely with the time course of intravenous51 or intracoronary155 L-arginine
endothelial dysfunction related to the stimu- supplementation at the time of reperfusion sig-
lated release of NO• and eventual accumu- nificantly decreased postischemic coronary
lation of neutrophils in postischemic tissue artery endothelial dysfunction and reduced
in both surgical 40,41,81-83 and nonsurgical infarct size; both were associated with
ischemia-reperfusion models. The reduction decreased neutrophil adherence to endothe-
in basal release of NO• by postischemic coro- lium and accumulation in the area at risk. In
nary vascular endothelium resulting from the the study by Nakanishi et al, infarct size was
initial deleterious actions of neutrophils am- reduced from 35 ± 2% of the area placed at
plifies the obligatory interaction between neu- risk to 18 ± 3% with 10mM intracoronary
trophils and endothelial cells, thereby trigger- L-arginine. Infarct size reduction was not
ing the adherence-dependent cascade of observed with D-arginine (10 mM, infarct size
neutrophils and subsequent neutrophil-medi- 49 ± 5% of area at risk).63 Neither postischemic
ated inflammatory component of reperfusion regional contractile function or myocardial
injury. blood flow were improved with L-arginine
treatment.
NO•-Related Therapeutic Basally expressed NO• seems to provide
Strategies endogenous cardioprotection which, when
NOS is selectively attenuated with inhibitors
such as L-NA or L-NAME, markers of injury
Enhancing Endogenous NO• Release are accentuated compared to noninhibited
NO•-related therapeutic strategies for at- groups.86,87 Consequently, in vivo infarct size
tenuating reperfusion injury are summarized was increased from 27 ± 2% area at risk in
in Table 1.1. Enhancing the release of untreated rabbits, to 51 ± 2% in rabbits given
endogenous NO• can be achieved by provid- intravenous L-NA at the time of reperfusion.88
ing the precursor L-arginine. Although L-argi- Similar observations have been made in surgi-
nine appears in the blood in sufficient con- cally relevant models using cardiopulmonary
centrations to saturate NOS, supplemental bypass and cardioplegia. This reduction in the
L-arginine has been shown to increase NO• contribution of basally released NO • to
release (directly or indirectly measured) by the endogenous cardioprotection, secondary to
coronary vascular endothelium.45,84,85 In iso- endothelial injury, may be a significant con-
lated rat aortic endothelial cells, 1 mM L-argi- tributor to postischemic (postcardioplegia)
nine increased the amperometrically measured infarction, systolic and diastolic dysfunction
release of NO• by approximately 40% above and vascular injury often observed in untreated
basal release.45 D-arginine at the same con- groups.2,40,44,61,89 In surgical models using
centration did not increase basally released antecedent ischemia (regional or global) with
NO•. subsequent cardioplegia and reperfusion,
Supplementation with L-arginine up to supplementing with L-arginine to compensate
10 mM concentrations in vitro resulted in for lost basal release of NO•, has shown sig-
decreased neutrophil adherence to activated nificant benefit. Sato et al83 used a canine
coronary artery endothelium, back to basal model of regional (left anterior descending
levels in unstimulated vessels, and significantly (LAD) coronary artery ligation for 90 min-
attenuated neutrophil-mediated injury utes, followed by cardioplegic arrest using
although L-arginine did not directly attenu- blood cardioplegia without (unsupplemented)
ate neutrophil superoxide anion production, or with 10 mM L-arginine with concomitant
Figure 1.6.84 D-arginine (10 mMol) did not intravenous infusion (4 mg/kg/min) starting
inhibit adherence, and carboxy-PT10, at release of the crossclamp. The LAD ligature
12 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 1.1. Strategies in nitric oxide therapy

Endogenous Approach Exogenous Approach

NO• precursor Authentic NO•


L-arginine Gas in solution
Acidified NaNO2

Agonist stimulators NO• donors


Acetylcholine Bioconversions (NTG,
Bradykinin SPM-5185)
Direct donors

Transfection with eNOS

was removed just before the second infusion later in the discussion.91 Along the same lines
of blood cardioplegia, thereby allowing deliv- of pericardioplegia use of L-arginine,
ery of cardioplegic solution to the previously Hiramatsu et al42,92 administered L-arginine
ischemic (revascularized) myocardial region. during the early phase of reperfusion in blood
Compared to the unsupplemented blood car- perfused isolated hearts, ostensibly targeting
dioplegia group, L-arginine supplementation reperfusion events, and reported better recov-
resulted in 1) a 33% increase in postischemic ery of postcardioplegia systolic function com-
systolic shortening of the area at risk and a pared to no adjunct treatment. This timing of
significant decrease in segment stiffness, 2) a administration of L-arginine is consistent with
30% reduction in infarct size (28 ± 4% vs 40 its antineutrophil effect in this and in vivo
± 4% of the area at risk, p < 0.05) and 3) near models. Similar beneficial effects of L-argin-
normalization of postischemic LAD endothe- ine administered at reperfusion were also re-
lial responses to acetylcholine, suggesting im- ported by Armani et al93 in a cell-free perfu-
proved endothelial function (Fig. 1.7). These sate system, suggesting that NO• derived from
beneficial effects were associated with lower supplemental L-arginine may have beneficial
myeloperoxidase activity levels in the area effects based on other than its antineutrophil
at risk used as a marker of neutrophil effects, i.e., quenching of superoxide anions.
accumulation, suggesting a reduction of neu- However, the benefits of L-arginine supple-
trophil-mediated injury with L-arginine mentation as a source of NO• in cell-free sys-
supplemented blood cardioplegia.83 These tems is controversial.
effects with L-arginine were reversed by
inclusion of the NOS inhibitor L-nitro-argi- Protection by Nitric Oxide
nine (L-NA) before administration of L-argi- Donors
nine-enhanced blood cardioplegia in a separate
In the study by Sato et al,83 only a modest
group. These results have been corroborated
decrease in infarct size was achieved compared
by other studies in which cardioplegia solu-
to other cardioprotective strategies, i.e.,
tions were supplemented with L-arginine.42
adenosine. This may represent a limitation of
Interestingly, a preischemic infusion of L-argi-
utilizing the endogenous cardioprotective
nine was found to be beneficial on post-
mechanisms of the heart with L-arginine
ischemic variables by Engelman et al90 in a
therapy in that the increased release of NO• is
nonsurgical model, while both pretreatment
dependent on the functional state of the
and supplementation of cardioplegia have been
endothelium. An injured endothelium in
reported to be beneficial in a surgical model.
which NO• generation mechanisms are im-
Interestingly, 3 mMol L-arginine given after
paired may show obtunded responses to
cardioplegia was detrimental, a subject addressed
Harnessing the Cardioprotective Potential of Nitric Oxide 13

Fig. 1.6. PANEL A. Effects of different


concentrations of L-arginine (L-Arg),
the NO • scavenger carboxy-PTIO
(PTIO) and D-arginine on PMN ad-
herence to coronary endothelium.
Coincubation with PAF is indicated by
the bracket at the bottom. Results are
presented as numbers of PMN per mm2
of endothelium. Bar heights represent
the mean ± s.e.m. *P<0.05 versus other
groups without asterisks. P<0.05 ver-
sus all other groups. PAF concentra-
tion—100 nMol. Numbers in the bars
represent numbers of coronary rings
studied. PANEL B. Effects of
unactivated and PAF-activated PMN,
and L-NA on endothelium-dependent
relaxation of coronary artery rings.
There is no difference between the con-
trol (CTRL) rings not incubated with
PMN and coincubation with
unactivated PMN groups. PAF activa-
tion of PMN caused a rightward shift
in the dose-response curve. L-NA +
L-Arg showed decreased responses to
acetylcholine. *P< 0.05 versus other
groups. PANEL C. Effects of 10 mM
L-arginine (L-Arg) and 10 mM D-Argi-
nine (D-Arg) on PMN-mediated alter-
ations in endothelium-dependent re-
laxation of coronary artery rings.
Responses to acetylcholine were signifi-
cantly greater in the L-Arg-treated rings
compared with D-Arg and PMN + PAF
rings. *P<0.05 versus other groups.
P<0.05 PMN + PAF versus L-Arg and
CTRL groups. D-Arg versus CTRL
group. n = number of rings used.
Adapted from Sato et al, Cardiovasc
Res 31; 1996:63-72.

L-arginine. Therefore, this limitation in the decreased infarct size by about 75%, which
endogenous capabilities of the heart may be was associated with decreased neutrophil
overcome by parenteral administration of accumulation in the area at risk (both non-
agents that donate NO• in vivo. This exog- necrotic and necrotic). These data suggest that
enous approach was first reported by Johnson NO• reduced infarct size by inhibiting neu-
et al using authentic nitric oxide gas (approxi- trophil-mediated damage. Subsequent studies
mately 10-20 nM in vivo concentration)94 and have been performed using a variety of organic
NaNO 2 as an NO • donor chemical 95 at NO• donor agents in nonsurgical models of
subvasodilator concentrations, administered coronary occlusion and reperfusion. These or-
only at the onset of reperfusion in a feline model ganic donors release NO• either spontaneously
of regional (90 minutes LAD occlusion fol- or after bioconversion reactions. Nitroglycerin
lowed by 4.5 hours of reperfusion) ischemia- is the prototype NO• donor agent, but is a
reperfusion. Both forms of NO • therapy poor NO • donor due to its prerequisite
14 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 1.7. Beneficial effects of blood car-


dioplegia supplemented with L-argin-
ine. Top panel. Segmental work of the
ischemic-reperfused myocardium de-
rived by integrating the area of the pres-
sure-segment length loop during con-
trol, after 40 minutes of LAD occlusion
and during reperfusion after cardiople-
gia arrest. Open bar = unsupplemented
blood cardioplegia; hatched bar = L-argi-
nine supplemented blood cardioplegia;
stippled bar = treatment with NOS in-
hibitor L-NA before cardioplegia.
Middle panel. Infarct size as area of ne-
crosis relative to the area at risk. Infarct
size was reduced by approximately 30%
in blood cardioplegia supplemented with
L-arginine, and this infarct size reduc-
tion was reversed by preceding L-argin-
ine with the NOS inhibitor L-nitro argi-
nine (L-NA). Lower panel. Neutrophil
accumulation in the nonischemic zone
(NIZ), ischemic non-necrotic zone (IZ),
and necrotic zone (NEZ) of the area at
risk, assayed by the neutrophil-specific-
enzyme myeloperoxidase in the myo-
cardium. Group legend same for top
panel. Adapted from Sato et al. J Tho-
racic Cardiovasc Surg 1995; 110:
302-314.83

bioconversion by a cysteine-containing risk.96 The nonactive form of the compound


enzyme that is partially depleted in the had no effects on any physiological variable.
microvasculature after ischemia-reperfusion, In a study by Nakanishi et al40 using a ca-
and tolerance develops over time. One such nine model of cardiopulmonary bypass and
cysteine-containing compound readily releases cardioplegia, hearts were subjected to 30 min-
NO• after biotransformation, SPM-5185 (N- utes of normothermic ischemia followed by
(3-hydroxy-pivaloyl)-S-(N’-acetylalanoyl)-L- one hour cardioplegia (4˚C multidose blood
cysteine ethyl ester (Schwarz Pharma AG, cardioplegia, 4:1 blood: crystalloid, Table 1.2).
Monheim, Germany).41,96 This NO•-donor Hearts received either standard blood car-
agent, when administered during reperfusion dioplegia (BCP) or blood cardioplegia with 10
after 1 hour LAD occlusion reduced infarct ∝mol/L SPM-5185 (BCP + SPM). After one
size from 41.7 ± 5.4% to 12.5 ± 3.2% of the hour of cardioplegic arrest, the heart was
area at risk, in association with a 58% reduc- reperfused for a total of 60 minutes, first in
tion in transmural neutrophil accumulation the beating empty state for 30 minutes and
(tissue myeloperoxidase activity) in the area at then after discontinuation of bypass for 30
Harnessing the Cardioprotective Potential of Nitric Oxide 15

minutes. Left ventricular function was assessed through this reaction can be increased by
by the slope of the end-systolic pressure- increasing either NO• or •O–2; the •O–2 specie
volume (impedance catheter) relation. Postis- is generated in larger quantities during the
chemic end-systolic pressure-volume relation early moments of reperfusion, with lower but
was depressed by 53.7% of preischemic val- still significant quantities released on a persis-
ues in the BCP group (from 8.2 ± 1.0 to 3.8 ± tent basis during the later phases of reper-
0.3 mm Hg/ml). In contrast, there was nearly fusion. Under physiological concentrations of
complete postischemic (Fig. 1.8) functional NO• (~ 5-10 nM) and •O–2, endogenous and
recovery in the BCP + SPM group (from 7.6 exogenous superoxide dismutase are effective
± 1.1 to 7.2 ± 1.2 mm Hg/ml). In coronary scavengers. However, in micromolar concen-
arteries isolated from these hearts, endothe- trations as may be reached in vitro, NO•
lium-dependent maximal relaxation to acetyl- effectively competes with superoxide dis-
choline was impaired by 27% in the BCP mutase to form peroxynitrite.
group, but recovered to ∃ 100% in the
BCP+SPM group. Myeloperoxidase activity, Equation 1:
an index of neutrophil accumulation in post- NO• + •O–2 % ONOO–
ischemic myocardium, was elevated in the ONOO– +H+ % ONOOH
BCP group (3.36 ± 0.58 U/100 mg tissue) ONOOH % •OH + NO +
2
but was significantly reduced in the (SOD, MPO)
BCP + SPM group to 1.27 ± 0.45 U/100 mg
tissue. Therefore, this study demonstrated that According to equation 1, the peroxynitrous
addition of 10 ∝mol/L nitric oxide donor acid (ONOOH) intermediate of ONOO–
SPM-5185 in blood cardioplegia improves undergoes a heterolytic cleavage in the pres-
postischemic ventricular performance and ence of transition metals (i.e., superoxide
postcardioplegia endothelial function in dismutase, myeloperoxidase) to form a
ischemically injured hearts, possibly via inhi- hydroxyl anion (•OH) plus a nitronium ion
bition of neutrophil-mediated damage. (NO2+).52 Therefore, in addition to reactive
peroxynitrite, the potential for cellular or tis-
Peroxynitrite: The Potential sue damage exists by the potent oxidant actions
Dark Side of Nitric Oxide of peroxynitrous acid (oxidation of iron and
zinc containing molecular center) and the
Therapy hydroxyl radical (lipid peroxidation,
The biradical quenching reaction between membrane damage).52,53 Peroxynitrite and
NO• and •O– 2 may be a double edged sword. peroxynitrous acid may be major sources of
On the one hand, NO• neutralizes a poten- cytotoxic hydroxyl radicals.53 Accordingly,
tially deleterious species of oxygen radical. On peroxynitrite has been directly or indirectly
the other hand, the reaction produces a po- (through the hydroxyl radial) associated with
tentially deleterious metabolite-peroxynitrite attenuation of mitochondrial respiration,97 cel-
(ONOO–) with subsequent degradation in the lular and tissue injury secondary to hemor-
presence of transition metals to the highly bio- rhagic shock, endotoxemia,98 in normal99 and
logically active hydroxyl radical. This reaction postischemic tissue ischemia-reperfusion100,101
of NO• and •O–2 in a 1:1 stoichiometry to form and atherosclerosis. Peroxynitrite is bacteri-
peroxynitrite progresses at essentially diffu- cidal, nitrosylates sulfhydryl moieties on pro-
sion-limited rates (6.7 x 109 M–1 sec–1), a teins, and causes protein cleavage and
rate which exceeds that between •O–2 and apoptosis. These latter events occur at micro-
superoxide dismutase by a factor of three molar concentrations.
(2 x 109 M-1 sec-1).52 In addition, this reaction On the other hand, numerous studies have
is exothermic with a release of 22 kcal/mol and reported physiological and cardioprotective
is, therefore, essentially irreversible. As seen in effects of ONOO– similar to NO•. It has been
equation 1, the amount of ONOO– produced reported that ONOO– attenuates platelet
16 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 1.2. Composition of blood cardioplegia

Constituent Value

Potassium ion (KCl) (∝mol/L) 20-25 (induction, terminal infusions),


10 (intermittent infusions)
Calcium ion (CPD) (∝mol/L) 0.3

Magnesium ion (derived from blood) (∝mol/L) 0.5

pH (THAM buffer) 8.2 (at 4˚C)


Osmolality (mOsm/L) 360-380

Hematocrit (%) 14-18 (or more)

Oxygen content (Ml O2/100 ml) 8-10

CPD, Citrate-phosphate-dextrose (blood); THAM, tris(hydroxymethyl)-aminomethane

Fig. 1.8. Postcardioplegia recovery of end-systolic pressure-volume slope as a percent of preischemic values. There
was a significant depression of left ventricular performance with unsupplemented blood cardioplegia (BCP) which
was significantly improved with the addition of 10 ∝Mol SPM-5185. *p < 0.05 vs BCP
Harnessing the Cardioprotective Potential of Nitric Oxide 17

aggregation, induces vasorelaxation, inhibits rate between NO • and • O – 2 potentially


adherence of neutrophils to stimulated outcompetes endogenous SOD for •O –2.
coronary artery102 or mesenteric artery103 Therefore, the presence of both NO• and •O-2
endothelium, preserves postischemic is favored under ischemia-reperfusion condi-
vasorelaxation responses to acetylcholine (an tions. The tissue concentration of ONOO–
agonist of endothelial NO• release), and may be in the nanomolar range in normal as
reduces myocardial infarct size.102 The reduc- well as pathological states, and that micromo-
tion in neutrophil adherence by low micro- lar concentrations are not likely to be achieved.
molar concentrations of ONOO– is associated However, studies in which ONOO– has been
with an attenuation of p-selectin surface shown to be deleterious have used micromo-
expression on endothelium103 similar to the lar to low millimolar concentrations of the
physiological effects of NO•. In the study by anion (or its precursors), while studies in which
Nossuli et al102 using a feline model of regional nanomolar concentrations of ONOO– have
ischemia and reperfusion, an approximately been used report a protective effect.
60% reduction in infarct size was associated
with significantly less endothelial dysfunction Physiological Environment
(basal and stimulated release of NO• in bioas- In addition to the rate of formation of
says) relative to a control group. ONOO–, degradation through breakdown
The resolution to this apparent schizo- reactions (Equation 1) and detoxification
phrenic effect of ONOO– where it is cast as a (Fig. 1.9) have a critical impact on the tissue
mediator of damage while in other conditions levels of ONOO– and its biological actions.
it exerts cardioprotective and vasculoprotective However, ONOO– is a relatively stable anion,
effects may be multifactoral, but may depend particularly at alkaline pH, and this stability
on 1) the concentrations of precursors NO• is sufficient to allow transcellular diffusion and
and •O–2 achieved under normal or patho- to engage a biological target such as membrane
physiological conditions and, hence, the physi- lipids, DNA, and tyrosine moieties on pro-
ological concentrations of ONOO– achieved, teins with which ONOO– can react in a
2) the environment in which ONOO– coex- destructive manner.
ists with other cell types and tissues, and the
presence or absence of endogenous detoxifi- The cellular detoxification mechanisms
cation mechanisms Figure 1.9. The balance that may drain ONOO– and prevent tissue
between the formation and breakdown (deg- accumulation of the anion include 1) plasma
radation and detoxification) of ONOO – bicarbonate buffering system and 2) thiol con-
determines, in part, the physiological conse- taining molecules, including albumin and glu-
quences of ONOO–, particularly in pathologi- tathione. NO• has been shown to S-nitrosylate
cal conditions. plasma albumin104 which, in addition to
increasing its half-life, also serves as an NO•
Concentration of ONOO– drain mechanism. Glutathione is present in
The generation of ONOO– is favored cardiac tissue and is considered an endogenous
when both NO• and •O–2 are formed at high antioxidant. ONOO– can nitrosylate reduced
concentrations; this condition may exist dur- glutathione and other thiol compounds to
ing the early moments of reperfusion with the form S-nitrosyl compounds which in turn can
reintroduction of molecular oxygen since the release NO•. The glutathione detoxification
generation of both radical species requires mechanism may be obtunded by oxidant-me-
molecular oxygen. Although there is substan- diated or pathologically-induced endothelial
tial copper-zinc SOD present in the myocar- injury101 such as in hypercholesterolemia.105
dium, potential oxygen radical scavengers are In addition, ONOO– may stimulate guanylate
depleted during ischemia-reperfusion which cyclase directly or through the formation of
increases the physiological concentrations of NO•. Therefore, ONOO– can serve as an
•O– , and the rapid diffusion-limited reaction
2 alternative NO• donor and as a stimulator of
18 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 1.9. The biradical reaction between nitric oxide NO• and superoxide radical •O–2 is nearly diffusion limited
and mainly irreversible, and requires equimolar concentration of the two substrates. The end product, peroxynitrite
(ONOO–) can be detoxified in blood by nitrosylating thiol compounds such as glutathione (GSH) to form
nitrosothiole, which may recycle to NO•. However, the lack of detoxifying reactions in crystalloid media allows
build-up of ONOO– with protein nitrosylation, DNA damage and formation of the highly reactive hydroxyl
radical. This pathway to injury may amplify the inflammatory response and •O–2 generation, thereby providing
more substrate for ONOO– formation. (Adapted with permission from Xin-Liang Ma, M.D., Ph.D.)

guanylate cyclase activity and have the potential Therefore, ONOO– in acellular environments
to be cardioprotective. would not be detoxified and may therefore
Crystalloid buffer systems (i.e., produce tissue injury, ostensibly either by
Langendorff isolated buffer perfused tissues, direct or •OH related mechanisms, while in
cell culture) do not contain endogenous com- biological environments, ONOO– would not
pounds that may detoxify ONOO–, while exert deleterious effects and may be protec-
plasma and tissue contain these potentially tive through nitrosylation of thiols (such as
detoxifying agents. In addition, buffer systems glutathione) with subsequent NO• donation.
do not contain neutrophils as targets for NO• This conclusion is supported by results pre-
antiaggregatory and antiadhesion actions. sented by Ma et al106 in which isolated hearts
Studies in which ONOO– has been shown to subjected to an ONOO- donor agent (SIN-1)
exert a deleterious effect were performed in in a blood environment were protected while
cell-free buffer perfused models99,101 while the those in a crystalloid environment were
studies which demonstrated a beneficial effect injured. This environmental effect may be im-
of authentic ONOO– were performed in in portant in cardiac surgery in which there are
vivo models in which plasma and tissue borne both crystalloid and blood formulations of car-
detoxifying substances were present or buffer- dioplegia solutions, with strong proponents for
perfused system contained neutrophils.102,103 each. Currently there is great interest in the
Harnessing the Cardioprotective Potential of Nitric Oxide 19

use of NO• donors or precursors (L-arginine) ing and left circumflex epicardial coronary
as supplements to cardioplegia solutions. arteries were excised and placed in organ cham-
Whether a similar dichotomy in the physi- bers to determine endothelial responses to ago-
ological effects of NO• will be observed in this nist stimulators of nitric oxide synthase.
area of surgical myocardial protection has not Coronary arteries from postcardioplegic
yet been reported. Indeed, the deleterious myocardium demonstrated marked differences
effects of L-arginine administered during with regard to both formulations and presence
reperfusion reported by Engleman et al91 for of ONOO–. Generally, blood cardioplegia
isolated crystalloid buffer perfused hearts may demonstrated better preservation of maximal
be related to this environment effect of NO• endothelial responses to acetylcholine
if indeed L-arginine supplementation of the (receptor dependent) than crystalloid car-
crystalloid buffer released enough NO• to dioplegia. The addition of ONOO– to blood
engage this NO•-ONOO-mechanism. cardioplegia demonstrated significantly
The question of whether ONOO– medi- greater responses (better function), while
ates differential effects depending on the blood ONOO– in the crystalloid cardioplegia group
or crystalloid environment was investigated by was associated with significantly poorer func-
Ronson et al107 in a canine model of myocar- tion (Fig. 1.10B). Therefore, ONOO– in a
dial protection with blood or crystalloid- crystalloid medium may indeed be deleteri-
cardioplegia solutions. Hearts were subjected ous, while it is beneficial in a blood medium
to 30 minutes of normothermic global is- with regards to postcardioplegia systolic func-
chemia on cardiopulmonary bypass, followed tion and coronary artery endothelial function.
by 60 minutes of hypothermic hyperkalemic Further investigation is necessary to deter-
intermittent cardioplegia using either crystal- mine the role of NO• and its metabolite
loid (Plegisol) or blood cardioplegia (8:1 ONOO– under different conditions. However,
blood:crystalloid), each with or without the majority of studies in normal or ischemic-
5 ∝mol/L authentic ONOO-, using the Myo- reperfused hearts in which cardioplegia has been
cardial Protection Systems (MPS, Quest Medi- supplemented with NO• donors or the pre-
cal, Inc.) delivery system to ensure accurate cursor L-arginine have reported net cardio-
concentrations of ONOO–. After 2 hours of protective effects of NO•.42,43,108,109
reperfusion, systolic and diastolic function
were determined using left ventricular pres- Nitric Oxide in Hypoxia-
sure-volume (impedance catheter) relations Reoxygenation Injury
analysis (Fig. 1.10A). In the crystalloid car-
Cyanotic heart defects are often corrected
dioplegia groups, the addition of ONOO– was
surgically using cardiopulmonary bypass and
associated with a 57% reduction (versus
elective chemical cardioplegia. Despite the
baseline) of systolic function compared to a
well-known tolerance of the immature heart
44 ± 2% reduction in the crystalloid cardiople-
to ischemia and reperfusion110-112 the imma-
gia group without ONOO– (p < 0.05). In con-
ture myocardium is seemingly intolerant of
trast, postischemic left ventricular function was
hypoxia-reoxygenation using high blood oxy-
generally better than crystalloid cardioplegia
gen tensions (∃ 400 mmHg).113-115 Key char-
with both formulations of blood cardioplegia,
acteristics in hypoxia-reoxygenation, with the
and there was significantly better recovery of
latter being accomplished on cardiopulmonary
systolic performance with BCP plus ONOO-
bypass, are 1) increased production of NO•
compared to BCP without ONOO-.
during the period of reoxygenation,116,117 2) a
Myocardial edema (% tissue water) was
loss if the endogenous antioxidant reserve
significantly increased by ONOO– in crystal-
coupled with an increase in the production of
loid (81.1 ± 0.3 vs 79.6 ± 0.4%) and blood
oxygen-derived free radicals which is amenable
cardioplegia (78.9 ± 0.3% vs 76.4 ± 0.32%)
to antioxidant therapy,118 3) increased produc-
in agreement with the chamber stiffness data.
tion of oxygen-derived free radicals during
After the experiment, the left anterior descend-
reoxygenation, and 4) a reduction in the
20 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 1.10. Physiological ef-


fects of ONOO– in blood
cardioplegia versus crystal-
loid cardioplegia (Plegisol)
environments. Panel A. Re-
covery of end-systolic pres-
sure-volume (impedance
catheter) relations after 2
hours of reperfusion as a
percent of baseline value of
slope of the relationship.
Panel B. Responses of post-
cardioplegia epicardial
coronary arteries to the
endothelial-dependent, re-
ceptor- dependent stimula-
tor of nitric oxide synthase,
acetylcholine at 500 nM or-
gan chamber concentration.
Responses are maximal
percent relaxation from
preconstricted (U46619)
values. In the BLOOD+
group, there was a signifi-
cant left shift in the concen-
tration-response curve com-
pared to BLOOD group
indicative of better function,
while in the PLEG+ group
there was a significant right
shift relative to PLEG group
indicative of receptor dys-
function. PLEG = Plegisol
crystalloid cardioplegia
without ONOO-; PLEG+
= Plegisol crystalloid
cardioplegia with 5∝M
ONOO-; BLOOD = blood
cardioplegia (8:1 blood:
crystalloid) without ONOO–;
BLOOD+ = blood cardio-
plegia without 5 ∝M ONOO-.
* = p<0.05 between indi-
cated groups.

production of oxygen-derived free radicals and beneficial effects. 116,120 In this case of
NO• by low or hypoxic reoxygenation.119 hypoxia-reoxygenation, ONOO—may be
Seemingly paradoxical also is the effect of NO• playing a major role in directing the effects of
in the model of hypoxia-reoxygenation injury. NO•-related therapy, even in the presence of
In contrast to the cardioprotection reported blood-borne detoxification mechanisms. The
for L-arginine or NO•-donor supplemented combination of increased production of both
therapy in ischemia-reperfusion models as dis- NO• and •O–2 during reoxygenation and the
cussed above, the same therapy in hypoxia- presence of reduced antioxidant reserve in the
reoxygenation models has deleterious heart may have permitted the unbridled forma-
effects. 116,120 and inhibitors of NOS had tion of ONOO- with subsequent production
Harnessing the Cardioprotective Potential of Nitric Oxide 21

of •OH, and hence, the deleterious effects. of apoptosis correlated with increased levels
However, the hypothesis that overproduction of c-myc and c-fos, gene products associated
of peroxynitrite in the hypoxic-reoxygenation with induction of mitogenesis and apop-
injury model is responsible or contributory to tosis.140 Other evidence of a protective role for
the deleterious effects of NO• has not been NO• against apoptotic cell death links the in-
tested. In any event, the mechanisms of NO• crease in NO• production by shear stress to
and related therapy are clearly different and decreases in endothelial cell apoptosis. After
independent of issues of different species, exposure of the endothelium to TNF-!, a
methods and laboratories since this same group known stimulator of apoptosis,141 shear stress
showed that in an ischemic-reperfusion model almost completely abrogated the appearance
(20 minutes antecedent normothermic is- of apoptosis compared to cells not subjected
chemia before cardioplegia) using the same to shear forces. This effect on reducing
animal model, that L-arginine augmented apoptosis was mimicked by NO • donors
NO• production and was cardioprotective, and through an inhibition of ICE-like and cysteine
this effect was reversed by NOS inhibitors.121 proteases.142 The potential increase in both
endothelial and myocardial apoptosis in vivo
Nitric Oxide and Myocardial secondary to decreases in endothelial nitric
Apoptosis oxide production and release with endothelial
As summarized above, NO• has important cell dysfunction following ischemia and
and potent modulatory effects on the inflam- reperfusion is not known and needs to be fur-
matory process related to postischemic injury ther evaluated.
and necrosis. Apoptosis, or genetically pro-
grammed cell death, as opposed to the explo- Summary
sive and inflammatory-initiating process of Nitric oxide, the molecule of the decade,
cellular necrosis,122 is known to occur in the is involved in a host of physiological effects,
heart during failure,123 infarction124 and is- particularly in the modulation of neutrophils
chemia-reperfusion of the myocardium125-127 and endothelial cells—both individually and
due to a change in the expression of spe- their interactions—during the complex pro-
cific gene products and production of spe- cesses instigated by ischemia and reperfusion.
cific cytoplasmic proteins.128-131 Some stud- The endothelium is an effective target for aug-
ies127,132,133 have shown that ischemia with menting endogenous NO• because 1) it is the
reperfusion, but not ischemia alone, induces vascular interface between neutrophils, cyto-
apoptosis of the myocardium; hypoxia is also kines and other blood-borne proinflammatory
a stimulus for apoptosis.134 mediators and the underlying tissue (myocytes,
vascular smooth muscle, conductile tissue, ar-
Apoptosis has been inhibited by a number chitectural structures) that are themselves tar-
of interventions including precondition- gets of injury, 2) the endothelium is the larg-
ing.135,136 In contrast, cardioprotective auta- est organ in the body, with an extensive surface
coids other than NO• have been associated area and distribution in organs, 3) it is respon-
with induction of apoptosis.137,138 However, sive to a number of physiological stimuli such
the role nitric oxide plays in the pathophysi- as shear stress. NO• plays a key role in the at-
ological development of apoptosis is not clear. tenuation of ischemic-reperfusion injury at
Lopez-Farre et al139 have demonstrated a link numerous levels of the process (Fig. 1.11), in-
between nitric oxide and endothelial cell cluding direct inhibition of the neutrophil (su-
apoptosis. In cultured endothelial cells, inhi- peroxide anion generation, adhesion molecule
bition of NO• synthesis by L-NAME signifi- expression, adherence to endothelium), direct
cantly increased endothelial cell apoptosis neutralization of superoxide radicals—a pri-
without altering any other environmental con- mary mediator of injury, inhibition of adhe-
dition. Furthermore, the increased expression sion molecule expression on the surface of the
22 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 1.11. The cardioprotective mechanisms of NO• related to inhibition of neutrophil and endothelial cell
interactions during reperfusion. NO• inhibits surface expression of p-selectin (p) and intercellular adhesion
molecule-1 (ICAM-1) and vascular cell adhesion molecule (VCAM). NO• also directly inhibits neutrophils and
superoxide (O•2) generation in addition to neutralizing O•2 via direct quenching. NO• can be augmented by
providing precursor L-arginine (L-arg) or administering direct NO• donor agents.

endothelium, and inhibition of synthesis of a metabolite such as peroxynitrite and hydroxyl


adhesion molecules at the level of nuclear tran- radical, and hemodynamic (vasodilator) con-
scription (NF-#B). Many studies support the sequences may be precipitated that are un-
observation that NO • exerts its cardio- wanted. Although nature has evolved this
protection primarily during the reperfusion pleuripotent mechanism of endogenous de-
phase, which would thereby guide the time fense against inflammatory-like conditions, the
course of its administration. Therapeutic strat- unraveling of mechanisms and harnessing of
egies applicable to ischemic-reperfusion injury this therapeutic potential will occupy scien-
include provision of the precursor to NO•- L- tific and drug development investigations for
arginine, which augments the endogenous a long time to come.
generation of NO• for availability in the com-
partments (intravascular compartment, inter- Acknowledgments
stitial compartment) most involved in the The authors are grateful for the assistance
acute and initiating steps in the inflammatory of Ms. Gail Nechtman in the preparation of
component of ischemic-reperfusion injury. the manuscript. We are indebted to the Carlyle
However, a ceiling in the effectiveness of this Fraser Heart Center for continued support of
approach may be encountered if the endothe- activities in the Cardiothoracic Research Labo-
lium is dysfunctional and impaired in its abil- ratory. Supported in part by a grant from the
ity to synthesize and release NO•. In this event, National Institutes of Health (NHLBI, HL
a wide variety of NO•-donor agents are avail- 46179).
able which may be administered. A word of
caution must be given in that there may be a
definite therapeutic window beyond which
tissue damage may be caused by NO• itself or
Harnessing the Cardioprotective Potential of Nitric Oxide 23

References 16. Yamabe H, Okumura K, Ishizaka H et al.


1. Vinten-Johansen J, Hammon JW, Jr. Car- Role of endothelium-derived nitric oxide in
diopulmonary Bypass: Principles and Practice. myocardial reactive hyperemia. Am J Physiol
In: Utley, Gravlee, eds. New York: Williams 1992; 263(1 Pt 2):H8-14.
& Wilkins 1993:155-206. 17. Lefer AM, Tsao PS, Ma X-L. Shock- and is-
chemia-induced mechanisms of impairment of
2. Vinten-Johansen J, Nakanishi K. Post- endothelium-mediated vasodilation. [Review].
cardioplegia acute cardiac dysfunction and Chest 1991; 100:160S-3S.
reperfusion injury. [Review]. J Cardiothorac 18. Lefer AM, Lefer DJ. Pharmacology of the
Vasc Anesth 1993; 7:6-18. endothelium in ischemia-reperfusion and cir-
3. Robicsek F, Schaper J. Reperfusion injury: culatory shock. Annual Review Pharmacology
Fact or myth? J Card Surg 1997; 12:133-7. and Toxicology 1993; 33:71-90.
4. Vinten-Johansen J. Reperfusion injury: Idle 19. Harrison DG, Kurz MA, Quillen JE et al.
curiosity or therapeutic vector? J Thrombosis Normal and pathophysiologic considerations
Thrombolysis 1997; 4:59-61. of endothelial regulation of vascular tone and
5. Vinten-Johansen J, Zhao Z-Q. Adenosine: their relevance to nitrate therapy. [Review].
Cardioprotection and Clinical Application. In: Am J Cardiol 1992; 70:11B-7B.
Mentzer, Kitakaze, Downey, Hori, eds. 20. Vanhoutte PM. Hypercholesterolaemia, ath-
Kluwer Academic Publishers 1997:49-70. erosclerosis and release of endothelium-de-
6. Chenoweth DE. Pathophysiology and tech- rived relaxing factor by aggregating platelets.
niques of cardiopulmonary bypass. In: Utley, Eur Heart J 1991; 12:25-32.
Betleski, eds. 2 ed. Baltimore: Williams & 21. Harrison DG. Endothelial dysfunction in ath-
Wilkins 1983: 49-60. erosclerosis. [Review]. Bas Res Cardiol 1994;
7. Kubes P, Suzuki M, Granger N. Nitric ox- 89 Suppl 1:87-102.
ide: An endogenous modulator of leukocyte 22. Lüscher TF, Yang Z, Diederich D et al. En-
adhesion. Proc Natl Acad Sci 1991; 88: dothelium-derived vasoactive substances:
4651-5. Potential role in hypertension, atheroscle-
8. Hammon JW, Jr., Vinten-Johansen J. Aug- rosis, and vascular occlusion. J Cardiovasc
mentation of microvascular nitric oxide Pharmacol 1989; 13\4:S63-S69
improves myocardial performance following 23. Cooke JP, Andon NA, Girerd XJ et al. Argi-
global ischemia. J Card Surg 1995; 10:423-7. nine restores cholinergic relaxation of hyper-
9. Lefer AM, Lefer DJ. Therapeutic role of ni- cholesterolemic rabbit thoracic aorta. Circu-
tric oxide donors in the treatment of cardio- lation 1991; 83:1057-62.
vascular disease. Drugs of the Future 1994; 24. Morita K, Ihnken K, Buckberg GD et al.
19:665-72. Pulmonary vasoconstriction due to impaired
10. Lefer DJ. Myocardial protective actions of nitric oxide production after cardiopulmonary
nitric oxide donors after myocardial ischemia bypass. Ann Thorac Surg 1996; 61:1775-80.
and reperfusion. [Review]. New Horiz 1995; 25. Furchgott RF, Zawadzki JV. The obligatory
3:105-12. role of endothelial cells in the relaxation of
11. Stewart JM, Wang J, Hintze TH. Role of arterial smooth muscle by acetylcholine. Na-
EDRF in the regulation of shear rate in large ture 1980; 288.
coronary arteries in conscious dogs. J Mol 26. Schrader J, Kostic MM, Borst M. Role of
Cell Cardiol 1994; 26(12):1625-33. nitric oxide, adenosine and ATP during re-
12. Smith TP, Jr., Canty JM, Jr. Modulation of active hyperemia. [Abstract] Biology of Ni-
coronary autoregulatory responses by nitric tric Oxide 1991; 125.
oxide. Evidence for flow-dependent resistance 27. Tsunoda R, Okumura K, Ishizaka H et al.
adjustments in conscious dogs. Circ Res Enhancement of myocardial reactive hyper-
1993; 73:232-40. emia with manganese- superoxide dismutase:
13. Quyyumi AA, Dakak N, Andrews NP et al. Role of endothelium-derived nitric oxide.
Endothelium-derived factors, Vasodilation. Cardiovasc Res 1996; 31(4):537-45.
Contribution of nitric oxide to metabolic 28. Depre C, Vanoverschelde JL, Goudemant JF
coronary vasodilation in the human heart. et al. Protection against ischemic injury by
Circulation 1995; 92:320-6. nonvasoactive concentrations of nitric oxide
14. Rubanyi GM. The role of endothelium in synthase inhibitors in the perfused rabbit
cardiovascular homeostasis and diseases. J heart. Circulation 1995; 92:1911-8.
Cardiovasc Pharmacol 1993; 23(Suppl 4): 29. Hattler BG, Gorcsan IJ, Shah N et al. A
S1-S14. potential role for nitric oxide in myocardial
15. Gryglewski RJ, Chlopicki S, Niezabitowski P. stunning. J Card Surg 1994; 9:425-9.
Coronary flow, Endothelium, Prostacyclin,
Nitric oxide, Adenosine. Endothelial control
of coronary flow in perfused guinea pig heart.
Bas Res Cardiol 1995; 90:119-24.
24 Ischemia-Reperfusion Injury in Cardiac Surgery

30. Shen W, Xu X, Ochoa M et al. Role of ni- 43. Sato H, Zhao Z-Q, McGee DS et al. Supple-
tric oxide in the regulation of oxygen con- mentation of blood cardioplegia with L-argi-
sumption in conscious dogs. Circ Res 1994; nine avoids regional postischemic injury by
75:1086-95. NO pathway. [Abstract] Ann Thorac Surg
31. Ma X-L, Lopez BL, Christopher TA et al. 1995.
Exogenous NO inhibits basal NO release 44. Hiramatsu T, Forbess JM, Miura T et al.
from vascular endothelium in vitro and in Effects of L-arginine and L-nitro-arginine
vivo. Am J Physiol 1996; 271:H2045-H2051 methyl ester on recovery of neonatal lamb
32. Finkel MS, Oddis CV, Jacob TD et al. Nega- hearts after cold ischemia. Evidence for an
tive inotropic effects of cytokines on the heart important role of endothelial production of
mediated by nitric oxide. Science 1992; nitric oxide. J Thorac Cardiovasc Surg 1995;
257:287-9. 109:81-7.
33. Kawamura T, Wakusawa R, Okada K et al. 45. Guo J-P, Murohara T, Buerke M et al. Di-
Elevation of cytokines during open heart sur- rect measurement of nitric oxide release from
gery with cardiopulmonary bypass: Participa- vascular endothelial cells. J Appl Physiol
tion of interleukin 8 and 6 in reperfusion 1996; 81:774-9.
injury [see comments]. Can J Anaesth 1993; 46. Ma X-L, Weyrich AS, Lefer DJ et al. Dimin-
40:1016-21. ished basal nitric oxide release after myocar-
34. Steinberg JB, Kapelanski DP, Olson JD et dial ischemia and reperfusion promotes neu-
al. Cytokine and complement levels in pa- trophil adherence to coronary endothelium.
tients undergoing cardiopulmonary bypass. J Circ Res 1993; 72:403-12.
Thorac Cardiovasc Surg 1993; 106:1008-16. 47. Engelman DT, Watanabe M, Engleman RM
35. Davies SW, Bailey J, Wedzicha JA et al. et al. Constitutive nitric oxide release is im-
Complement activation and lung permeabil- paired after ischemia and reperfusion. J
ity during cardiopulmonary bypass. Ann Thorac Cardiovasc Surg 1995; 110:1047-53.
Thorac Surg 1991; 51:693-7. 48. Buga GM, Gold ME, Fukuto JM et al. Shear
36. Kirklin JK, Westaby S, Blackstone EH et al. stress-induced release of nitric oxide from
Complement and the damaging effects of endothelial cells grown on beads. Hyperten-
cardiopulmonary bypass. J Thorac Cardiovasc sion 1991; 17:187-93.
Surg 1983; 86:845-57. 49. Cooke JP, Stamler J, Andon N et al. Flow
37. Weyrich AS, Ma X-L, Buerke M et al. Physi- stimulates endothelial cells to release a
ological concentrations of nitric oxide do not nitrovasodilator that is potentiated by reduced
elicit an acute negative inotropic effect in thiol. Am J Physiol-Heart Circ Physiol 1990;
unstimulated cardiac muscle. Circ Res 1994; 259:H804-H812.
75:692-700. 50. Nerem RM, Harrison DG, Taylor WR et al.
38. Hasebe N, Shen Y-T, Vatner SF. Inhibition Hemodynamics and vascular endothelial bi-
of endothelium-derived relaxing factor en- ology. [Review]. J Cardiovasc Pharmacol
hances myocardial stunning in conscious 1993; 21 Suppl 1:S6-10.
dogs. Circulation 1993; 88:2862-71. 51. Weyrich AS, Ma X-L, Lefer AM. The role
39. Kelm M, Schrader J. Control of coronary of L-arginine in ameliorating reperfusion in-
vascular tone by nitric oxide. Circ Res 1990; jury after myocardial ischemia in the cat.
66:1561-75. Circulation 1992; 86:279-88.
40. Nakanishi K, Zhao Z-Q, Vinten-Johansen J 52. Beckman JS, Koppenol WH. Nitric oxide,
et al. Blood cardioplegia enhanced with ni- superoxide, and peroxynitrite: The good, the
tric oxide donor SPM-5185 counteracts pos- bad, and the ugly. Am J Physiol 1996;
tischemic endothelial and ventricular dysfunc- 271:C1424-C1437
tion. J Thorac Cardiovasc Surg 1995; 109: 53. Beckman JS, Beckman TW, Chen J et al.
1146-54. Apparent hydroxyl radical production by
41. Lefer DJ, Nakanishi K, Vinten-Johansen J. peroxynitrite: Implications for endothelial
Endothelial and myocardial cell protection by injury from nitric oxide and superoxide. Proc
a cysteine- containing nitric oxide donor af- Natl Acad Sci USA 1990; 87:1620-4.
ter myocardial ischemia and reperfusion. J 54. Gryglewski RJ, Palmer RMJ, Moncada S.
Cardiovasc Pharmacol 1993; 22 Suppl 7: Superoxide anion is involved in the break-
S34-43. down of endothelium-derived relaxing factor.
42. Hiramatsu T, Forbess JM, Miura T et al. Nature 1986; 320:454-60.
Effect of L-Arginine cardioplegia on recovery 55. Rubanyi GM, Vanhoutte PM. Superoxide
of neonatal lamb hearts after 2 hours of anions and hyperoxia inactivate endothelium-
cold ischemia. Ann Thorac Surg 1995; 60: derived relaxing factor. Am J Physiol 1986;
1187-92. 250:H822-H827
Harnessing the Cardioprotective Potential of Nitric Oxide 25

56. Rogers NE, Ignarro LJ. Constitutive nitric 69. Lefer DJ, Nakanishi K, Vinten-Johansen J et
oxide synthase from cerebellum is reversibly al. Cardiac venous endothelial dysfunction
inhibited by nitric oxide formed from L-argi- after myocardial ischemia and reperfusion in
nine. Biochem Biophys Res Commun 1992; dogs. Am J Physiol 1992; 263:H850-H856
189:242-9. 70. Boyle EM, Pohlman TH, Cornejo CJ et al.
57. Buga GM, Griscavage JM, Rogers NE et al. Endothelial cell injury in cardiovascular sur-
Negative feedback regulation of endothelial gery: Ischemia-reperfusion. Ann Thorac Surg
cell function by nitric oxide. Circ Res 1993; 1996; 62:1868-75.
73:808-12. 71. Moore KL, Patel KD, Bruehl RE et al.
58. Tsao PS, Aoki N, Lefer DJ et al. Time course P-selectin glycoprotein ligand-1 mediates roll-
of endothelial dysfunction and myocardial ing of human neutrophils on p-selectin. J Cell
injury during myocardial ischemia and Biol 1995; 128:661-71.
reperfusion in the cat. Circulation 1990; 72. McEver RP, Cummings RD. Role of PSGL-
82:1402-12. 1 Binding to selectins in Leukocyte Recruit-
59. Tsao PS, Lefer AM. Time course and mecha- ment. J Clin Invest 1997; 100:485-92.
nism of endothelial dysfunction in isolated 73. Davenpeck KL, Gauthier TW, Albertine KH
ischemic- and hypoxic-perfused rat hearts. Am et al. Role of P-selectin in microvascular leu-
J Physiol 1990; 259:H1660-H1666 kocyte-endothelial interaction in splanchnic
60. Dignan RJ, Dyke CM, Abd-Elfattah AS et ischemia-reperfusion. Am J Physiol 1994;
al. Coronary artery endothelial cell and 267:H622-30.
smooth muscle dysfunction after global myo- 74. Geng J-G, Bevilacqua MP, Moore KL et al.
cardial ischemia. Ann Thorac Surg 1992; Rapid neutrophil adhesion to activated endot-
53:311-7. helium mediated by GMP-140. Nat 1990;
61. Nakanishi K, Zhao Z-Q, Vinten-Johansen J 343:757-60.
et al. Coronary artery endothelial dysfunction 75. Jerome SN, Dore M, Paulson JC et al. P-
after ischemia, blood cardioplegia, and selectin and ICAM-1-dependent adherence
reperfusion. Ann Thorac Surg 1994; 58: reactions: Role in the genesis of postis-
191-9. chemic no-reflow. Am J Physiol 1994;
62. VanBenthuysen KM, McMurtry IF, Horwitz 266: H1316-21.
LD. Reperfusion after acute coronary occlu- 76. Kubes P, Jutila M, Payne D. Therapeutic
sion in dogs impairs endothelium-dependent potential of inhibiting leukocyte rolling in
relaxation to acetylcholine and augments con- ischemia/reperfusion. J Clin Invest 1995;
tractile reactivity in vitro. J Clin Invest 1987; 95:2510-9.
79:265-74. 77. DeCaterina R, Libby O, Peng H et al. Ni-
63. Nakanishi K, Vinten-Johansen J, Lefer DJ et tric oxide decreases cytokine-induced endot-
al. Intracoronary L-arginine during reper- helial activation. J Clin Invest 1995; 96:60-8.
fusion improves endothelial function and re- 78. Ohashi Y, Kawashima S, Hirata K et al. Ni-
duces infarct size. Am J Physiol 1992; tric oxide inhibits neutrophil adhesion to
263:H1650-H1658. cytokine-activated cardiac myocytes. Am J
64. Lefer AM, Tsao PS, Lefer DJ et al. Role of Physiol 1997; 272:H2807-H2814
endothelial dysfunction in the pathogenesis of 79. Dreyer WJ, Michael LH, West MW et al.
reperfusion injury after myocardial ischemia. Neutrophil accumulation in ischemic canine
FASEB J 1991; 5:2029-34. myocardium: Insights into time course, dis-
65. Kaeffer N, Richard V, Francois A et al. Pre- tribution, and mechanism of localization dur-
conditioning prevents chronic reperfusion-in- ing early reperfusion. Circulation 1991;
duced coronary endothelial dysfunction in 84:400-11.
rats. Am J Physiol 1996; 271:H842-H849 80. Entman ML, Michael L, Rossen RD et al.
66. Seccombe JF, Schaff HV. Coronary artery Inflammation in the course of early myocar-
endothelial function after myocardial ischemia dial ischemia. FASEB J 1991; 5:2529-37.
and reperfusion. [Review]. Ann Thorac Surg 81. Wilson I, Gillinov AM, Curtis WE et al.
1995; 60:778-88. Inhibition of neutrophil adherence improves
67. Seccombe JF, Pearson PJ, Schaff HV. Oxy- postischemic ventricular performance of the
gen radical-mediated vascular injury selectively neonatal heart. Circulation 1993; 88:II372-9.
inhibits receptor-dependent release of nitric 82. Byrne JG, Smith WJ, Murphy MP et al.
oxide from canine coronary arteries. J Thorac Complete prevention of myocardial stunning,
Cardiovasc Surg 1994; 107:505-9. contracture, low- reflow, and edema after
68. Lefer AM, Ma X-L, Weyrich A et al. Endot- heart transplantation by blocking neutrophil
helial dysfunction and neutrophil adherence adhesion molecules during reperfusion. J
as critical events in the development of Thorac Cardiovasc Surg 1992; 104:1589-96.
reperfusion injury. Agents Actions Suppl.
1993; 41:127-35.
26 Ischemia-Reperfusion Injury in Cardiac Surgery

83. Sato H, Zhao Z-Q, McGee DS et al. Supple- 95. Johnson G, III, Tsao PS, Mulloy D et al.
mental L-arginine during cardioplegic arrest Cardioprotective effects of acidified sodium
and reperfusion avoids regional postischemic nitrite in myocardial ischemia with reper-
injury. J Thorac Cardiovasc Surg 1995; fusion. J Pharmacol Exp Thera 1990; 252:
110:302-14. 35-41.
84. Sato H, Zhao Z-Q, Vinten-Johansen J. 96. Lefer DJ, Nakanishi K, Johnston WE et al.
L-arginine inhibits neutrophil adherence and Antineutrophil and myocardial protection
coronary artery dysfunction. Cardiovasc Res actions of a novel nitric oxide donor after
1996; 31:63-72. acute myocardial ischemia and reperfusion in
85. Palmer RMJ, Rees DD, Ashton DS et al. dogs. Circulation 1993; 88:2337-50.
L-arginine is the physiological precursor for 97. Xie YW, Wolin MS. Role of nitric oxide and
the formation of nitric oxide in endothelium- its interaction with superoxide in the suppres-
dependent relaxation. Biochem Biophys Res sion of cardiac muscle mitochondrial respira-
Comm 1988; 153:1251-6. tion. Circulation 1996; 94:2580-6.
86. Sato H, Zhao Z-Q, Jordan JE et al. Basal 98. Szabo C, Farago M, Horvath I et al. Hem-
nitric oxide expresses endogenous cardio- orrhagic hypotension impairs endothelium-
protection during reperfusion by inhibition of dependent relaxations in the renal artery of
neutrophil-mediated damage after surgical the cat. Circulatory Shock 1992; 36(3):
revascularization. [In Press] J Thorac 238-41.
Cardiovasc Surg 1996. 99. Lopez BL, Liu G-L, Christopher TA et al.
87. Sato H, Zhao Z-Q, Jordan JE et al. Basal Peroxynitrite, the product of nitric oxide and
nitric oxide expresses endogenous cardio- superoxide, causes myocardial injury in the
protection during reperfusion by inhibition of isolated perfused rat heart. Cor Art Dis 1997;
neutrophil-mediated damage after surgical 8:149-53.
revascularization. J Thorac Cardiovasc Surg 100. Schulz R, Wambolt R. Nitric oxide, Myo-
1997; 113:399-409. cardial ischemia, Reperfusion, L-name,
88. Williams MW, Taft CS, Ramnauth S et al. L-nmma, L-arginine, Stunning, Rabbit heart.
Endogenous nitric oxide (NO) protects Inhibition of nitric oxide synthesis protects
against ischaemia- reperfusion injury in the the isolated working rabbit heart from is-
rabbit. Cardiovasc Res 1995; 30:79-86. chaemia-reperfusion injury. Cardiovasc Res
89. Hudspeth DA, Nakanishi K, Vinten-Johansen 1995; 30:432-9.
J et al. Adenosine in blood cardioplegia pre- 101. Ma X-L, Lopez BL, Liu G-L et al. Peroxy-
vents postischemic dysfunction in ischemically nitrite aggravates myocardial reperfusion in-
injured hearts. Ann Thorac Surg 1994; jury in the isolated perfused rat heart.
58:1637-44. Cardiovasc Res 1997.
90. Engelman DT, Watanabe M, Maulik N et 102. Nossuli TO, Hayward R, Scalia R et al.
al. L-arginine reduces endothelial inflamma- Peroxynitrite reduces myocardial infarct size
tion and myocardial stunning during ischemia and preserves coronary endothelium after is-
reperfusion. Ann Thorac Surg 1995; 60: chemia and reperfusion in cats. Circulation
1275-81. 1997; 96:2317-24.
91. Engelman DT, Watanabe M, Maulik N et 103. Lefer DJ, Scalia R, Campbell B et al. Peroxy-
al. Critical timing of nitric oxide supplemen- nitrite inhibits leukocyte-endothelial cell in-
tation in cardioplegic arrest and reperfusion. teractions and protects against ischemia-
Circulation 1996; 94(suppl II):II-407-II-411 reperfusion injury in rats. J Clin Invest 1997;
92. Hiramatsu T, Forbess JM, Miura T et al. 99:684-91.
Additive effects of L-arginine infusion and 104. Stamler JS, Jaraki O, Osborne J et al. Nitric
leukocyte depletion on recovery after hypo- oxide circulates in mammalian plasma prima-
thermic ischemia in neonatal lamb hearts. J rily as an S-nitroso adduct of serum albumin.
Thorac Cardiovasc Surg 1995; 110:172-9. Proc Natl Acad Sci 1992; 89:7674-7.
93. Amrani M, Chester AH, Jayakumar J et al. 105. Ma X-L, Lopez BL, Liu G-L et al. Hyperc-
L-arginine reverses low coronary reflow and holesterolemia impairs a detoxification mecha-
enhances postischaemic recovery of cardiac nism against peroxynitrite and renders the
mechanical function. Cardiovasc Res 1995; vascular tissue more susceptible to oxidative
30:200-4. injury. Circ Res 1997; 80:894-901.
94. Johnson G, III, Tsao PS, Lefer AM. Cardio- 106. Lopez BL, Christopher TA, Ma X-L. The
protective effects of authentic nitric oxide in effects of peroxynitrite on the myocardium
myocardial ischemia with reperfusion. Crit during ischemia-reperfusion are dependent
Care Med 1991; 19:244-52. upon the biological environment. [Abstract]
Endothel 1995; 3:76
Harnessing the Cardioprotective Potential of Nitric Oxide 27

107. Ronson RS, Thourani VH, Ma X-L et al. The 112. Hammon JW, Jr., Boucek RJ, Jr. Myocar-
nitric oxide product peroxynitrite is benefi- dial Protection in Cardiac Surgery. In: Rob-
cial in blood cardioplegia, but is deleterious erts, ed. New York: Marcel Dekker, Inc.;
in crystalloid cardioplegia in vivo. [In Press] 1987: 283-294.
Circulation (Suppl) 1998. 113. Julia P, Kofsky ER, Buckberg GD et al. Stud-
108. Nakanishi K, Zhao Z-Q, Vinten-Johansen J ies of myocardial protection in the immature
et al. Blood cardioplegia enhanced with an heart. III. Models of ischemic and hypoxic/
organic nitric oxide donor reverses postis- ischemic injury in the immature puppy heart.
chemic ventricular dysfunction and coronary J Thorac Cardiovasc Surg 1991; 101:14-22.
endothelial damage. Surg Forum 1993; 114. Ihnken K, Morita K, Buckberg GD et al.
44:245-7. Studies of hypoxemic/reoxygenation injury:
109. Nakanishi K, Zhao Z-Q, Vinten-Johansen J Without aortic clamping. III. Comparison of
et al. Blood cardioplegia enhanced with the the magnitude of damage by hypoxemia/
nitric oxide donor SPM-5185 counteracts reoxygenation versus ischemia/reperfusion. J
postischemic endothelial and ventricular dys- Thorac Cardiovasc Surg 1995; 110:1182-9.
function. J Thorac Cardiovasc Surg 1995; 115. Ihnken K, Morita K, Buckberg GD et al.
109:1146-54. Studies of hypoxemic/reoxygenation injury:
110. Aoki M, Kawata H, Mayer JE, Jr. Coronary Without aortic clamping. II. Evidence for
endothelial injury by cold crystalloid cardio- reoxygenation damage. J Thorac Cardiovasc
plegic solution in neonatal lambs. Circulation Surg 1995; 110:1171-81.
1992; 86:II346-51. 116. Morita K, Sherman MP, Buckberg GD et al.
111. Kohman LJ, Veit LJ. Neonatal myocardium Studies of hypoxemic/reoxygenation injury:
resists reperfusion injury. J Surg Res 1991; Without aortic clamping. V. Role of the L-
51:133-7. arginine-nitric oxide pathway: The nitric ox-
ide paradox. J Thorac Cardiovasc Surg 1995;
110:1200-11.
CHAPTER 2

The Cellular Basis of Immediate


Lethal Reperfusion Injury
H. M. Piper and D. García-Dorado

I
n cardiac surgery, myocardium may Lethal Reperfusion Injury:
suffer from ischemia either because it has Definition of Terms
been ischemic prior to the surgical
Lethal reperfusion injury is defined as
intervention or because it is made intention-
injury caused by restoration of blood flow after
ally or becomes inadvertently ischemic dur-
an ischemic episode leading to death of cells
ing the intervention. As a result of a success-
that were only reversibly injured during that
ful revascularization or the end of the surgical
preceding ischemic episode. For lethal
manipulations, the ischemic myocardium may
reperfusion injury to occur, ischemia has to
be reperfused. Ischemic or postischemic loss
set the stage without producing irreversible
of viable myocardium is one of the major foes
injury already itself. The definition thus comes
impairing an improvement of cardiac func-
with the corollary that ischemic alterations of
tion after cardiac surgery. Many procedures
cellular conditions are necessary prerequisites
have been designed for the purpose to protect
for lethal reperfusion injury, but not in them-
myocardium against ischemic injury. The pos-
selves sufficient causes for cell death. When
sibility—and need—to protect myocardium
after extended ischemia myocardium is
during the first minutes to hours of reperfusion
reperfused by simple restoration of coronary
against “reperfusion injury” has been largely
blood flow, analysis of the developing necro-
neglected. This may seem surprising as the
sis does not permit to distinguish between cell
cardiac surgeon can control precisely the con-
death caused by the ischemia or by reperfusion.
ditions of reperfusion. This chapter is focussed
The only valid criterion for the existence of
on the possibilities of protecting myocardial
reperfusion injury is whether modification of
cells from cell death specifically by modifying
the conditions of reperfusion can prevent cell
the early conditions of reperfusion, i.e., to pro-
death, otherwise occurring in ischemic-
tect against the early causes of lethal reper-
reperfused myocardium. This criterion appears
fusion injury. It describes the novel strategies
simpler than it is to apply, since many modifi-
developed during the past decade in experi-
cations of reperfusion conditions are possible
mental research. The chapter does not deal
and failure to find one that reduces cell death
with two other closely related topics which
in reperfused myocardium does not disprove
have been discussed extensively in several other
the existence of reperfusion injury. As we ar-
recent reviews. These are: reversible postis-
gue below, causes of certain forms of lethal
chemic dysfunction, so-called stunning, and
reperfusion injury have now been identified
delayed causes of lethal reperfusion injury, e.g.,
and thus some modifications of the conditions
by activation of blood-borne factors.
of reperfusion are indeed known to provide

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


The Cellular Basis of Immediate Lethal Reperfusion Injury 29

protection. If such a modification preventing The impact of oxygen radicals on the myocar-
cell death in reperfused myocardium has been dial cells after ischemia may be increased since
identified, yet another question may be raised. their antioxidative defense is reduced. Oxy-
In experimental models, cell death is normally gen radicals are highly reactive and therefore
assessed within a few hours of reperfusion. It potentially very toxic molecules. It is there-
therefore remains possible that the tested fore understandable that they seemed to be a
modifications of reperfusion conditions only prime candidate for cause of lethal reperfusion
delays the full expression of cell death beyond injury. A large number of experimental stud-
the time of assessment and thus falsely sug- ies has been performed in which ischemic-
gests prevention of lethal reperfusion injury. reperfused myocardium has been treated by
The whole topic of lethal reperfusion injury means directed against oxygen radicals. Super-
can be differentiated conceptually as follows: oxide dismutase either alone or in combina-
Reversible injury must be delineated from tion with catalase upon reperfusion has been
lethal, irreversible injury. In this overview we used in experimental studies aiming at reduc-
will concentrate on the development of the tion of infarct size by reperfusion protection.
acute form of cell death, i.e., necrosis occur- In a smaller number of studies chemical anti-
ring within the first minutes to first few hours oxidants were used for the same purpose.
of reperfusion. In the normal clinical setting, Several reviews from the past ten years have
myocardial injury due to ischemia-reperfusion summarized these studies and came to the
may appear as a single entity. For the under- sobering conclusion that the experimental
standing of reperfusion injury, ischemic injury studies have failed to prove the hypothesis that
must be clearly separated and the dependency oxygen radicals are crucially involved in the
of reperfusion injury on preceding ischemic genesis of lethal reperfusion injury.1-5 This
changes must be clarified. Injury of ischemic- conclusion was drawn because there is about
reperfused myocardium is complex, involving an equal number of studies demonstrating
injury of vascular cells as well as cardio- improvement or no improvement of infarct
myocytes. We will here primarily discuss size when antioxidant strategies are applied
whether lethal reperfusion injury of cardio- during reperfusion. Part of the discrepancies
myocytes (as opposed to other cells in myo- among results may be explained by experimen-
cardium) can occur during the first minutes tal flaws but a large part seems due to differ-
after reflow in the setting of ischemia- ences in the experimental models used. The
reperfusion, i.e., we will concentrate on im- general feeling is that a phenomenon depend-
mediate lethal (necrotic) reperfusion injury. ing so much on the specific features of an
This can be distinguished from delayed lethal experimental model is not of general relevance.
(necrotic) reperfusion injury which, e. g., may This is a plausible opinion. It must be empha-
be delivered to the cardiomyocytes by activated sized, however, that the lack of unequivocal
polymorphic neutrophils, and also from evidence from experimental animal studies
induction of apoptosis of cardiomyocytes in does not exclude the possibility that the human
reperfused myocardium. case represents one of the positive cases where
oxygen radicals contribute to myocardial
A Short Note on the Role reperfusion injury.
of Oxygen Radicals Many researchers who had tried hard to
prove the importance of oxygen radicals have
There is clear evidence that in reperfused
thereafter lost hope altogether that myocar-
myocardium oxygen radicals are generated at
dium could be protected at the time of
a higher rate than in normal myocardium.
reperfusion or, in other words, they lost con-
Several sources seem to contribute to that
fidence in the existence of lethal reperfusion
increased production of oxygen radicals:blood
injury. This frustration does usually not apply
borne cells such as activated neutrophiles as
to reversible reperfusion injury. In fact, even
well as constitutive cells of the myocardium
those researchers who feel that there is no such
such as cardiomyocytes and endothelial cells.
30 Ischemia-Reperfusion Injury in Cardiac Surgery

thing as lethal reperfusion injury would nor- as a permissive factor for hypercontracture elic-
mally admit that there are transient aggrava- ited by re-energization and can also contrib-
tions of myocardial metabolic or functional ute to further Ca2+ overload. Cell swelling is
disorder during reperfusion which are caused the other major final cause for immediate
by the reperfusion situation. Mechanical stun- lethal reperfusion injury. It originates in the
ning and reperfusion arrhythmias are such reperfusion situation from a too rapid normal-
phenomena.6 In the pathogenesis of these ization of extracellular osmolality (cause 3),
reversible dysfunctions oxygen radicals seem leaving the intercellular fluid hyperosmolar.
in fact to play an important part. A critical
discussion of these forms of reversible Role of Re-Energization
reperfusion injury is also interesting but not About two decades ago Hearse and co-
the objective of this chapter. workers7 demonstrated that in the oxygen
depleted and reoxygenated myocardium severe
Three Initial Causes myocardial injury, characterized by myofibril-
of Immediate Lethal lar hypercontracture and sarcolemmal disrup-
tion, may develop with the onset of reoxy-
Reperfusion Injury genation. It has been demonstrated by Ganote
We address the question whether causes for
and co-workers8,9 that this injury is due to the
immediate lethal reperfusion injury of
resumption of energy production upon
cardiomyocytes exist in mammalian hearts and
reoxygenation. This phenomenon of severe cell
how one can interfere with their mechanisms.
injury immediately provoked by re-ener-
Since specific strategies against immediate
gization has been termed “oxygen paradox”.
reperfusion injury have never been applied to
It has remained an open question for a long
human myocardium in vivo except for
time whether the oxygen paradox represents
antioxidative treatment, it must remain open
genuine “reoxygenation injury” or just a dra-
now if reperfusion protection represents a use-
matic manifestation of injury that has already
ful strategy in human therapy. Three poten-
developed during the oxygen depletion period.
tial initial causes of immediate reperfusion
The presence of contraction bands in infarcted
injury, apart from oxygen radicals, have been
myocardium is a histological indicator of oxy-
experimentally investigated in considerable
gen paradox injury in ischemic-reperfused
detail, and will be briefly discussed:
myocardium. Histologic analysis clearly dem-
cause 1: Re-energization, onstrates that when reperfusion is performed
cause 2: Rapid normalization of tis- early enough to produce some myocardial sal-
sue pH, vage, infarcts are composed almost exclusively
cause 3: Rapid normalization of tis- of contraction band necrosis reflecting
sue osmolality. hypercontracture of myocytes, and there is
These potential initial causes are not evidence indicating that this hypercontracture
entirely independent. As outlined below occurs during the first minutes of reflow. Al-
mechanical disruption of the sarcolemma though contraction bands can be observed in
appears to be the endpoint of immediate lethal the absence of necrosis in specific circum-
reperfusion injury. Hypercontracture of the stances (as an artifact in biopsies), in the setting
myofibrils is probably one of the major final of reperfusion hypercontractured myocytes
causes. Hypercontracture is made possible by invariably present signs of necrosis, indicat-
re-energization of the ischemic cell (cause 1) ing that, as opposed to what happens in iso-
in which destructive contractile forces are gen- lated cardiomyocytes, reperfusion-induced
erated due to Ca2+ overload and increased hypercontracture is associated to sarcolemmal
cytoskeletal fragility. Ischemic acidosis can disruption and cell death.
attenuate this contractile activation. Rapid Details of the causal mechanism of the
normalization of tissue pH (cause 2) can act oxygen paradox have now been identified in
The Cellular Basis of Immediate Lethal Reperfusion Injury 31

experimental studies using isolated cardio- the sarcolemma is rapidly activated to remove
myocytes (see below). These studies have access Na+ from the interior of the cell. It was
shown that the oxygen paradox is indeed injury shown on the cellular level that the re-ener-
1. brought about by the process of reoxy- gized cardiomyocyte can retain sufficient meta-
genation and bolic competence to rapidly reactivate the SR
2. based on a mechanism within the myo- Ca2+ pump and the sarcolemmal Na+ pump
cardial cell. during the early phase of reoxygenation, even
Re-energization causes lethal cell injury by if the cell had been extensively depleted of its
provocation of hypercontracture. The mecha- energy stores and suffered from severe Ca2+
nism is the following: After prolonged energy and Na+ overload before re-energization.10-12
depletion, cytosolic Ca2+ concentration is dra- It seems that cells in which these pumps have
matically increased. Upon re-energization of been crucially damaged during the ischemic
the myocardial cell, made possible by resup- period are in principle unable to recover and
ply of oxygen to mitochondria, two processes cannot be, therefore, subject to reperfusion
are simultaneously activated: injury in the sense of the definition given above
1. The energy supply to cation pumps (Fig. 2.2).
initiates recovery of the cellular cation It is the resupply of energy to the myo-
balance; fibrilar elements in the presence of an increase
2. resupply of energy to the myofibrillar of cytosolic Ca2+ concentration which may
elements initiates contractile activation. become deleterious for the reoxygenated cell
Under conditions of energy depletion, i.e., (Fig. 2.1, lower part). This is because during
in ischemic or hypoxic myocardium, the cy- the initial phase of reoxygenation the cytosolic
tosol of the myocardial cells becomes loaded Ca2+ is still largely elevated and myofibrillar
with Na+ and Ca2+. Recovery of energy pro- activation therefore leads to uncontrolled,
duction upon the resupply with oxygen and excessive force generation. This sustained force
metabolic substrates rapidly reactivates two generation causes hypercontraction. A
major cation pumps (Fig. 2.1): namely the hypercontracting cardiac muscle cell becomes
Ca2+ pump (Ca2+-ATPase) of the sarcoplasmic severely injured in its cytoskeletal structures
reticulum (SR) and the Na+ pump (Na+-K+- as the deformation of cytoskeletal elements
ATPase) of the sarcolemma, unless these beyond the degree found under normal con-
pumps are themselves injured by the preced- tractile shortening is no longer readily revers-
ing ischemic conditions. Activation of the Ca2+ ible. The resulting state of irreversible cell
pump of the SR leads to a temporary seques- shortening is called “hypercontracture.” In tis-
tration of excess Ca2+ within this intracellular sue, hypercontraction of adjacent cells may
storage organelle.10,11 If the capacity of this lead to mutual cellular disruptions and necro-
organelle is too small for the amount of Ca2+ sis. This pathomechanism of reoxygenation-
accumulated in the cytosol, a cycle of continu- induced mechanical injury can be prevented
ous release and reuptake of Ca2+ from and into if the contractile machinery is inhibited dur-
the SR is initiated. These spontaneous oscilla- ing the first stage of energy recovery, for the
tions come to an end only if the major mecha- time needed to reestablish a normal cellular
nism for Ca2+ extrusion from the cytosol is cation control. It has been demonstrated in
sufficiently activated, i.e., the Na +/Ca 2+ several studies that a direct blocker of the
exchanger of the sarcolemma.11 The ability of myofibrils, 2,3-butanedionemonoxime
this exchanger to remove Ca2+ from the cyto- (BDM), can be used experimentally to inhibit
sol depends on the magnitude of the trans- the myofibrillar machinery during the early
sarcolemmal Na+ gradient. Restoration of a “vulnerable phase” of reoxygenation.13-17 These
sufficiently large Na+ gradient across the sar- studies from different groups have involved
colemma is therefore the prerequisite for different models (isolated myocytes,13 isolated
extrusion of Ca2+ from reoxygenated myocar- rat heart,14 isolated guinea pig heart,17 in situ
dial cells. It is essential that the Na+ pump of pig15 and dog heart16), different conditions
32 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 2.1. Scheme of cation control and initiation of hypercontracture in the reoxygenated cardiomyocyte. On
reactivation of oxidative phosphorylation in mitochondria (MITO), the Na+ pump (1) is reactivated generating
a transsarcolemmal Na+ gradient which provides a driving force for the Na+/Ca2+ exchanger (2) in its “forward
mode”. Re-energization of the Ca2+ pump (3) of the sarcoplasmic reticulum (SR) causes sequestration of excessive
cytosolic Ca2+ into this organelle. Overload of the SR leads to Ca2+ release. Repetitive Ca2+ uptake and release by
the SR results in Ca2+ cycling between SR and cytosol. Reactivation of oxidative phosphorylation in mitochondria
provides energy also to the contractile machinery. Energy supply in the presence of high cytosolic Ca2+ concen-
tration causes uncontrolled contractile activation and consecutively mechanical injury of cell structures.

(anoxia reoxygenation,13,14 ischemia-reperfusion17 to reestablish a normal cation control, the ini-


transient coronary occlusion15,16) and different tial phase of Ca2+ recovery in the reoxygenated
end-points including ventricular function,17 cell may be divided into two stages:
hypercontracture,13 histochemically deter- 1. An early stage during which the cyto-
mined infarct size15,16 or extension of myo- solic Ca2+ level falls due to uptake of
cardial necrosis as assessed by quantitative his- Ca2+ into the SR and
tology after 24 hours of reperfusion.15 In the 2. a second stage during which Ca2+ is
study by García-Dorado et al,15 for example, shifted in an oscillatory manner be-
the left descending coronary artery was oc- tween cytosol and SR until a sufficient
cluded for 45 min in an in vivo model (pig) of proportion of the Ca2+ level is extruded
regional ischemia. Upon reperfusion, BDM across the sarcolemma.10 The Ca2+ os-
was added to the coronary flow and remained cillations of this stage can also cause
there for the first 60 min of normoxic hypercontraction. This is not solely ex-
reperfusion. Infarct size determinations after plained, however, by the magnitude of
24 hours of reperfusion then demonstrated a the Ca2+ peak concentrations occurring
reduction by half in BDM treated hearts. during oscillations. It has been shown
It has recently been demonstrated that that the susceptibility of reoxygenated
hypercontraction may also be elicited by a cardiomyocytes to develop hyper-
closely related mechanism.19 In cells capable contracture at a given elevation of
The Cellular Basis of Immediate Lethal Reperfusion Injury 33

Fig. 2.2. Scheme of Na+ and Ca2+ control in the reoxygenated cardiomyocyte. Upper half: When reactivation of
the Na+ pump (1) creates a large transsarcolemmal Na+ gradient and the membrane potential (mV) is large, the
Na+/Ca2+ exchanger (2) is driven in its “forward mode”. The cytosolic Ca2+ overload may thus be reduced. Lower
half: When Na+ influx through Na+/H+ exchanger (3), Na+/HCO3- symporter (4) or Na+ channels (5) diminishes
the transsarcolemmal Na+ gradient and the membrane potential is small, the Na+/Ca2+ exchanger is driven in its
“reverse mode”. The cytosolic Ca2+ overload may thus be increased.

cytosolic Ca2+ is increased after a pro- after short-lived ischemia, exhibiting


longed period of hypoxic energy depletion.18 stunning, have shown rather a reduc-
This means that hypercontraction in tion of myofibrillar Ca2+ sensitivity.19
energy depleted and repleted myocar- In vitro it is possible to protect the
dial cells may be elicited by Ca2+ con- reoxygenated myocardial cell from
centrations in the cytosol which would hypercontracture by damping the os-
not cause harm to a normal cell. The cillatory movements between Ca2+ and
cause for this increased susceptibility cytosol, thus reducing the high peak
seems to reside in an increased fragility concentrations of cytosolic Ca2+.18 This
of cytoskeletal elements which can no can be achieved by specific blockade of
longer resist large contraction forces. An Ca2+ uptake into or release from the SR,
alternative explanation would be that as with cyclopiazonic acid or ryanodine,
the myofibrillar sensitivity to Ca2+ is in- respectively.19 Interestingly, the volatile
creased in reoxygenated cardio- anesthetic halothane can also be used
myocytes. This has not yet been stud- to inhibit SR function and thereby pro-
ied directly. It seems unlikely, however, vide protection.20 Halothane applied
since studies on reperfused myocardium upon reoxygenation has been shown to
34 Ischemia-Reperfusion Injury in Cardiac Surgery

protect isolated cardiomyocytes, hy- cardial cells are allowed to restore a normal
poxia-reoxygenated hearts21 and is- intracellular acid-base balance. It has been
chemic-reperfused in vivo22 myocar- demonstrated in vitro that continuation of
dium against hypercontracture and extracellular acidosis, and thereby intracellu-
lethal reperfusion injury. lar acidosis, during the early phase of
reoxygenation protects myocardial cells against
Role of Rapid Normalization the development of hypercontracture during
of Tissue pH this phase. For reperfusion of myocardium in
The cytosolic pH in cardiomyocytes in vivo the situation is less clear. Inhibitors of the
reperfused myocardium has a pronounced Na +/H+ exchanger were found to protect
influence on the development of hyper- against the development of hypercontracture
contracture. After prolonged ischemia, the and necrosis during reperfusion only when
cytosolic pH is markedly lowered because present during the previous ischemic
anaerobic metabolism and the breakdown of period.26-28 The most likely explanation for the
ATP produce an excess of H+. This leads to an discrepancy between the in vitro and the in
acidification of both the intracellular and the vivo studies is at present that in blood per-
interstitial spaces. Upon reperfusion the pH fused hearts intracellular acidosis cannot be
in the interstitial space is rapidly renormalized maintained for a sufficiently long time after
and a gradient is thus generated between the initiation of reperfusion if only the Na+/H+
cytosol, containing still high H+ concentra- exchanger is inhibited. This is because the
tions, and the interstitium, where the H+ con- myocardial cell possesses also another route for
centrations are already re-normalized. This the transsarcolemmal extrusion of acid equiva-
causes an activation of the H+ extruding lents, i.e., the Na+/HCO3– symporter which
mechanisms of the cardiomyocytes, i. e., the works in parallel to the Na+/H+ exchanger and
Na + /H + exchanger and the Na + /HCO 3 - is active in normal bicarbonate containing flu-
symporter.23,24 This process has two conse- ids. Unfortunately, specific inhibitors for this
quences (Fig. 2.3): mechanism are not yet available for research
or therapy.
1. Intracellular acidosis is rapidly reduced.
Intracellular acidosis inhibits, however, Role of Rapid Normalization
the myofibrillar machinery, i.e., it ex- of Tissue Osmolality
erts an effect similar to the presence of One of the major causes for water influx
BDM during the early phase of into the ischemic-reperfused myocardial cell
reperfusion.25 Rapid extrusion of excess seems to be cytosolic Na + overload. The
H+ from the reoxygenated cell thus re- Na+/H+ exchanger plays a major role in cell
moves a potentially protective agent. volume regulation.29,30 In ischemic myocar-
2. Activation of the Na+/H+ exchanger dium the end products of anaerobic metabo-
causes a net influx of Na+ into the cy- lism also accumulate, thus increasing the
tosol. Depending on the ability of the osmotic load in the intracellular and the in-
Na+ pump to remove this excess load terstitial space.30 If during reperfusion the
of Na+, it may come to a secondary ac- extracellular excess of osmotically active mol-
tivation of the Na +/Ca 2+ exchange ecules is rapidly washed out, an osmotic gra-
mechanism, transporting Na+ in the dient between the intracellular and the extra-
outward direction and Ca2+ in the in- cellular space is generated.31 Cellular uptake
ward direction. This coupled mecha- of water and, through the consecutive increase
nism may enhance the preexisting Ca2+ in intracellular pressure, mechanical stretch of
overload of the cells. the sarcolemma meets a myocardial cell whose
Rapid H+ removal and secondary Ca2+ mechanical fragility was increased during the
uptake thus both favor the development of preceding energy depletion.32-34 As is the case
hypercontracture if ischemic-reperfused myo- for hypercontracture, swelling per se is
The Cellular Basis of Immediate Lethal Reperfusion Injury 35

Fig. 2.3. Scheme of H+ control and its consequences in the cardiomyocyte upon reperfusion. Ischemia causes intra-
and extracellular acidification. Intracellular acidosis inhibits contractile activation. Upon reperfusion the extracel-
lular H+ concentration is rapidly reduced and thus a transsarcolemmal H+ gradient created. Consecutively, intra-
cellular excess H+ is removed via the Na+/H+ exchanger (1) and the Na+/HCO3- symporter (2). As long as high
intracellular H+ concentration is present, the contractile machinery is impaired and hypercontracture prevented.
Na+ influx through (1) and (2) can cause net Ca2+ influx through the Na+/Ca2+ exchanger (3) in its “reverse mode”.
This increases cytosolic Ca2+ overload, favoring uncontrolled contractile activation.

normally not able to disrupt the sarcolemma, dicate that attenuation of the additional me-
as demonstrated by the maintenance of sar- chanical stress imposed by swelling can limit
colemmal integrity in isolated cardiomyocytes myocardial necrosis during reperfusion.36-38
subjected to osmotic stress in normoxic con- The mechanism of sarcolemmal fragility
ditions. However, the mechanical stress caused secondary to energy deprivation is not under-
by swelling may add up with other sources of stood in detail. Alterations in the lipidic com-
stress and then result in cell deterioration position of the cell membrane, as suggested
(Fig. 2.4). In isolated cardiomyocytes, osmotic by the protective effect of treatments preserv-
stress results in sarcolemmal disruption only ing the turnover of phospholipids during
if the cell develops hypercontracture and has energy deprivation,34 changes in sarcolemmal
previously been submitted to prolonged energy proteins39,40 or changes in the sarcolemma-
deprivation. 35 The combination of these cytoskeleton anchorage41,42 could play a role.
factors seems to increase sarcolemmal fragility There is also evidence that sarcolemmal fra-
and render the cell thus more susceptible to gility induced by ischemia can be aggravated
damage by osmotic stress. The results of stud- during the first moments of reperfusion.33,40
ies with highly hyperosmotic reperfusion in- It has recently been demonstrated that
36 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 2.4. Scheme of osmotic injury of the reoxygenated cardiomyocyte. Upon reperfusion a transsarcolemmal
osmotic gradient is created. This leads to water influx and cell swelling. Cytoskeletal fragility increases during
ischemia. Circumstances of ischemia and reperfusion augment independently sarcolemmal fragility. Increased
cytoskeletal and sarcolemmal fragility and cell swelling together favor rupture of the sarcolemma.

enhanced susceptibility of reoxygenated myo- enced by cell-to-cell interactions.43,44 Histo-


cardial cells to osmotic injury can be reduced logic observations have shown that the areas
by specific interventions during the early phase of contraction-band necrosis induced by
of reoxygenation.33 Effective measures were transient coronary occlusion followed by
additions of NO-donors in high concentra- reperfusion are composed of hypercontracted
tion and of an antilipid peroxidant or means myocytes connected to each other to form a
increasing the cellular glutathione pool. The continuum, of which the often complex
results suggest that mechanical fragility of the geometry cannot be explained by gradients of
sarcolemma is increased by radical mechanisms flow or microvascular distribution.45 Com-
during the early period of reoxygenation. It puter simulation studies indicate that some
must be said clearly that, in this role of kind of cell-to-cell interaction must be taken
enhancing sarcolemmal fragility, oxygen radi- into account to explain these histological fea-
cals are a factor of secondary importance for tures, and that in the absence of such interac-
reperfusion injury, when seen in relation to tion, hypercontracted myocytes should be scat-
the whole scenario. tered across the area of risk instead of forming
continuous zones of necrosis. It has been sug-
Following Initial Causes: gested that this cell-to-cell interaction could
Spreading of Necrosis be mechanical, the exchange of forces imposed
by tight intercellular junctions tearing apart
Several lines of evidence indicate that
the sarcolemma of myocytes hypercontracting
reoxygenation-induced hypercontracture and
during in situ reperfusion, and damaging the
sarcolemmal disruption are markedly influ-
The Cellular Basis of Immediate Lethal Reperfusion Injury 37

sarcolemma of adjacent cells.43,44 But the ally controlled cellular response to moderate
interaction between adjacent myocytes lead- cell injury or to the influence of various
ing to cell-to-cell progression of hyper- cytokines. In contrast, necrotic cell death is
contracture also could be chemical. Ca2+ and the consequence of severe structural cell dam-
other second messengers may diffuse freely age and is not transcriptionally regulated. Cells
through gap junctions and thereby transmit which have entered the apoptotic process
the trigger for hypercontracture. Recent stud- retain physical integrity of the plasmalemma
ies in pairs of isolated cardiomyocytes have initially even though its chemical structure
demonstrated that hypercontracture of one cell may change. The point where the process
induced by sarcolemmal disruption or becomes irreversible seems to be the activa-
microinjection of Ca2+ can induce hyper- tion of endonucleases severing genomic DNA
contracture of adjacent cells, that this trans- at internucleosomal sites, what is then taken
mission is associated to passage of gap junc- as a characteristic feature of cell death by
tion permanent dye to the adjacent cell, and apoptosis. In a number of recent papers evi-
that the gap junction uncoupler heptanol pre- dence for apoptotic cell injury in ischemic-
vents both dye passage and transmission of reperfused myocardium and in border zones
hypercontracture. The intracoronary admin- of ischemic myocardium has been demon-
istration of heptanol during the first minutes strated.47-53 This gives rise to the question if
of reperfusion significantly reduces infarct size reperfusion of severely ischemic myocardium
in the in situ pig heart submitted to transient can be followed by delayed apoptotic cell death
coronary occlusion.46 These results are con- which could abolish all short-lived protective
sistent with the hypothesis that cell-to-cell effects against the acute onset of necrosis dur-
transmission of hypercontracture may cause ing reperfusion. The real contribution of
spreading of necrosis, and thus contributes to apoptosis to cardiomyocyte death has not yet
reperfusion injury. been established. To date, the factors initiat-
ing apoptosis in ischemic-reperfused myocar-
The Problem of Delay dium are still unclear. It is also an open ques-
of Necrosis and Initiation tion whether the apoptotic process observed
in reperfused myocardium is due to triggers
of Apoptosis during the time of ischemia or during
Some interventions applied at the time of reperfusion. If the latter is the case, it seems
reperfusion can apparently reduce the extent reasonable to expect that possibilities are found
of irreversible tissue injury when this is inves- to inhibit the full development of apoptotic
tigated early but in fact may only delay the cell death as the whole process of apoptosis is
manifestation of irreversible injury. If this is a multi-stage metabolic mechanism with many
the case such an intervention does not provide possible sites of inhibition.
true reperfusion protection. There are indeed
some cases known where an apparent protec-
tive effect was later found to be only imitated
Conclusions
After prolonged periods of energy deple-
by a time delay in development of the mark-
tion the ischemic myocardial cell can be jeop-
ers of necrotic tissue injury. This must be dis-
ardized by specific causes within the reper-
tinguished from injury of myocardium by
fusion period (Fig. 2.5). These causes can be
causes appearing not during the early but
viewed as unwanted aspects of the recovery
during the late phase of reperfusion, e.g., tis-
process itself limiting its efficiency. Under-
sue injury by invasion of activated neutrophils.
standing of the basic causes has opened novel
It is another question whether reperfused perspectives for specific interference with these
myocardium may also become subject to serious pathomechanisms. The experimental
apoptosis, i.e., programmed cell death, even results encourage the development of thera-
if effectively protected against immediate peutic approaches to reduce infarct size by
necrotic injury. Apoptosis is a transcription- specific measures applied during the early
38 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 2.5. Scheme of factors contributing to immediate lethal (necrotic) reperfusion injury of the cardiomyocyte.

phase of reperfusion. The principles of the strategy has hindered research on this impor-
protective interventions during the early stage tant pathophysiological problem during recent
of reperfusion are: years. Knowledge on the basic causal mecha-
1. Inhibition of contractile activation; nisms of reperfusion injury, reviewed in this
2. prevention of intracellular Ca2+ oscillations; chapter, does no longer justify abstention from
3. preservation of intracellular acidosis; intensive research on new principles of
4. suppression of causes favoring sarcolemmal reperfusion protection. Cardiac surgery may
fragility; profit immediately from this research since in
5. reduction of cell swelling; most cardiac surgery procedures the reper-
6. prevention of spreading of necrosis. fusion conditions can be modified at will.
This does not exclude that measures aimed
to limit necrosis or apoptosis occurring later Acknowledgement
during reperfusion are of additional value. Supported by the BIOMED II Programme
To date, all these strategies for reperfusion of the European Union, grants PL 95-1254
protection have only been studied experimen- and PL 95-0838.
tally and the number of studies is relatively
small. The results of these studies conclusively
demonstrated that it is possible to markedly References
reduce myocardial necrosis by treatments 1. Yellon DM, Jennings RB, eds. Myocardial
applied at the time of reperfusion, and pro- Protection. The Pathophysiology of Reper-
vide precise mechanistic explanation for this fusion and Reperfusion injury. New York:
Raven, 1992.
beneficial effect within the frame of our cur- 2. Przyklenk K, ed. Special Issue: Lethal Myo-
rent understanding of myocyte death during cardial Reperfusion injury. J Thromb Throm-
ischemia-reperfusion. This is in contrast to the bolysis 1997; 4:1-154.
wealth of investigations testing the strategy of 3. Ferrari R, Hearse DJ. Reperfusion injury:
Does it exist and does it have clinical rel-
preventing injury due to oxygen free radicals evance? J Thromb Thrombolysis 1997; 4:
in reperfusion injury. The failure of the latter 25-34.
The Cellular Basis of Immediate Lethal Reperfusion Injury 39

4. Kloner RA. Does reperfusion injury exist in 17. Habazettl H, Palmisano BW, Bosnjak ZJ et
humans? J Am Coll Cardiol 1993; 21: al. Initial reperfusion with 2,3 butanedione
537-545. monoxime is better than hyperkalemic reper-
5. Downey JM, Yellon DM. Do free radicals fusion after cardioplegic arrest in isolated
contribute to myocardial cell death dur- guinea pig hearts. Eur J Cardiothorac Surg
ing ischemia-reperfusion? In: Yellon CM, 1996; 10:897-904.
Jennings RB, eds. Myocardial Protection: The 18. Ladilov YV, Siegmund B, Piper HM. Simu-
Pathophysiology of Reperfusion and Reper- lated ischemia increases the susceptibility of
fusion injury. New York: Raven, 1992:35-57. rat cardiomyocytes to hypercontracture. Circ
6. Bolli R. Myocardial stunning in man. Circu- Res 1997; 80:69-75.
lation 1992; 86:1671-1691. 19. Gao WD, Liu Y, Mellgren R et al. Intrinsic
7. Hearse DJ, Humphrey SM, Chain EB. myofilament alterations underlying the de-
Abrupt reoxygenation of the anoxic potas- creased contractility of stunned myocardium.
sium-arrested perfused rat heart: A study of A consequence of Ca2+-dependent proteoly-
myocardial enzyme release. J Mol Cell Cardiol sis? Circ Res 1996; 78:455-465.
1973; 5:395-407. 20. Siegmund B, Schlack W, Piper HM. Hal-
8. Ganote CE. Contraction band necrosis and othane protects cardiomyocytes against reoxy-
irreversible myocardial injury. J Mol Cell genation-induced hypercontracture. Circula-
Cardiol 1983; 15:67-73. tion, in press.
9. Ganote CE, Vander Heide RS. Importance 21. Schlack W, Hollmann M, Stunneck J et al.
of mechanical factors in ischemic and reper- Effect of halothane on myocardial reoxy-
fusion injury. In: Piper HM, ed. Pathophysi- genation injury in the isolated rat heart. Br J
ology of severe ischemic myocardial injury. Anaesth 1996; 76:860-867.
Dordrecht: Kluwer, 1990:337-355. 22. Preckel B, Schlack W, Comfère T et al. Ef-
10. Siegmund B, Zude R, Piper HM. Recovery fects of inhalational anaesthetics on myocar-
of anoxic-reoxygenated cardiomyocytes from dial reperfusion injury in vivo. Pflügers Arch
severe Ca2+ overload. Am J Physiol 1992; 1997, suppl to 433:R15.
263:H1262-H1269. 23. Lagadic-Gossmann D, Vaughan-Jones RD.
11. Siegmund B, Ladilov YV, Piper HM. Impor- Coupling of dual acid extrusion in the guinea-
tance of Na+ for the recovery of Ca2+ control pig isolated ventricular myocytes to a1 and
in reoxygenated cardiomyocytes. Am J Physiol b-adrenoceptor. J Physiol 1993; 464:49-73.
1994; 267:H506-H513. 24. Piper HM, Balser C, Ladilov YV et al. The
12. Siegmund B, Koop A, Klietz T et al. Sar- role of Na+/H+ exchange in ischemia-reper-
colemmal integrity and metabolic compe- fusion. Basic Res Cardiol 1996; 91:191-202.
tence of cardiomyocytes under anoxia- 25. Ladilov YV, Siegmund B, Piper HM. Pro-
reoxygenation. Am J Physiol 1990; 258: tection of the reoxygenated cardiomyocyte
H285-H291. against hypercontracture by inhibition of
13. Siegmund B, Klietz T, Schwartz P et al. Na+/H+ exchange. Am J Physiol 1995; 268:
Temporary contractile blockade prevents H1531-H1539.
hypercontracture in anoxic-reoxygenated 26. Bugge E, Ytrehus K. Inhibition of sodium-
cardiomyocytes. Am J Physiol 1991; 260: hydrogen exchange reduces infarct size in the
H426-H435. isolated rat heart—a protective additive to
14. Schlüter KD, Schwartz P, Siegmund B et al. ischemic preconditioning. Cardiovasc Res
Prevention of the oxygen paradox in hypoxic- 1995; 29:269-274.
reoxygenated hearts. Am J Physiol 1991; 27. Klein HH, Pich S, Bohle RM et al. Myocar-
261:H416-H432. dial protection by Na+/H+ exchange inhibi-
15. García-Dorado D, Théroux P, Duran JM et tion in ischemic reperfused porcine hearts.
al. Selective inhibition of the contractile ap- Circulation 1995; 92:912-917.
paratus: A new approach to modification of 28. García-Dorado D, González MA, Barrabés JA
infarct size, infarct composition, and infarct et al. Prevention of ischemic rigor contrac-
geometry during coronary artery occlusion ture during coronary occlusion by inhibition
and reperfusion. Circulation 1992; 85: of Na+-H+ exchange. Cardiovasc Res 1997;
1160-1174. 96:3579-3586.
16. Schlack W, Uebing A, Schäfer M et al. Re- 29. Inserte J, García-Dorado D, Ruiz-Meana M
gional contractile blockade at the onset of et al. The Na+-H+ exchange occurring dur-
reperfusion reduces infarct size in the dog ing hypoxia in the genesis of reoxygenation-
heart. Pflügers Arch 1994; 428:134-141. induced myocardial oedema. J Mol Cell
Cardiol 1997; 29:1167-1175.
40 Ischemia-Reperfusion Injury in Cardiac Surgery

30. Grinstein S, Woodside M, Sardet C et al. 43. Ganote CE. Cell-to-cell interactions contrib-
Activation of the Na+/H+ antiporter during uting to the oxygen paradox. Bas Res Cardiol
cell volume regulation. Evidence for a phos- 1985; 80:141-146.
phorylation-independent mechanism. J Biol 44. Frank JS, Brandy AJ, Fransworth BS et al.
Chem 1992; 267:23828-23828. Ultrastructure and function of isolated
31. García-Dorado D, Oliveras J. Myocardial myocytes after calcium depletion and reple-
edema: A preventable cause of reperfusion tion. Am J Physiol 1986; 250:H265-H275.
injury. Cardiovasc Res 1993; 27:1555-1563. 45. Solares J, García-Dorado D, Oliveras J et al.
32. Armstrong SC, Ganote CE. Effects of the Contraction band necrosis at the lateral bor-
protein phosphatase inhibitors okadaic acid ders of the area at risk in reperfused infarcts.
and calyculin A on metabolically inhibited Observations in a pig model of in situ coro-
and ischemic isolated myocytes. J Mol Cell nary occlusion. Virchows Arch 1995; 426:
Cardiol 1992; 24:869-884. 393-399.
33. Schlüter KD, Jakob G, Ruiz-Meana M et al. 46. García-Dorado D, Inserte J, Ruiz-Meana M
Protection of reoxygenated cardiomyocytes et al. Gap junction uncoupler heptanol pre-
against osmotic fragility by NO donors. Am vents cell-to-cell progression of hyper-
J Physiol 1996; 271:H428-H434. contracture and limits necrosis during myo-
34. Ruiz-Meana M, García-Dorado D, Julia M. cardial reperfusion. Circulation 1997; 35:
González MA et al. Pretreatment with 80-89.
trimetazidine increases sarcolemmal me- 47. Bardales RH, Hailey LS, Xie SS et al. In situ
chanical resistance in reoxygenated myocytes. apoptosis assay for the detection of early acute
Cardiovasc Res 1996; 32:587-592. myocardial infarction. Am J Pathol 1996;
35. Ruiz-Meana M, García-Dorado D, González 149:821-829.
MA et al. Effect of osmotic stress on sar- 48. Fliss H, Gattinger D. Apoptosis in ischemic
colemmal integrity of isolated cardiomyocytes and reperfused rat myocardium. Circ Res
following transient metabolic inhibition. 1996; 79:949-956.
Cardiovasc Res 1995; 30:64-69. 49. Suzuki H, Wildhirt SM, Dudek RR et al.
36. Kloner RA, Reimer KA, Willerson JT et al. Induction of apoptosis in myocardial infarc-
Reduction of experimental myocardial infarct tion and its possible relationship to nitric
size with hyperosmotic mannitol. Proc Soc oxide synthase in macrophages. Tissue Cell
Exp Biol Med 1976; 151:677-683. 1996; 28:89-97.
37. Tranum-Jensen J, Janse MJ, Fiolet WT et al. 50. Veinot JP, Gattinger DA, Fliss H. Early
Tissue osmolality, cell swelling, and reper- apoptosis in human myocardial infarcts. Hum
fusion in acute regional myocardial ischemia Pathol 1997; 28:485-492.
in the isolated porcine heart. Circ Res 1981; 51. Saraste A, Pulkki K, Kallajoki M et al.
49:364-381. Apoptosis in human acute myocardial infarc-
38. García-Dorado D, Théroux P, Muñoz R et tion. Circulation 1997; 95:320-323.
al. Favorable effects of hyperosmotic reper- 52. Kajstura J, Cheng W, Reiss K et al. Apoptotic
fusion on myocardial edema and infarct size. and necrotic myocytes cell deaths are inde-
Am J Physiol 1992; 262:H17-H22. pendent contributing variables of infarct size
39. Iizuka K, Kawaguchi H, Kitabatake A. Ef- in rats. Lab Invest 1996; 74:86-107.
fects of thiol protease inhibitors on fodrin 53. Gottlieb RA, Burleson KO, Kloner RA et al.
degradation during hypoxia in cultured Reperfusion injury induces apoptosis in rab-
myocytes. J Mol Cell Cardiol 1993; 25: bit cardiomyocytes. J Clin Invest 1994;
101-1109. 94:1621-1628.
40. Yoshida K, Inui M, Saido TC et al. Reper-
fusion of rat heart after brief ischemia induced
proteolysis of calspectin (nonerythroid spec-
trin of fodrin) by calpain. Circ Res 1995;
77:603-610.
41. Steenbergen C, Hill ML, Jennings RB.
Cytoskeletal damage during myocardial is-
chemia: Changes in vinculin immunofluores-
cence staining during total in vitro ischemia
in canine heart. Circ Res 1987; 60:478-486.
42. Ganote CE, Vander Heide RS. Cytoskeletal
lesions in anoxic myocardial injury: A con-
ventional and high voltage electron micro-
scopic and immunofluorescence study. Am J
Pathol 1987; 129:327-344.
CHAPTER 3

Apoptosis in Ischemia—
Reperfusion Injury
Harald Darius, Waltraud Ibe and Michael Buerke

A
poptosis or “programmed cell death” therapeutic goal in the future to aim at is still
has recently been recognized to be unresolved.
involved in several cardiovascular
diseases. There are some histologic criteria as Characteristics of Apoptosis
well as histochemical, biochemical and gel elec- Apoptosis and necrosis are two indepen-
trophoretic methods developed for the detec- dent phenomena determining the survival of
tion of apoptosis in isolated cells, tissues or cells within an organ structure or isolated cells
organs. In myocarditis and heart failure evi- in response to environmental factors. Since
dence is accumulating that apoptosis may play necrosis is more like an accident of nature due
a role in their pathophysiology. In isolated to malnutrition of a cell or tissue damage
cardiomyocytes hypoxia and reoxygenation caused by ischemia or toxic concentrations of
cause apoptosis, cytokines at physiologic con- environmental toxins or biological mediators
centrations induce apoptosis and intracellular e.g., oxygen derived radicals, apoptosis repre-
promotors and inhibitors of apoptosis have sents a genetically determined process. Dur-
been identified. In animal models of myocar- ing necrosis the membrane permeability is
dial ischemia and reperfusion apoptotic cells increased, swelling of cells and organelles
have been detected in the area at risk not results from altered ion pump activity leading
undergoing necrosis. Reperfusion itself either to increased sodium retention. DNA degra-
seems to induce apoptosis or at least signifi- dation occurs nonspecifically resulting in frag-
cantly augments this process, depending on ments of nondefined length showing in gel
the experimental conditions and animal spe- electrophoresis as a smear. Cell swelling and
cies studied. In autopsy studies of human myo- nuclear destruction result in cytolysis, release
cardial infarction, victims apoptotic cells were of cytoplasmic constituents and induction of
identified in the border zone between success- an inflammatory reaction.
fully reperfused myocardium and the core
ischemic area undergoing necrosis. Intracel- In contrast to necrosis, apoptosis or pro-
lular regulatory proteins (e.g., Bcl-2, Bax, Fas, grammed cell death (PCD) represents a normal
p53) were identified to be involved in feature of cell and tissue development (Table
apoptosis induction or reduced expression. 3.1). This is of specific relevance in the fetus
The pathophysiologic role of apoptosis in and in some instances of cell replacement in
human myocardial infarction and reperfusion adult tissues (e.g., thymus). PCD is a tightly
as well as in surgical ischemia and reperfusion regulated and energy dependent process
still has to be determined. If the reduction or induced by a genetic program and requiring
induction of apoptosis may be an useful gene transcription. Cells undergoing necrosis

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


42 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 3.1. Specific features of apoptosis in contrast to necrosis

Necrosis Apoptosis

- Death of tissue segments - Death limited to single cells


- Membrane destruction - Membrane remains intact
- Cell swelling - Cell shrinkage
- Nonspecific DNA degradation - Chromatin condensation
- Inflammatory response - Phagocytosis without inflammation
- No energy required - Energy required
- Nonregulated event - Highly regulated event
- No gene requirement - Gene transcription (de novo)

can be easily differentiated from those fragments (Fig. 3.1A). Light microscopy, fluo-
undergoing PCD since PCD results in cell rescence microscopy or flow cytometry
shrinkage, blebbing of the cellular membrane (Fig. 3.1B) may be used after labeling the
and condensation of the nuclear chromatin. DNA fragments using a TUNEL assay
Since apoptotic bodies are formed no or very (Terminal deoxynucleotidyl transferase-medi-
little cytoplasmic constituents are released into ated dUTP Nick End-Labeling) or Annexin
the surrounding environment and, unlike in V antibodies to detect the terminal DNA frag-
necrosis, there is no evidence of a localized in- ments. The most reliable but laborious method
flammatory reaction. The apoptotic bodies are is the detection of the characteristic cellular
phagocytosed instead. Nuclear condensation features of apoptosis described above by elec-
is followed by nuclear disintegration and DNA tron microscopy. An alternative method for
fragmentation into segments of 180-200 bp the detection of apoptosis in a tissue segment
length or multiples of this. These fragments is the visualization of DNA strand breaks of a
can become visualized easily in gel electro- rather specific segment length between 180 and
phoresis, where a laddering phenomenon is 200 base pairs in agarose gel electrophoresis by
seen with characteristic bands of the respec- the characteristic laddering phenomenon.
tive segment length.
There is a wide variety of environmental Apoptosis in Cardiac Disease
factors and biological mediators that have been Programmed cell death or apoptosis re-
demonstrated to cause PCD in vitro in cell cently has been demonstrated in several car-
culture or isolated tissues (Table 3.2). In vivo diac disease states. Mallat et al1 demonstrated
however, a number of noncardiovascular and convincing evidence of apoptosis being heavily
cardiovascular disorders have been identified involved in the degenerative process of the
to result in an accelerated rate of apoptosis arrhythmogenic right ventricular dysplasia,
(Table 3.3). which often is not differentiated from Uhl’s
disease. In end stage heart failure all samples
Detection of Apoptosis derived from hearts of patients with dilated
There are a number of methods for the cardiomyopathy showed histologic evidence of
detection of apoptosis in isolated cells or tis- apoptosis and a positive laddering phenom-
sues, however none of which is regarded as enon in gel electrophoresis. In contrast, only
being entirely specific (Table 3.4). Thus, very one out of three hearts from patients with end
often two or more independent methods are stage coronary artery disease had detectable
utilized for the confirmation of apoptotic cells. DNA fragmentation as a sign of apoptosis.2
The methods are either based on histologic or When hearts from congestive cardiac failure
cytologic examination or on detection of DNA patients were analyzed and compared to hearts
Apoptosis in Ischemia: Reperfusion Injury 43

Table 3.2. Environmental factors and biologic mediators for which an increased rate
of apoptosis has been demonstrated

Physiologic stimuli Nonphysiologic stimuli

- steroids - heat,
- calcium - UV-irradiation
- tumor necrosis factor - infections
- transforming growth factor - cytotoxic T-cells
- depletion of essential growth factors - oncogenes, tumor-suppressor gene
- loss of cell-cell contact - chemotherapeutics

Table 3.3. Altered rates of apoptosis observed in noncardiovascular and in


cardiovascular disorders

Noncardiovascular disorders
Increased rate Diminished rate

- AIDS - malignancies
- IDDM I* - autoimmune disease
- myelodysplastic syndrome - viral infection
- ischemic lesions
- neurodegenerate disorders
*: IDDM-I: insulin dependent diabetes mellitus Type I
Cardiovascular disorders
Increased rate of apoptosis

- myocardial infarction
- reperfusion damage
- arterial hypertension
- arrhythmogenic right ventricular dysplasia
- dilated cardiomyopathy
- atherosclerotic plaque
- restenosis after ballooning

from patients who died due to motor accidents Experimental and Clinical
the morphologic features of apoptosis were Evidence of Apoptosis in Acute
increased 232-fold. In confocal microscopy
DNA-strand breaks were associated with chro- Myocardial Infarction
matin condensation and fragmentation.3 and Reperfusion
Other groups presented evidence of apoptosis During the last few years increasing evidence
in myocarditis, left ventricular hypertrophy became available from animal experiments and
and coronary artery dilatation. Thus, apoptosis from human autopsy studies showing a role
may play a major role in cardiomyocyte cell of apoptosis in myocardial ischemia with or
death and may help to explain some of the without reperfusion. In isolated cardiac
uniform reactions of the heart towards a wide myocytes the hypoxia-induced decrease in
variety of damaging agents or environmental intracellular glutathione seems to be responsible
factors. for the reoxygenation-dependent c-jun kinase
44 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 3.4. Methods for the detection of apoptosis in isolated cells or tissues

Morphologic methods:

- Electron microscopy
- TUNEL-Assay using peroxidase, alkaline phosphatase, or fluorescein labeling
- Detection of DNA fragments in flow cytometry
Chemical methods detecting nuclear strand breaks:

- ELISA for histone detection (cell death detection assay)


- Laddering phenomenon in gel electrophoresis
- Binding of labeled Annexin V to phosphatidylserine in flow cytometry
Detection of regulatory proteins (e.g., Bcl-2, Bax, Fas) by antibody-based detection in cells (light
microscopy) or tissue homogenates (ELISA) or Western blot

activation, which is selectively inhibited by condensation was markedly different between


tyrosine kinase inhibitors or N-acetyl cysteine.4 reperfused and permanently ischemic myocar-
A number of extracellular mediators and in- dium. Interestingly, induction of granulocy-
tracellular signaling proteins have been iden- topenia in the rabbits did not alter the
tified as contributing factors to cardiomyocyte development of apoptosis, an effect very dif-
apoptosis. Physiologic concentrations of TNF- ferent from necrosis, which is highly depen-
alpha induced apoptosis in isolated rat dent on neutrophil infiltration.9 In contrast
cardiomyocytes probably causing apoptosis by to the experiments performed in rabbit myo-
activating the endogenous mediator cardial ischemia, rat myocardium seems to be
sphingosine.5 Other authors suggested the more susceptible to ischemia for the induc-
involvement of intracellular signaling proteins tion of apoptosis. In a model of permanent
like p53,6 Fas7 and others.8 However, these coronary artery occlusion, Kajstura et al10 ana-
data were obtained in vitro using isolated rat lyzed infarct size and rate of apoptotic cells
cardiomyocytes in most studies. Thus, the after 20 min-7 days. They report that apoptosis
physiologic relevance of these findings for the is the most prominent mechanism of cell death
pathophysiologic sequealae of events in human in the majority of cardiomyocytes within the
myocardial ischemia and reperfusion is still first 2 days of infarction. This corresponds to
unclear. In addition to the in vitro experiments the immunocytochemically detected increase
there are a number of reports from animal in Bcl-2 and Fas expression, which are
experiments in vivo concerned with the increased 18- and 131-fold.10 The results
detection of apoptosis during myocardial reported by Fliss and Garringer11 correspond
ischemia and/or reperfusion. It is not entirely quite well with the data referred to above, since
clear yet if ischemia alone is a sufficiently Fliss and Garringer were able to show that
strong stimulus to induce apoptosis. Gottlieb development of apoptosis is present during the
et al9 were not able to detect signs of apoptosis first hours of permanent coronary artery liga-
in rabbit myocardial tissue undergoing per- tion in the rat; however reperfusion markedly
manent ischemia for 4.5 hours. In contrast, if accelerates and augments this process.
the myocardium was reperfused for 4 hours Although the conditions in autopsy studies are
following a period of 30 min of ischemia a far from being as standardized as compared to
substantial number of cardiomyocytes under- animal or in vitro experiments, there is grow-
went apoptosis, detected either by gel electro- ing evidence that apoptosis is a phenomenon
phoresis, by in situ nick end labeling or elec- also present in acute human myocardial inf-
tron microscopy. In transmission electron arction. Itoh et al12 were able to identify
microscopy the pattern of nuclear chromatin apoptotic cells by nick end labeling and DNA
Apoptosis in Ischemia: Reperfusion Injury 45

Fig. 3.1A.) Nuclear staining in rabbit vascular smooth muscle cells following incubation of the cells with actino-
mycin D. Nuclei show chromatin condensation and the formation of apoptotic bodies. B.) Flow cytometric DNA
analysis of rabbit vascular smooth muscle cells. On the left panels cells under control conditions exert their DNA
in haploid or diploid manner, with very few cells being in the S-phase (peak to the right). Following incubation
with very high concentrations of nitroprusside sodium (NPN, 10 mM) cells undergo apoptosis and the number
of haploid and diploid cells decreases markedly, with many cell fragments exerting DNA strands of varying length
(peak to the left).

agarose gel electrophoresis in hearts of patients human myocardium are already visible after
who died during acute myocardial infarctions. about two hours of myocardial ischemia, thus
In their histologic examinations they observed possibly representing an early histologic sign
most apoptotic cells in areas staining strongly of acute myocardial infarction. Saraste et al14
with hematoxylin-eosin, commonly accepted analyzed human autopsy specimens and found
as an indicator of necrosis. Other groups13 sug- that apoptotic cells were present primarily in
gested, that the early signs of apoptosis in the border zone of histologically infarcted
46 Ischemia-Reperfusion Injury in Cardiac Surgery

myocardium. Only very few apoptotic cells detection of digoxigenin labeled genomic
were detectable in areas remote to the infarct DNA by the TUNEL reaction using 3'-OH
zone. The authors speculate that apoptosis may end labeling with terminal deoxynucleotidyl
provide a possible target for therapeutic transferase.
nterventions during evolving myocardial inf- In nontreated rats ischemia and reperfusion
arction in humans. Some of the intracellular resulted in a significant loss of creatine kinase
signaling proteins possibly involved in human activity from the ischemic area in the range of
cardiomyocyte cell death during acute myo- 570 IU/100 mg protein. This CK loss was
cardial infarction were identified by Misao et almost completely prevented if the rats were
al.15 They found a very high incidence of Bcl-2 pretreated with an intraperitoneal injection of
positive cells, an inhibitor of apoptosis, in areas insulin-like growth factor (10 ∝g) one hour
salvaged by thrombolysis immediately adjacent prior to coronary artery ligation. A compa-
to infarcted tissue. In contrast, in old myocar- rable but somewhat less pronounced cardio-
dial infarctions they found almost no Bcl-2 protective effect was seen dose-dependently
positive cells. However, immunoreactivity for with injection of up to 100 ∝g/kg bw of
Bax, an accelerator of apoptosis, showed a very C1-esterase inhibitor 2 min prior to initiation
high incidence in old infarctions, whereas only of reperfusion, preventing the activation of the
2 of 15 hearts with acute MI showed Bax posi- classical complement pathway (Fig. 3.2). Simi-
tive specimens. lar cardioprotective effects of C1-esterase
inhibitor have been described previously in a
Prevention of Apoptosis During cat and a porcine model of myocardial
Ischemia and Reperfusion ischemia and reperfusion.17-19 Interestingly,
In a rat model of acute myocardial ischemia both cardioprotective agents studied very
and reperfusion, M. Buerke was able to dem- effectively prevented neutrophil infiltration
onstrate in two different sets of experiments into the ischemic area as evidenced by a sig-
performed in Allan M. Lefers group and in nificantly reduced myeloperoxidase content,
our group the prevention of apoptosis by phar- which was confirmed by histologic analysis.
macologic interventions given immediately In both studies the prevention of neutrophil
prior to initiation of reperfusion.16,17 In ether infiltration was thought to be intimately
anesthetized rats the heart was exteriorized involved in the reduction of reperfusion dam-
briefly and a silk slip knot was placed around age or were even thought to represent the
the left coronary artery. The heart was returned mechanisms of action (Fig. 3.2).
immediately to the chest cavity and the lungs In both sets of experiments histologic evi-
excavated in order to allow the animals to dence of apoptosis was obtained in the
breathe spontaneously. Following 20 min of ischemic areas of vehicle treated rats and a high
ischemia the slip knot was removed to initiate number of TUNEL positive i.e., cells under-
reperfusion for 24 hours. Infarct size was quan- going apoptosis was identified. In the IGF-1
titated by measuring the creatine kinase (CK) experiments sham operated rats had only 4%
loss within the left ventricular free wall of apoptotic nuclei identified, whereas rats
(ischemic region) if compared to the CK con- undergoing myocardial ischemia and reper-
tent in the septum i.e., nonischemic region. fusion exerted up to 62% positive cells. The
Leukocyte infiltration was measured by number of TUNEL positive nuclei was sig-
myeloperoxide content or visual analysis of nificantly reduced by treating the rats prior to
histologic slides stained with hematoxylin- reperfusion with 1 ∝g of IGF-1, which reduced
eosin. Histologic analysis was carried out in the apoptotic cells to about 28%.19 In a series
additional rats whose hearts were perfusion of experiments with inhibition of complement
fixed after 24 and 48 hrs of reperfusion. activation, the number of TUNEL positive
Immunohistochemical staining for apoptosis cells increased from 3% in sham operated ani-
was carried out by direct immunoperoxidase mals to about 33% in rats undergoing ischemia
and reperfusion. This substantial increase in
Apoptosis in Ischemia: Reperfusion Injury 47

Fig. 3.2. Upper panel: Infarct size in rats with myocardial ischemia for 20 min and reperfusion for 24 hours
measured as creatine kinase (CK) difference between the left ventricular free wall (ischemic area) and the interven-
tricular septum (nonischemic area). Rats were treated by intraperitoneal injection of insulin like growth factor
(IGF-1), 10 ∝g given 1 hr prior to coronary artery ligation or C1-esterase inhibitor (C1-INH) 100 ∝g/kg 2 min
prior to reperfusion (data taken from references 16 and 19). Lower panel: Neutrophil infiltration in the infarcted
myocardium quantified by measuring the activity of the neutrophil specific enzyme myeloperoxidase. Treatment
with IGF-1 or C1-INH significantly reduced the neutrophil infiltration into the area at risk, a process thought to
be the mechanism of action for the diminished reperfusion damage demonstrated by the reduced CK loss. (Data
taken from references 16 and 19)

Fig. 3.3. TUNEL reaction positive cardiomyocytes in the border zone of infarcted rat myocardium. Rats under-
went 20 min of ischemia and 24 hours of reperfusion. A: Apoptosisim vehicle treated animals; B: reduced apoptosis
in rats treated with C1-INH.
48 Ischemia-Reperfusion Injury in Cardiac Surgery

apoptotic cells was significantly reduced in C1- 10. Kajstura J, Cheng W, Reiss K et al. Apoptotic
esterase inhibitor treated animals to 13% and necrotic myocyte cell deaths are indepen-
dent contributing variables of infarct size in
(Fig. 3.3). rats. Lab Invest 1996; 74:86-107.
11. Fliss H, Garringer D. Apoptosis in ischemic
References and reperfused rat myocardium. Circ Res
1. Mallat Z, Tedgui A, Fontalioran F et al. Evi- 1996; 79:949-56.
dence of apoptosis in arrhythmogenic right 12. Itoh G, Tamura J, Suzuki M et al. DNA frag-
ventricular dysplasia. N Engl J Med 1996; mentation of human infarcted myocardial
335:1190-11962. cells demonstrated by the nick end labeling
method and DNA agarose gel electrophore-
2. Narula J, Haider N, Virmani R et al. Apop- sis. Am J Pathol 1995; 146:1325-31.
tosis in myocytes in end-stage heart failure. 13. Bardales RH, Hailey LS, Xie SS et al. In situ
N Engl J Med 1996; 335:1182-9. apoptosis assay for the detection of early acute
3. Olivetti G, Abbi R, Quaini F et al. Apoptosis myocardial infarction. Am J Pathol 1996;
in the failing human heart. N Engl J Med 149:821-9.
1997; 336:1131-41 14. Saraste A, Pulkki K, Kallajoki M et al.
4. Laderoute KR, Webster KA. Hypoxia/ Apoptosis in human acute myocardial infarc-
reoxygenation stimulates Jun kinase activ- tion. Circulation. 1997; 95:320-3.
ity through redox signaling in cardiac 15. Misao J, Hayakawa Y, Ohne M et al. Ex-
myocytes. Circ Res 1997; 80:336-3445. pression of bcl-2 protein, an inhibitor of
5. Krown KA, Page MT, Nguyen C et al. Tu- apoptosis, and Bax, an accelerator of apop-
mor necrosis factor alpha-induced apoptosis tosis, in ventricular myocytes of human hearts
in cardiac myocytes. Involvement of the sph- with myocardial infarction. Circulation 1996,
ingolipid signaling cascade in cardiac cell 94:1506-151216.
death. J Clin Invest 1996; 98:2854-65. 16. Buerke M, Murohara T, Skurk C et al.
6. Long X, Boluyt MO, Hipolito ML et al. p53 Cardioprotective effect of insulin-like growth
and the hypoxia-induced apoptosis of cultured factor I in myocardial ischemia followed by
neonatal rat cardiac myocytes. J Clin Invest reperfusion. Proc Natl Acad Sci USA 1995;
1997; 99:2635-43. 92:8031-5.
7. Tanaka M, Ito H, Adachi S et al. Hypoxia 17. Buerke M, Murohara T, Lefer AM. Cardio-
induces apoptosis with enhanced expression protective effects of a C1 esterase inhibitor
of Fas antigen messenger RNA in cultured in myocardial ischemia and reperfusion. Cir-
neonatal rat cardiomyocytes. Circ Res 1994; culation 1995; 91:393-402.
75:426-33. 18. Horstick G, Heimann A, Gotze O et al.
8. Gottlieb RA, Gruol DL, Zhu JY et al. Pre- Intracoronary application of C1 esterase in-
conditioning in rabbit cardiomyocytes. Role hibitor improves cardiac function and reduces
of pH, vacuolar proton ATPase, and apop- myocardial necrosis in an experimental model
tosis. J Clin Invest 1996; 97:2391-2398. of ischemia and reperfusion. Circulation
9. Gottlieb RA, Burleson KO, Kloner RA et al. 1997; 95:701-8.
Reperfusion injury induces apoptosis in rab- 19. Buerke M, Prüfer D, Dahm M et al. Inhibi-
bit cardiomyocytes. J Clin Invest 1994; tion of the classical complement pathway re-
94:1621-810. duces reperfusion injury and induction of
apoptosis. Eur Heart J 1996; 17:597 (Abstr.).
CHAPTER 4

Changes in Myocardial Gene


Expression Following Ischemia
and Reperfusion
John W. C. Entwistle III and Andrew S. Wechsler

T
he heart has the remarkable ability to myocardial stunning, in which the injured
adapt to a wide variety of physiological myocytes exhibit temporary contractile dys-
and pathological conditions. Some function despite adequate substrate delivery.
examples of change in the structure and func- By definition, stunned myocardium will regain
tion of the heart include normal growth and full mechanical function given time. The
development, response to pressure overload, mechanism through which the stunned heart
volume overload, hypothyroidism and responds to such injury is largely unknown,
ischemic injury. While the external manifes- but it appears that the basis for the cellular
tations of these changes are very different, they response to IR injury is at the molecular level.
share several features at the subcellular level. While the initiating event in stunning may be
The single common denominator is an alter- transient, it may also be involved in the initia-
ation in expression of the myocellular proteins. tion of a complex and coordinated cascade of
However, the initiating signals and subsequent molecular changes.
subcellular changes are diverse. This creates the Several factors have been implicated in the
situation in which the morphological changes onset of myocardial stunning, including the
seen in the myocardium and the resultant loss of high-energy phosphates, oxidative stress
changes in function fall into one of several through the generation of oxygen free radi-
defined categories, such as hypertrophy or car- cals, and altered myocardial cytosolic calcium
diomyopathy. It is likely that most if not all of levels and myofibrillar calcium sensitivity (see
the myocardial responses to injury have a sig- reviews by Bolli1 and Kusuoka2). In addition,
nificant component based on changes in the sympathetic overstimulation occurs during
expression of genes encoding structural or ischemia through neuronal release of cat-
functional proteins. echolamines, which may cause direct myo-
Ischemia and reperfusion (IR) have pro- cellular damage, compounding the injurious
found effects on the function and structure of effects (see review by Schömig3). These fac-
the myocardium. At one extreme, prolonged tors may play a significant role in both the
ischemia results in cellular necrosis. Since the initiation of stunning and in the initiation of
adult myocyte is incapable of division, the the molecular changes responsible for the
surviving cells must hypertrophy to restore altered myocardial phenotype seen during
contractile function. However, when the recovery.
ischemic insult is less severe, the result is

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


50 Ischemia-Reperfusion Injury in Cardiac Surgery

Basics of Myocardial Gene myocardium as an adaptive response to exter-


Expression nal signals.
The phenotypic changes involved in myo-
Adaptive and maladaptive changes in the
cardial recovery from IR injury may take hours
heart are both geometric and the consequence
to days to be detectable when monitoring pro-
of altered protein content and/or configura-
tein levels because of the slow rate of change
tion. The protein composition of the myocyte
in steady-state protein concentration. Many
can be regulated at one or more sites.
models of IR injury, such as the isolated heart,
Pretranslational control mechanisms include
are not suited for long-term studies. Thus,
those that affect the rate at which specific
analysis of steady-state mRNA levels has
mRNAs are produced (transcriptional), or
become a popular method for detecting alter-
those that affect their processing, nucleocyto-
ations in gene expression early after physiologic
plasmic transport and stability (posttranscrip-
events. The major assumption, however, is that
tional). The selection of mRNAs to be syn-
protein levels will eventually mirror those of
thesized into protein and the rate at which this
their respective mRNAs. When studied in
occurs are types of translational control. Post-
models of acute injury, mRNA levels gener-
translational regulation involves the process-
ally predict the direction of change in the cor-
ing, transportation and stability of the pro-
responding protein, indicating that transcrip-
tein products. Control may be exerted at one
tional control mechanisms are important in
or more levels for each mRNA and protein
regulating the phenotype of the heart after
product. Some well-described examples of the
injury.
regulation of myocardial gene expression in-
Studies of altered gene expression after
clude those seen during growth and develop-
myocardial injury have utilized a variety of
ment,4-6 or associated with overload-induced
models, including cultured myocytes, in vitro
myocardial hypertrophy.7-12 In the fetal heart,
beating hearts, and in vivo preparations. While
myosin and !-actin are present as “fetal”
changes in gene expression seen in isolated
isoforms. Atrial natriuretic factor is produced
myocyte preparations are clearly the result of
by both the atria and ventricles in the fetus,
IR injury to the myocytes, this may not repre-
and the proteins of the fetal sarcoplasmic
sent changes in the intact heart. Fibroblasts
reticulum (SR) are downregulated.6,13 After
and endothelial cells are just a few of the other
birth and during development, there is a
cellular components of the heart capable of
gradual shift in the expressed MHC and actin
responding to IR injury. Indeed, both of these
isoforms to those typical of the adult heart. In
cell types are extremely active in regulating
addition, atrial natriuretic factor (ANF) pro-
their local environment and produce sub-
duction in the ventricles disappears, and the
stances that act in a paracrine manner to
expression of SR gene products increases. Both
influence the myocytes. Thus, while it may be
hormonal and mechanical stimuli govern these
scientifically interesting to isolate myocytes in
changes. Hypertrophy in adult tissue in
an attempt to localize changes in gene expres-
response to pressure overload, but not volume
sion, failure to account for interactions with
overload,14 mimics gene expression in the fetal
other cell types and alterations of the mechani-
heart. This includes a switch in the dominant
cal properties of the myocyte represent an
isoforms for myosin and !-actin, re-expression
important shortcoming of such studies.
of ANF by the ventricles, and decreased
In intact heart models of IR injury, both
expression of the sarcoplasmic and endoplas-
regional and global models of IR have been
mic reticulum calcium ATPase-2 (SERCA2)
employed. Regional models of ischemia and
and phospholamban (plb). Changes in
reperfusion generally treat the nonischemic
myocardial gene expression occur in response
border zones as normal control tissue. How-
to a variety of other stimuli, and each elicits a
ever, there is a distinct possibility that these
characteristic pattern of gene expression. This
areas are also affected by the IR injury as the
phenotypic change permits remodeling of the
mechanical stresses seen in the “control” areas
Changes in Myocardial Gene Expression Following Ischemia and Reperfusion 51

are altered during ischemia and reperfusion, proteins, including several kinases. Ca2+/
until the function of the ischemic area recov- calmodulin-dependent protein kinase (CAM
ers fully. In addition, paracrine or nervous kinase) phosphorylates the Ca 2+ /cAMP
stimulation may also occur in these “control” response element binding protein (CREB).
areas as a result of injury, further altering the The phosphorylated CREB then can bind to
milieu of the cells in the border zone. the Ca2+/cAMP response element (CRE) in
the promoter region of target genes, and
Mechanisms of Gene Changes influence the rate of transcription of those
in Stunning genes. Addition of the calcium ionophore
A23187 to cultured cardiac myocytes induces
Stunning, when severe, may persist for sev-
an increase in the steady state mRNA level for
eral days before recovery of contractile func-
the immediate early gene (IEG) c-fos.18 c-fos
tion occurs, and extends beyond the termina-
and the other IEGs are transcription factors.
tion of the initial ischemic stimulus. While the
Once expressed, these IEGs affect the rate of
cellular and molecular mechanisms that
transcription of numerous other genes through
underlie this delay remain unknown, this
binding to the TPA (12-O-tetradeconoyl-
adaptive process in response to IR may involve
phorbol-13-acetate) response element (TRE)
specific qualitative changes in cell phenotype
in the promoter region of its target genes. The
that are the consequence of changes in myo-
CRE and TRE are found in the promoter
cardial gene expression. As the phenotype of
region of many genes. Thus, the effects of cal-
the heart is altered, these changes could alter
cium on gene expression, both directly through
the function and/or cellular morphology of the
the CRE, and indirectly through specific IEGs
stunned heart, and hence prolong recovery.
and TRE, may be widespread.
Two putative causes of stunning are increased
cytosolic calcium levels and alpha-adrenergic
receptor stimulation. Both are potent signals Alpha-Adrenergic Receptor
that govern transcriptional activity of genes Stimulation as a Regulator
important for contractility and hypertro- of Gene Expression
phy.15-17 Thus, the events that initiate contrac- The !1-adrenergic receptor has been linked
tile dysfunction seen in stunning may also be directly to changes in the expression of several
stimuli for alterations in gene expression that genes involved in cardiac growth and devel-
may govern the rate and manner of myocar- opment.19;20 It may serve as one of the pri-
dial recovery. Therefore, delayed recovery from mary instigators in altering gene expression
contractile dysfunction may reflect changes in following pressure-overload or other forms of
myocardial gene expression that are either a cardiac injury. In cultured neonatal rat ven-
maladaptive response to noxious stimuli or an tricular cardiomyocytes, !1-adrenergic stimu-
adaptive response to the hypertrophy signals lation with phenylephrine (PE) induced the
from mechanical stress induced by stunning. production and release of atrial natriuretic fac-
tor (ANF) within 12 hours, with increases in
ANF mRNA evident by 6 hours.19 In addi-
Calcium as a Regulator tion to regulating ANF expression, PE induced
of Gene Expression expression of the myosin light chain-2 tran-
Calcium is an important second messen- script, but failed to affect the mRNA level for
ger in the cell, with a variety of effects ranging the cardiac-specific sodium channel.21 This
from a role in contraction to transcriptional indicates that induction of transcription by
control. Many genes are responsive to changes !1-adrenergic receptor stimulation has speci-
in the free calcium concentration in the cell. ficity and is not a generalized process. !1-ad-
Calcium may bind to calmodulin, a calcium- renergic stimulation has a comprehensive effect
binding regulatory protein. The calcium- on cardiac gene expression. PE exerts its effect
calmodulin complex alters the activity of target on gene expression through a consensus
52 Ischemia-Reperfusion Injury in Cardiac Surgery

sequence in the promoter region of the respon- myocellular phenotype at a cellular and
sive genes. In the ANF promoter, the sequence molecular level are important. Whether
conferring PE-responsiveness has been iden- myocellular stretch, intracellular calcium con-
tified and termed the phenylephrine response centrations, and/or catecholamine stimulation
element (PERE). ! skeletal actin (!SkA) and of the myocyte act together or independently
&-MHC contain homologous regions corre- in regulation of gene expression in stunned
sponding to the PERE of ANF.22 The !1-adr- hearts is not well understood. These factors,
energic system is thought to be at least partly however, initiate a complex series of events that
responsible for the development of cardiac eventually result in altered functional and
hypertrophy,23,24 and it is characterized by in- structural characteristics. Among the first
creases in !SkA, &-MHC and ANF, which all changes seen are induction of stress proteins
respond to PE stimulation. and immediate-early genes (IEGs). Stress pro-
teins serve to help the heart survive the initial
Myocellular Stretch insult, while the IEGs are transcription fac-
as a Regulator of Gene tors that direct the expression of a multitude
of other genes under their control. Some of
Expression the genes with altered expression are structural
In addition to changes in gene expression and functional proteins which result in a new
induced through chemical mediators such as cellular phenotype. These changes in gene
calcium and !-adrenergic receptor agonists, expression alter the functional characteristics
mechanical factors such as myocyte stretch of the myocyte and thus influence the way in
may play a role in myocardial remodeling and which the heart recovers from injury.
associated changes in myocardial gene expres-
sion. Myocardium in globally stunned hearts Changes in Stress Proteins and IEGs
may be subject to abnormal stresses that elicit
There is a large, highly conserved, family
molecular markers typical of stretch-induced
of proteins whose expression is altered under
hypertrophy. For example, decreased contrac-
times of cellular stress. These stress-response
tility may lead to chamber dilatation. Stretch
proteins act to improve cellular survival under
imposed upon cardiac myocytes alters the ex-
adverse conditions, and include members of
pression of several myocardial genes, includ-
the heat shock protein (HSP) and glucose-re-
ing those encoding heat shock protein 68
sponsive protein (GRP) families. Hsp70 gene
(HSP68),25 c-fos,25,26 c-myc,25 !SkA,27 and
products are thought to assist in the import of
&-MHC.25 Ventricular loading, a determinant
nascent polypeptides into mitochondria32 and
of stretch, is an independent regulator of gene
to act as chaperones in the folding of dena-
expression in the absence of stunning. Both
tured proteins.33 Changes in levels of mRNA
ventricular overloading, as in pressure over-
encoding heat shock proteins have been well
load hypertrophy, and underloading28-30 have
established in hearts subjected to ischemia and
been linked to changes in myocardial gene
reperfusion.34-37
expression. Such changes can be modulated
The glucose response protein grp78 is
by pacing the heterotopically transplanted
another stress-response protein. Grp78 is a
heart.31 These findings collectively suggest that
constitutively expressed protein of the endo-
the load upon the ventricle during reperfusion
plasmic reticulum (ER) that is induced dur-
may affect the molecular phenotype of the
ing glucose starvation or increased calcium
stunned myocardium.
levels, and it affects protein translocation and
folding (see review by Haas38). Thus, within
Changes in Gene Expression the ER, grp78 appears to have a role similar
in Ischemia and Stunning to that of hsp70.39,40 Despite this similarity,
Regardless of the initiating signal(s) changes in expression of grp78 have not been
involved in stunning, the signals that alter the seen in global IR injury.37,41 However, we have
Changes in Myocardial Gene Expression Following Ischemia and Reperfusion 53

recently demonstrated an increase in the IEGs has been demonstrated after acute
steady-state mRNA levels encoding grp78 only pressure overload of the ventricle,53-55 regional
under conditions of ventricular unloading stunning,50 and in global myocardial ischemia
during reperfusion. Thus, while the hsp70 and and reperfusion.37,51 The timing and pattern
grp78 share similar functional characteristics, of IEG induction varies with the nature of the
the signals responsible for their expression in injury. The complicated interactions between
IR injury differ. This is consistent with previ- the different transcription factors may in part
ous data that some signals may coordinately dictate the response of the cell to ischemia/
or discordantly regulate hsp and grp expres- reperfusion injury, and the pattern of the IEG
sion.42 The result, however, is that IR injury response may be responsible for the different
induces a pattern of expression amongst the phenotypic changes seen in different models
stress-responsive proteins that appears to be of cardiac injury. Due to the complex interac-
protective in nature. tions between the different IEGs, the tempo-
The immediate-early genes (IEGs) are ral pattern of IEG expression has important
nuclear transcription factors that are involved implications in the resulting effects on the
in the regulation of gene expression in normal regulation of structural genes.
growth and development, as well as in alter- In global IR injury, we have demonstrated
ations of gene expression seen after injury. The a temporal pattern of IEG expression, with
net result of IEG expression is determined by different IEG mRNA profiles at 1 and 2 hours
the relative levels of the individual IEGs (see of reperfusion.41 This pattern of rapid changes
review by Herschman43). Members of the fos in levels of mRNA encoding individual IEGs
and jun families exert their actions as dimers. has also been demonstrated in regional
c-fos and jun-c are able to form heterodimers ischemia and reperfusion.50
that bind to the AP-1 sequence(s) in the pro- In regional ischemia and reperfusion,
moter region of certain genes. The presence expression of stress-related genes is dependent
of these binding sites provides the mechanism upon the region of the heart examined.
through which alterations in IEG expression Increased expression of the IEGs c-fos and jun-
change expression of structural cellular genes c, and the heat shock proteins hsc70 and hsp70
and thus alter the phenotype of the injured are limited to the ischemic area after IR injury,
cell. The c-fos/jun-c dimer may regulate car- as demonstrated by in situ hybridization.
diac gene expression in a positive44 or nega- However, mRNA encoding the IEG jun-B is
tive45 manner, depending on the specific gene seen in both the ischemic and nonischemic
of interest. When the IEG jun-B binds to jun- border zones.48 Temporal and regional differ-
c, c-fos binding activity of the dimer is inhib- ences in IEG expression are important since
ited.46,47 Altered expression of the IEGs has the net result of IEG expression depends upon
been demonstrated in IR injury, with increases the ratio between jun-c, jun-B and c-fos, due
in the steady-state levels of mRNA encoding to the interactions between these proteins.46,47
jun-c,41,48-50 jun-B48,50 c-fos,37,41,48,50 egr-150 However, altered IEG expression is only mean-
and c-myc.41 However, the IEG induction is ingful in that they directly impact upon the
clearly not universal in IR injury as no signifi- expression of other proteins within the cell that
cant induction was detected in the levels of have functional importance.
mRNAs encoding the IEGs egr-141,48 or
c-myc.37,50-52 Changes in Growth Factors
Induction of IEGs in response to injury While the most important function of the
provides a mechanism through which an ini- heart is supplying blood and nutrients to the
tial extracellular signal is transduced to body, it also is the source of a variety of factors
upregulate other transcription factors which that influence the activity of other cells.
can then orchestrate further changes in gene Interleukin-8 (IL-8) affects adhesion of neu-
expression, culminating in altered mechani- trophils to cardiomyocytes and results in
cal properties of the heart. Rapid induction of cytotoxicity of myocytes in vitro. IL-8 mRNA
54 Ischemia-Reperfusion Injury in Cardiac Surgery

is increased after in vivo regional IR injury, physiologic conditions. In addition, the heart
suggesting that locally produced factors play a appears to respond to IR injury through
deleterious role in IR injury.56 Similar increases increased production of some of these factors.
in the mRNAs encoding IL-6, tumor necrosis Insulin-like growth factor (IGF) has been
factor alpha (TNF-alpha), and IL-1beta have implicated in DNA synthesis and cellular pro-
been shown in the ischemic zone of hearts fol- liferation. Increases in IGF-1 and its receptor,
lowing 1-3 hours of reperfusion.57 The mRNA IGF-1R, are seen in surviving myocytes after
encoding monocyte chemoattractant protein-1 regional infarction.62 In contrast, mRNA
(MCP-1), which may play a role in monocyte encoding IGF-2 is increased by operative, but
trafficking into IR-injured myocardium, is not IR, stress following repetitive brief coro-
increased in the endothelium of small veins nary artery occlusions, while the mRNA
within the heart following IR injury.58 While encoding one receptor subtype, IGF binding
MCP-1 induction is not demonstrated in the protein-5 (IGFBP-5), is increased following
myocytes themselves, the action of MCP-1 IR injury.63 Since IGF exerts its action through
affects the environment of the myocytes, thus IGFBP, the induction of the receptor may play
influencing recovery and function. A further an important role in the adaptive processes that
link between IR injury and the initiation of occur in the heart en route to recovery. Simi-
the inflammatory response following acute larly, the heart undergoing transient ischemia
myocardial ischemia is seen in the increase in and reperfusion responds with an increase in
the mRNA encoding endothelial leukocyte mRNA encoding vascular endothelial growth
adhesion molecule 1 (E-selectin). E-selectin factor (VEGF).64,65 Local inducible VEGF
mRNA levels are not influenced by ischemia production following IR injury may be the
alone, but increase only after reperfusion.59 In mechanism for coronary collateral develop-
addition, intracellular adhesion molecule-1 ment in ischemia. In addition, mRNAs
(ICAM-1) has been implicated in neutrophil- encoding hepatocyte growth factor (HGF) and
mediated myocardial injury following IR. In its receptor c-Met, which have been implicated
situ hybridization techniques have been used in tissue regeneration and angiogensis, are
to demonstrate that ICAM-1 mRNA levels are increased after IR injury.66 While HGF mRNA
increased in ischemic myocardium within 3 levels are increased in endothelial cells and
hours of reperfusion, and are increased in interstitial cells, mRNA for c-Met was
nonischemic areas within the same heart by increased only in capillary endothelial cells.
24 hours.60 The induction of ICAM-1 in the Following regional myocardial infarction,
border zones may increase the area of injury mRNA encoding the growth factor transform-
due to additional damage to viable cells from ing growth factor &1 (TGF-&1) is increased
the inflammatory process. after 24-48 hours.67 The increase is localized
Endothelin-1 (ET-1), a potent vasocon- to myocytes, whereas increase in mRNA for
strictive peptide, is increased systemically after heparin-binding growth factor 1 (HBGF-1)
myocardial infarction. Some of the circulat- is increased in the walls of blood vessels.52
ing ET-1 may be cardiac in origin, as the While the cells of origin of many of these
mRNA encoding ET-1 is increased in injured growth factors are not myocytes, myocytes are
myocardium after 90 minutes of regional influenced by the altered chemical environ-
ischemia and 150 minutes of reperfusion.61 ment that results.
This increase in mRNA levels is correlated with Ischemia and reperfusion is associated with
an increase in immunoreactivity for ET-1 in increased production of several factors that
the area. These data demonstrate that changes regulate the way in which the myocardium
in cardiac gene expression may play a substan- responds to its local environment. While these
tial role in the overall response of the organ- changes may be important in the injury
ism to myocardial stunning. response of the heart, the changes within the
Cells within the heart are capable of pro- structure and function of the myocytes may
ducing a variety of growth factors under
Changes in Myocardial Gene Expression Following Ischemia and Reperfusion 55

be even more important to the survival of the proteins can have a dramatic impact upon
organism. intracellular calcium flux and therefore myo-
cardial contraction, these data provide a pos-
Changes in Structural or Functional sible mechanism to explain prolonged contrac-
Proteins tile dysfunction after global stunning.
Alterations in either the amount or isoform Energy production and consumption are
prevalence of a structural or functional myo- markedly altered in ischemic and postischemic
cardial protein can have a significant impact myocardium. The expression of several genes
on the contractile properties of the heart. This involved in these processes is also altered in
has been extensively illustrated in the contrac- IR injury and may be related to some of the
tile changes shown in association with nor- alterations observed. In regional ischemia, the
mal development, acute injury, and end-stage mRNA encoding the cardiac glucose trans-
heart disease. The mechanisms of myocardial porter GLUT1 is increased in both ischemic
recovery after IR injury may involve similar and nonischemic myocardium, whereas that
changes in the expression of structural or func- for GLUT4 is unaltered.70,71 The cardiac ATP-
tional myocardial proteins. sensitive potassium channel (KATP) serves to
We have studied changes in the expression protect the heart during ischemia. Its mRNA
of several proteins of the sarcoplasmic reticu- is increased in myocardium subjected to pro-
lum (SR). These include the SR calcium longed periods (60 minutes) of regional-
ATPase (SERCA2) responsible for calcium ischemia followed by 24 hours of reper-
uptake into the SR, its regulatory protein, fusion.72 While such changes cannot influence
phospholamban, and the SR calcium release the immediate recovery of stunned myocar-
channel, the ryanodine receptor (RyR). dium, they may regulate the rate and degree
Changes in the ratio of SERCA2 to phos- of recovery.
pholamban are associated with altered contrac- The changes in gene expression seen in IR
tile properties of the myocardium during nor- injury are selective. Most genes investigated
mal myocardial development or end-stage show no significant change in mRNA levels.
failure. After global IR injury, there is discor- In addition, the mRNA levels of some genes
dant regulation of the mRNA species encod- are depressed by ischemia and reperfusion.
ing these proteins, such that altered SERCA2 However, this may be more difficult to detect
to phospholamban ratio results.41 Over time, in the early period following injury. Since the
these changes in steady state mRNA levels half-life of most mRNA species is measured
would be reflected in alterations at the pro- in hours, an early decline in mRNA levels may
tein level, which would produce altered con- signify an active degradation of the message
tractile properties in the recovering heart. In as well as a decrease in active production of
contrast to the changes seen after a single that message. The mRNA for creatine kinase
period of global IR injury, a different pattern M is decreased 40% in ischemic myocardium,
of change is seen after brief periods of repeti- whereas that encoding myosin heavy chain is
tive ischemia in both a porcine model of unaltered.36
regional IR68 and rabbit model of global IR
injury.69 Thus, the manner in which IR injury Implications of Changes in Gene
occurs may influence the direction of change Expression
for individual mRNA species. Finally, we have The cellular stress response is a complex
found that the manner of reperfusion signifi- orchestration of molecular signals that allow
cantly impacts the expression of SR proteins. the cell to adapt to a rapidly changing envi-
Ventricular unloading during reperfusion ame- ronment. These signals are ubiquitous, and
liorates changes in SR gene expression seen in their expression varies at different times dur-
global stunning.41 Because alterations in the ing growth and development and at times of
relative ratios of SR calcium regulatory cellular stress or injury. It includes members
56 Ischemia-Reperfusion Injury in Cardiac Surgery

of the heat shock family of proteins and nuclear References


transcription factors. Ischemia and reperfusion 1. Bolli R. Mechanism of myocardial “stun-
injury has been associated with altered expres- ning”. Circ. 1990; 82:723-738.
sion of several genes of this class. 2. Kusuoka H, Marban E. Cellular mechanisms
Changes in myocardial gene expression of myocardial stunning. Ann Rev Physiol
have been demonstrated in a wide variety of 1992; 54:243-256.
3. Schomig A. Catecholamines in myocardial
injury models, suggesting that this phenom- ischemia: Systemic and cardiac release. Circ
enon is a vital part of the adaptive mechanisms 1990; 82:II-13-II-22
of the stressed heart. In particular, changes in 4. Lompre AM, Anger M, Levitsky D.
contractile function have been correlated with Sarco(endo)plasmic reticulum calcium
altered expression of functional proteins, such pumps in the cardiovascular system: Func-
tion and gene expression. J Mol Cell Cardiol
as the genes encoding SR proteins. Indeed, 1994; 26:1109-1121.
changes in the expression of mRNAs for SR 5. Moorman AFM, Vermeulen JLM, Koban
proteins have been noted in hypothyroid- MU et al. Patterns of expression of sarcoplas-
ism,7,73 hyperthyroidism,7,73,74 ventricular mic reticulum Ca2+-ATPase and phospho-
lamban mRNAs during rat heart develop-
hypertrophy following overload7,8,75 and in ment. Circ Res 1995;76:616-625
congestive heart failure. In failed human 6. Brillantes AMB, Bezprozvannaya S, Marks
hearts, decreased mRNA encoding SERCA2 AR. Developmental and tissue-specific regu-
was correlated with a decrease in the cardiac lation of rabbit skeletal and cardiac muscle
index prior to explantation.76 Thus, changes calcium channels involved in excitation-
contraction coupling. Circ Res 1994; 75:
in expression of SR gene products in several 503-510.
models of myocardial remodeling are accom- 7. Nagai R, Zarain-Herzberg A, Brandl CJ et
panied by altered contractile properties, con- al. Regulation of myocardial Ca2+-ATPase and
sistent with the importance of the SR pheno- phospholamban mRNA expression in re-
sponse to pressure overload and thyroid hor-
type in myocardial function. mone. Proc Natl Acad Sci USA 1989; 86:
2966-2970.
Conclusions 8. de la Bastie D, Levitsky D, Rappaport L et
Myocardial stunning has at least two major al.Function of the sarcoplasmic reticulum and
expression of its Ca2+-ATPase gene in pres-
components: functional and molecular. The sure overload-induced cardiac hypertrophy in
functional component is the reduced contrac- the rat. Circ Res 1990; 66:554-564.
tility of the myocardium and depressed 9. Lattion AL, Michel JB, Arnauld E et al.
organellar (mitochondrial and SR) function Myocardial recruitment during ANF mRNA
increase with volume overload in the rat. Am
seen after a significant but nonlethal injury. J Physiol 1986; 251:H890-H896.
There is also a myocardial molecular response 10. Mercadier JJ, Samuel JL, Michel JB et al.
to stunning. This coordinated molecular Atrial natriuretic factor gene expression in rat
response reflects the degree of myocardial in- ventricle during experimental hypertension.
jury, since interventions known to improve Am J Physiol 1989; 257:H979-H987.
11. Black FM, Packer SE, Parker TG et al. The
myocardial function after global ischemia vascular smooth muscle !-actin gene is reac-
result in alterations in the pattern of this tivated during cardiac hypertrophy provoked
response. Given that amelioration of stunning by load. J Clin Invest 1991; 88:1581-1588.
is associated with specific changes in this mo- 12. Schiaffino S, Samuel JL, Sassoon D et al.
Nonsynchronous accumulation of !-skeletal
lecular response, we propose that the injured actin and &-myosin heavy chain mRNAs dur-
myocardium has initiated a reparative program ing early stages of pressure-overload-induced
to restore proper myocardial function. hypertrophy demonstrated by in situ hybrid-
ization. Circ Res 1989;64:937-948.
13. Knowlton KU. Atrial natriuretic factor: A
molecular marker for cardiac specific, embry-
onic, and inducible gene expression. Heart
Failure 1992; June/July:121-128.
Changes in Myocardial Gene Expression Following Ischemia and Reperfusion 57

14. Schaub MC, Hefti MA, Harder BA et al. 26. Komuro I, Kaida T, Shibazaki Y et al.
Various hypertrophic stimuli induce distinct Stretching cardiac myocytes stimulates proto-
phenotypes in cardiomyocytes. J Mol Med oncogene expression. J Biol Chem 1990;
1997; 75:901-920. 265:3595-3598.
15. Bishopric NH, Kedes L. Adrenergic regula- 27. Komuro I, Katoh Y, Kaida T et al. Mechani-
tion of the skeletal !-actin gene promoter cal loading stimulates cell hypertrophy and
during myocardial cell hypertrophy. Proc Natl specific gene expression in cultured rat car-
Acad Sci USA 1991; 88:2132-2136. diac myocytes: Possible role of protein kinase
16. Bishopric NH, Simpson PC, Ordahl CP. In- C activation. J Biol Chem 1991; 266:1265-
duction of the skeletal !-actin gene in !1- 1268.
adrenoceptor-mediated hypertrophy of rat 28. Samarel AM, Engelmann GL. Contractile
cardiac myocytes. J Clin Invest 1987;80:1194- activity modulates myosin heavy chain-& ex-
1199. pression in neonatal rat heart cells. Am J
17. Waspe LE, Ordahl CP, Simpson P. The car- Physiol 1991; 261:H1067-H1077.
diac &-myosin heavy chain isogene is induced 29. Klein I, Ojamaa K, Samarel AM et al. He-
selectively in !1-adrenergic receptor-stimulated modynamic regulation of myosin heavy chain
hypertrophy of cultured rat heart myocytes. gene expression: Studies in the transplanted
J Clin Invest 1990;85:1206-1214. rat heart. J Clin Invest 1992; 89:68-73.
18. Sadoshima J, Izumo S. Mechanical stretch 30. Buchwald A, Klein HH, Lindert S et al. Ef-
rapidly activates multiple signal transduction fect of intracoronary superoxide dismutase on
pathways in cardiac myocytes: Potential in- regional function in stunned myocardium. J
volvement of an autocrine/paracrine mecha- Cardiovasc Pharm 1989; 13:258-264.
nism. EMBO J 1993;12:1681-1692. 31. Geenen DL, Malhotra A, Buttrick PM et al.
19. Knowlton KU, Baracchini E, Ross RS et al. Increased heart rate prevents the isomyosin
Coregulation of the atrial natriuretic factor shift after cardiac transplantation in the rat.
and cardiac myosin light chain-2 genes dur- Circ Res 1992; 70:554-558.
ing !-adrenergic stimulation of neonatal rat 32. Gambill BD, Voos W, Kang PJ et al. A dual
ventricular cells: Identification of cis sequences role for mitochondrial Heat Shock Protein 70
within an embryonic and a constitutive in membrane translocation of preproteins. J
contractile protein gene which mediate in- Cell Biol 1993; 123:109-117.
ducible expression. J Biol Chem 1991; 266: 33. Lindquist S, Craig EA. The heat shock pro-
7759-7768. teins. Ann Rev Genet 1988; 22:631-677.
20. Springhorn JP, Ellingsen O, Berger H-J et 34. Knowlton AA, Brecher P, Apstein CS. Rapid
al. Transcriptional regulation in cardiac expression of heat shock protein after brief
muscle: Coordinate expression of Id with a cardiac ischemia. J Clin Invest 1991; 87:
neonatal phenotype during development and 139-147.
following a hypertrophic stimulus in adult rat 35. Dillman WH, Mehta HB, Barrieux A et al.
ventricular myocytes in vitro. J Biol Chem Ischemia of the dog heart induces the appear-
1992; 267:14360-14365. ance of a cardiac mRNA coding for a pro-
21. Shubeita HE, McDonough PM, Harris AN tein with migration characteristics similar to
et al. Endothelin induction of inositol phos- heat-shock/stress protein 71. Circ Res 1986;
pholipid hydrolysis, sarcomere assembly, and 59:110-114.
cardiac gene expression in ventricular myo- 36. Mehta HB, Popovich BK, Dillman WH. Is-
cytes: A paracrine mechanism for myocardial chemia induces changes in the level of
cell hypertrophy. J Biol Chem 1990; 265: mRNAs coding for stress protein 71 and cre-
20555-20562. atine kinase M. Circ Res 1988; 63:512-517.
22. Ardati A, Nemer M. A nuclear pathway for 37. Wechsler AS, Entwistle JCI, Yeh T Jr et al.
!1-adrenergic receptor signaling in cardiac Early gene changes in myocardial ischemia.
cells. EMBO J 1993;12:5131-5139. Ann Thorac Surg 1994; 58:1282-1284.
23. Simpson P. Norepinephrine-stimulated hyper- 38. Haas IG. BiP (GRP78), an essential hsp70
trophy of cultured rat myocardial cells is an resident protein in the endoplasmic reticulum.
!1-adrenergic response. J Clin Invest 1983; Experientia 1994; 50:1012-1020.
72:732-738. 39. Kozutsumi Y, Segal M, Normington K. The
24. Simpson P, Bishopric N, Coughlin S et al. presence of malfolded proteins in the endo-
Dual trophic effects of the alpha1-adrenergic plasmic reticulum signals the induction of
receptor in cultured neonatal rat heart muscle glucose-regulated proteins. Nature 1988;
cells. J Mol Cell Cardiol 1986; 18:45-58. 332:462-464.
25. Delcayre C, Klug D, Van Thiem N et al. 40. Pelham HRB. Speculations on the functions
Aortic perfusion pressure as early determinant of the major heat shock and glucose-related
of &-isomyosin expression in perfused hearts. proteins. Cell 1986; 46:959-961.
Am J Physiol 1992;263:H1537-H1545
58 Ischemia-Reperfusion Injury in Cardiac Surgery

41. Entwistle JWC III. Changes in the expres- 54. Pollack PS, Houser SR, Budjak R et al. c-myc
sion of sarcoplasmic reticulum protein and gene expression is localized to the myocyte
stress-related genes in stunned myocardium: following hemodynamic overload in vivo. J
Effects of unloading during early reperfusion. Cell Biochem 1994; 54:78-84.
Virginia Commonwealth University 1998; 55. Rozich JD, Barnes MA, Schmid PG et al.
1-83. Load effects on gene expression during car-
42. Watowich SS, Morimoto RI. Complex regu- diac hypertrophy. J Mol Cell Cardiol 1995;
lation of heat shock- and glucose-responsive 27:485-499.
genes in human cells. Mol Cell Biol 1988; 56. Kukielka GL, Smith CW, LaRosa GL et al.
8:393-405. Interleukin-8 gene induction in the myocar-
43. Herschman HR. Primary response genes in- dium after ischemia and reperfusion in vivo.
duced by growth factors and tumor promot- J Clin Invest 1995; 95:89-103.
ers. Annual Review of Biochemistry 1991; 57. Chandrasekar B, Colston JT, Freeman GL.
60:281-319. Induction of proinflammatory cytokine and
44. Bishopric NH, Jayasena V, Webster KA. Posi- antioxidant enzyme gene expression follow-
tive regulation of the skeletal !-actin gene by ing brief myocardial ischemia. Clin Exp
Fos and Jun in cardiac myocytes. J Biol Immunol 1997; 108:346-351.
Chem 1992; 267:25535-25540. 58. Kumar AG, Ballantyne CM, Michael LH et
45. McBride K, Robitaille L, Tremblay S et al. al. Induction of monocyte chemoattractant
fos/jun repression of cardiac-specific transcrip- protein-1 in the small veins of the ischemic
tion in quiescent and growth-stimulated and reperfused canine myocardium. Circ.
myocytes is targeted at a tissue-specific cis 1997; 95:693-700.
element. Mol Cell Biol 1993; 13:600-612. 59. Billups KL, Palladino MA, Hinton BT et al.
46. Chiu R, Angel P, Karin M. Jun-b differs in Expression of E-selectin mRNA during is-
its biological properties from, and is a nega- chemia/reperfusion injury. J Lab Clin Med
tive regulator of, c-jun. Cell 1989; 59: 1995; 125:626-633.
979-986. 60. Youker KA, Hawkins HK, Kukielka GL et
47. Schutte J, Viallet J, Nau M et al. jun-b in- al. Molecular evidence for induction of in-
hibits and c-fos stimulates the transforming tracellular adhesion molecule- in the viable
and trans-activating activities of c-jun. Cell border zone associated with ischemia-reper-
1989; 59:987-997. fusion injury of the dog heart. Circ. 1994;
48. Plumier JC, Robertson HA, Currie RW. Dif- 89:2736-2746.
ferential accumulation of mRNA for imme- 61. Tonnessen T, Giaid A, Saleh D et al. In-
diate early genes and heat shock genes in creased in vivo expression and production of
heart after ischemic injury. J Mol Cell Cardiol endothelin-1 by porcine cardiomyocytes sub-
1996; 28:1251-1260. jected to ischemia. Circ Res 1995; 76:
49. Mizukami Y, Yoshida K, Morimoto S. A 767-772.
novel mechanism of JNK1 activation. Nuclear 62. Reiss K, Kajstura J, Zhang X et al. Acute
translocation and activation of JNK1 during myocardial infarction leads to upregulation of
ischemia and reperfusion. J Biol Chem 1997; the IGF-1 autocrine system, DNA replication,
272:16657-16662. and nuclear mitotic division in remaining vi-
50. Brand T, Sharma HS, Fleischmann KE et al. able cardiac myocytes. Exp Cell Res 1994;
Protooncogene expression in porcine myocar- 213:463-472.
dium subjected to ischemia and reperfusion. 63. Kluge A, Zimmermann R, Münkel B et al.
Circ Res 1992; 71:1351-1360. Insulin-like growth factor II is an experimen-
51. Wechsler AS, Entwistle JWCI, Ding M et al. tal stress inducible gene in a porcine model
Myocardial stunning: Association with altered of brief coronary occlusions. Cardiovasc Res
gene expression. J Card Surg 1994; 9: 1995; 29:708-716.
537-542. 64. Banai S, Shweiki D, Pinson A et al. Up-
52. Sharma HS, Wünsch M, Brand T et al. regulation of vascular endothelial growth fac-
Molecular biology of the coronary vascu- tor expression induced by myocardial is-
lar and myocardial responses to ischemia. chaemia: Implications for coronary angiogen-
J Cardiovasc Pharm 1992; 20(Suppl 1): esis. Cardiovasc Res 1994; 28:1176-1179.
S23-S31. 65. Hashimoto E, Ogita T, Nakaoka T et al.
53. Komuro I, Kurabayashi M, Takaku F et al. Rapid induction of vascular endothelial
Expression of cellular oncogenes in the myo- growth factor expression bt transient ischemia
cardium during the developmental stage and in rat heart. Am J Physiol 1994; 267:H1948-
pressure-overloaded hypertrophy of the rat H1954.
heart. Circ Res 1988; 62:1075-1079.
Changes in Myocardial Gene Expression Following Ischemia and Reperfusion 59

66. Ono K, Matsumori A, Shioi T et al. En- 73. Arai M, Otsu K, MacLennan DH et al. Ef-
hanced expression of hepatocyte growth fect of thyroid hormone on the expression of
factor/c-Met by myocardial ischemia and mRNA encoding sarcoplasmic reticulum pro-
reperfusion in a rat model. Circ 1997; 95: teins. Circ Res 1991; 69:266-276.
2552-2558. 74. Kimura Y, Otsu K, Nishida K et al. Thyroid
67. Thompson NL, Bazoberry F, Speir EH et al. hormone enhances Ca2+ pumping activity of
Transforming growth factor beta-1 in acute the cardiac sarcoplasmic reticulum by increas-
myocardial infarction in rats. Growth Factors ing Ca 2+ATPase and decreasing phospho-
1988; 1:91-99. lamban expression. J Mol Cell Cardiol 1994;
68. Frass O, Sharma HS, Knöll R et al. Enhanced 26:1145-1154.
gene expression in stunned porcine myocar- 75. Komuro I, Kurabayashi M, Shibazaki Y et al.
dium. Cardiovasc Res 1993; 27:2037-2043. Molecular cloning and characterization of a
69. Crandall JD. Modulation of sarcoplasmic Ca2+ + Mg2+-dependent adenosine triphos-
reticulum gene expression following global phatase from rat cardiac sarcoplasmic reticu-
myocardial ischemia and reperfusion. Virginia lum: Regulation of its expression by pressure
Commonwealth University 1996; 21-36. overload and developmental stage. J Clin In-
70. Brosius FCI, Liu Y, Nguyen N et al. Persis- vest 1989; 83:1102-1108.
tent myocardial ischemia increases GLUT1 76. Mercadier JJ, Lompre AM, Duc P et al. Al-
glucose transporter expression in both is- tered sarcoplasmic reticulum Ca2+-ATPase
chemic and nonischemic heart regions. J Mol gene expression in the human ventricle dur-
Cell Cardiol 1997; 29:1675-1685. ing end-stage heart failure. J Clin Invest
71. Brosius FCI, Nguyen N, Egert S et al. In- 1990; 85:305-309.
creased sarcolemml glucose transporter abun-
dance in myocardial ischemia. Amer J Cardiol
1997; 80:77A-84A.
72. Akao M, Otani H, Horie M et al. Myocar-
dial ischemia induces different regulation of
KATP channel gene expression in rat hearts.
J Clin Invest 1997; 100:3053-3059.
Section II:
Methods to Prevent
Ischemia-Reperfusion Injury
CHAPTER 5

The NO-Donor L-Arginine


Reduces the Reperfusion Injury
after Heart Transplantation
Gábór Szabó

I
schemia-reperfusion injury is a common pathway plays an important role in ischemia-
condition during cardiac surgery. reperfusion injury. In the heart, NO is syn-
Myocardial performance within the first thesized from oxygen and L-arginine by the
hours after the surgical procedure determines coronary endothelium,4 cardiac myocytes5 and
the patient’s state not only during the postop- endocardial cells.6 It has a host of physiologi-
erative period but also for the long time out- cal effects, including autoregulatory modula-
come, especially after heart transplantation tion of coronary blood flow7,8 and inhibition
when an extended time of ischemia is followed of neutrophil endothelial interaction9,10 as well
by reperfusion. Most studies about the effects as platelet aggregation. In contrast to the well
of myocardial ischemia and reperfusion focus understood physiological roles of NO in heart
on myocardial injury and the recovery of con- function, its significance under pathophysio-
tractile function. It is now appreciated that the logical states remains unclear. On one hand
survival of the heart as a whole is in part NO is shown to have a beneficial effect by
dependent on the ability of the microcircula- increasing postischemic blood flow and
tion to deliver and distribute blood flow ad- decreasing the ‘no-reflow’ phenomenon,11
equately during reperfusion. Recent studies decreasing leukocyte adhesion and of cell
show the importance of protecting the mi- adhesions molecules12 and quenching free radi-
crovasculature to attenuate reperfusion in- cals, particularly superoxide radicals.13 Con-
jury.1-3 Preservation of microvascular reactiv- trarily, some authors report deleterious effects
ity may be especially important in those hearts of NO during cardiac ischemia and reper-
undergoing cold ischemic storage for trans- fusion probably by a direct negative inotropic
plantation. Potential impairment of the right action of NO, or inhibition of mitochondrial
ventricular endothelium-dependent microvas- respiration or formation of peroxynitrite, a
cular response after harvesting and implanta- precursor of highly oxidant species.14-16
tion is also a likely contributing factor in In a recent study, we investigated the short
right ventricular failure after cardiac and long time effects of altered NO-synthesis
transplantation.2 on reperfusion injury after reversible deep
hypothermic ischemia in a modified hetero-
Effects of Nitric Oxide topic rat heart transplantation model.
Enhancement of NO-synthesis was achieved
During the past few years, several studies by L-arginine and blocking of NO synthesis
reported that the L-arginine-nitric oxide (NO) by L-NAME. Left ventricular pressure-volume

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


62 Ischemia-Reperfusion Injury in Cardiac Surgery

relationships were measured by an intraven- endothelium-dependent vasodilatation after


tricular balloon which was connected to a pres- infusion of acetylcholine (30 ∝g/min),
sure transducer and myocardial blood flow was L-NAME significantly impaired myocardial
assessed by the hydrogen-clearance tech- blood flow (Fig. 5.3). The endothelium-inde-
nique.17,18 This study demonstrated that the pendent vasodilatation after infusion of
L-arginine-NO pathway has an important sodium-nitroprussid (200 ∝g/min) was not
beneficial effect on reperfusion injury after influenced by altered NO-synthesis. Although
reversible deep hypothermic ischemia in the systolic and diastolic function and baseline
heterotopically transplanted rat heart. The MBF were similar after 24 hours of reperfusion
rapid infusion of the NO-precursor L-argin- in all groups, endothelium-dependent vasodi-
ine (40 mg/kg) during the first five minutes latation was still significantly better in the
of reperfusion led to an increased functional L-arginine than in the control group and both
recovery after 60 minutes of reperfusion showed a better endothelium-dependent
(Figs. 5.1, 5.2). Simultaneous administration vasodilatation than the L-NAME group in-
of L-arginine and L-NAME (10 mg/kg), an dicating a persisting beneficial effect of
inhibitor of endogenous NO-synthesis, NO-synthesis on endothelial function dur-
antagonized these effects suggesting that func- ing reperfusion (Fig. 5.3).
tional changes can be attributed to NOS Histologic findings revealed a slight edema
enhancement or inhibition, respectively. Fur- and inflammatory perivascular infiltrate com-
thermore, L-NAME alone reduced recovery posed predominantly of polymorphonuclear
of left ventricular function. While adminis- neutrophils and lymphocytes in all treated
tration of L-arginine led to a leftward shift of transplanted hearts in comparison with the
the left ventricular systolic pressure-volume native hearts of the recipients after 60 min-
curves suggesting higher contractility in com- utes of reperfusion. While, however, only spo-
parison to control, the left ventricular systolic radic adherent neutrophils and scare cardiac
pressure-volume curves showed a significant infiltration were found in the L-arginine
rightward shift in the L-NAME group indi- group, the control- and the L-arginine +
cating decreased systolic function after L-NAME group showed an increased infiltra-
reperfusion. The slope of this relationship tion of neutrophils. These findings were more
Emax was significantly higher in the L-argin- profound in the L-NAME group where also
ine and significantly lower in the L-NAME extended infiltrated areas were found includ-
group. Active diastolic function, myocardial ing not only the perivascular but the cardiac
relaxation, was significantly improved after muscle tissue. After 24 hours, no differences
L-arginine as indicated by the lower isovolumic were observed between the groups also in com-
relaxation constant (TE) and severely impaired parison to the native hearts.
by application of L-NAME (Fig. 5.2). Left
ventricular compliance did not differ between Reduction of Reperfusion Injury
the groups except the L-NAME treated ani- by Nitric Oxide: Potential
mals after 60 minutes of reperfusion. The fact
that passive diastolic function was also Mechanisms
impaired (Fig. 5.2) suggests that the relative The exact mechanism by which endo-
integrity of endogenous NO-synthesis may be genous NO synthesis has a beneficial effect
essential for limiting reperfusion injury. on reperfusion injury remains controversially
The reduction of reperfusion injury by discussed. One possible mechanism would be
L-arginine can be explained by an increase through increased postischemic coronary
of postischemic myocardial blood flow blood flow,19-21 assuming that endothelial NO
(Table 5.1) and decrease of neutrophil adher- release is increased by supplying more substrate
ence. In contrast, L-NAME led to a signifi- for NO production.19 Endothelium derived
cant decrease of myocardial blood flow. NO is a potent vasodilator and exerts its effect
Whereas L-arginine significantly improved the by stimulating soluble guanylate cyclase in the
The NO-Donor L-Arginine Reduces the Reperfusion Injury after Heart Transplantation 63

Fig. 5.1. Left ventricular systolic pressure-volume relationships (left panel) and the slope of the left ventricular
systolic pressure-volume relationships (Emax, right panel) in the heterotopically transplanted rat heart after 60
minutes and 24 hours of reperfusion. L-Arg, L-arginine, L-NAME, nitro-L-arginine methyl ester, LVPSP, left
ventricular peak systolic pressure, LVV left ventricular volume. All values are given as mean ± SEM. *p < 0.05 all
groups vs. control at a given time point, Üp < 0.05 24 hours vs. 60 minutes.

underlying vascular smooth muscle cells, studies also showed a protective effect of NO-
thereby elevating intracellular levels of cGMP synthesis regarding functional recovery11,24 and
and inducing relaxation of vascular smooth postischemic blood flow11 in the setting of
muscle. 22 The significantly higher post- regional ischemia and reperfusion. Pinsky et
ischemic myocardial blood flow after L-argin- al25 demonstrated a significantly higher graft
ine and the significantly lower postischemic survival and better functional recovery assessed
myocardial blood flow after L-NAME in our by a semiquantitative scoring system after
preparation would support this hypothesis. application of the exogenous NO-donor ni-
These results are consistent with the study of troglycerin in heterotopic transplants after 12
Hiramatsu et al19 who observed a significant hours of ischemia.
increase of coronary blood flow and subse- NO is not only a vasodilator but also in-
quent improvement of left ventricular func- hibits interaction between the vessel wall and
tional recovery after cold ischemia in blood circulating blood elements.26-28 It has been
perfused isolated lamb hearts if L-arginine was shown that neutrophil-endothelial interaction
supplemented during the first 20 minutes of plays a crucial role in reperfusion injury.
reperfusion. In the same study, L-NAME Incited by the paracrine factors released by the
resulted in a significant decrease of post- endothelium, the neutrophils begin to gener-
ischemic coronary blood flow and left ven- ate oxygen derived free radicals as well as pro-
tricular functional recovery. Amrani et al23 teases which compound the endothelial injury
showed a 31% loss of postischemic coronary and induce myocyte necrosis. In addition, the
flow by application of L-NMMA, another in- cytokines released from the activated neutro-
hibitor of NO-synthesis, after four-hour cold phils induce the expression of other adhesion
ischemia in oxygenated, Krebs-Henseleit, molecules (e.g., ICAM-1, E-selectine) that
buffer-perfused, isolated rat hearts. Other sustain the inflammatory process.29 Recent
64 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 5.2. Left ventricular end-diastolic pressure-volume relationships in the heterotopically transplanted rat heart
after 60 minutes and 24 hours of reperfusion (left panel). The changes of myocardial relaxation (TE, isovolumic
relaxation constant) are shown on the right panel. L-Arg, L-arginine, L-NAME, nitro-L-arginine methyl ester,
LVEDP, left ventricular end-diastolic pressure, LVV left ventricular volume. All values are given as mean ± SEM.
*p < 0.05 all groups vs. control at a given time point, †p < 0.05 24 hours vs. 60 minutes.

Table 5.1. Parameters of the coronary circulation

Control L-Arg L-NAME L-NAME+L-Arg

MBF [ml min-1 g-1]


60 minutes 1.9 ± 0.2 3.6 ± 0.4# 1.2 ± 0.1# 2.0 ± 0.2
24 hours 3.4 ± 0.3* 3.9 ± 0.4 3.0 ± 0.2* 3.8 ± 0.3*
CVR [U/g]
60 minutes 47 ± 6 25 ± 3# 75 ± 7# 52 ±8
24 hours 36 ± 9* 27 ± 10 34 ± 4* 28 ± 5*
L-Arg, L-arginine, L-NAME, nitro-L-arginine methyl ester. MBF, myocardial blood flow, CVR, coronary
vascular resistance. All values are given as mean ± SEM at an intraventricular volume of 80 ∝l.
*p < 0.05 24 hours vs. 60 minutes; #p < 0.05 vs. control

studies24,25,30,31 have shown that modulation NO-donor treated animals after regional
of the NO-pathway may influence neutrophil- ischemia and reperfusion. Furthermore,
mediated reperfusion injury. Pinsky et al25 and decreased expression of endothelial and soluble
Pabla et al 31 demonstrated a histologic adhesion molecules could be found after treat-
evidence of decreased neutrophil adhesion ment with NO-donor.24,30,32
under enhanced and increased neutrophil NO may also play a direct role by genera-
adhesion under attenuated NO-synthesis. In tion of radicals. NO itself is a primary radical
a previous study of Pabla et al 11 cardiac species and is inactivated by superoxide radi-
myeloperoxidase activity, an index of neutro- cals. 33,34 Conversely, NO may neutralize
phil accumulation was significantly lower in superoxide radicals.35 Decreased endothelial
The NO-Donor L-Arginine Reduces the Reperfusion Injury after Heart Transplantation 65

Fig. 5.3. Myocardial blood flow response after application of the endothelium-dependent vasodilator acetylcholine
(left panel) and the endothelium-independent vasodilator sodium nitroprussid (right panel). L-Arg, L-arginine,
L-NAME, nitro-L-arginine methyl ester. All values are given as mean ± SEM. *p < 0.05 all groups vs. control at
a given time point. Endothelium-dependent vasodilatation significantly (p < 0.05) improved in all treatment
groups after 24 hours in comparison to 60 minutes of reperfusion (not indicated in the graph).

production of NO with L-NAME infusion of superoxide is permitted to react with NO


may therefore lead to an enhanced generation because of an approximately 100-fold higher
of free radicals and increases endothelial and presence of superoxide dismutase, it is possible
myocardial injury during reperfusion. On the that under ischemia/reperfusion, increased
other hand, L-arginine may reduce free radi- concentrations of superoxide anion and
cal mediated injury. Siegfried et al13 described decreased concentrations of superoxide
cardioprotection and attenuation of endothe- dismutase allow the generation of peroxynitrite
lial dysfunction by organic nitric oxide donors in a significant amount. Maulik et al39 sug-
in myocardial ischemia-reperfusion. They gested that NO plays a significant role in trans-
hypothesized that NO may quench other free membrane signaling via cGMP by opposing
radicals, particularly superoxide radicals and the effects of phosphodiesterases by inhibit-
this may contribute to its protective effect. In ing the ischemia/reperfusion-induced phos-
contrast, some authors suggested that NO has phodiesteratic break down. They postulated
a detrimental influence during ischemia and an on- and off-NO-signaling which is linked
reperfusion,14,15,36 primarily due to generation to its free radical chemistry.
of peroxynitrite from superoxide anion and The relative importance of different pos-
NO. Normally, free NO is catabolized by sible effects of NO remains unclear during
binding with hemoglobin to form nitrosyl- reperfusion. Several authors underline, that the
hemoglobin which is subsequently oxidized to beneficial effects of NO are primarily due to
form nitrate and to a lesser content nitrite in a the modulation of postischemic blood
relatively slow reaction.37 However, NO also flow.11,19,23,40,41 The fact that NO showed a
may form peroxynitrite as described above in protection against ischemia-reperfusion injury
a rapid diffusion limited reaction.38 Although in crystalloid perfused heart preparations,
under normal conditions only a small amount where neutrophil-mediated actions of NO
66 Ischemia-Reperfusion Injury in Cardiac Surgery

seem to be of minor importance, would sup- a clear and marked expression of eNOS in
port this hypothesis. In contrast, in a recent endothelial cells and no expression of iNOS
study31 NO or L-arginine had only a cardio- in native fresh hearts by immunohistology. The
protective effect if polymorphonuclear leuko- semiquantitative analysis of the immunohis-
cytes were present underlining the importance tologic stainings showed a significant decrease
of NO-mediated inhibition of neutrophil- of eNOS after 60 minutes of deep hypother-
endothelial interaction. Despite a decreased mia and 60 minutes of reperfusion whereas
postischemic blood flow, Naseem et al 15 the decrease was less pronounced in the con-
reported better functional recovery after block- trol hearts and more pronounced in the
ing of NO-synthesis by attenuated free radi- L-NAME-treated group. On the other hand
cal generation. Takeuchi et al42 found in crys- iNOS was expressed in all ischemic groups
talloid perfused isolated rabbit hearts that without any differences between the groups.
L-arginine had no effect on postischemic myo- This is in agreement with the study of Wang
cardial blood flow, but significantly decreased et al45 who found after ischemia-reperfusion,
functional recovery after normoterm no-flow that tissue activity of eNOS in L-arginine
ischemia. They suggested that NO has a direct treated hearts was approximately the same as
detrimental action by inhibition of mitochon- in nonischemic hearts while control vehicle
drial respiration or accumulation of intracel- treated hearts showed a significant decrease of
lular calcium during reperfusion. A possible eNOS activity. Interestingly, application of a
explanation for the conflicting results regard- blocker of NO-synthesis, L-NNA did not
ing the role of NO during reperfusion may be cause any further changes in eNOS activity.
that NO may have an adverse effect on This phenomenon is most likely, because
ischemia-reperfusion injury depending on the ischemia and reperfusion reduced eNOS
time of application: the blocking of NO-syn- activity to such a degree that addition of NO-
thesis was shown to be beneficial during blocker did not further inhibit the NOS
ischemia and NO-donation may improve activity. In contrast to Wang et al,45 who sub-
functional recovery during reperfusion.14,16 jected the hearts to a 30-minute global warm
Furthermore, the applied doses of NO- ischemia, in our previous40,44 studies, after
blockers/donors may also play a role in the deep hypothermia and reperfusion eNOS
net effect of these drugs.14,16 In vasoactive activity was probably moderately impaired,
doses, the net effect of NO-donors is prob- and therefore blocking of NO-synthesis by
ably dominated by the vasodilatative property L-NAME may have led to a further decrease
of NO. In nonvasoactive doses, a protective of eNOS activity. Consistent with the report
role of endogenous NO-synthesis was shown of Wang et al, the fact that iNOS expression
only during reperfusion, while during ischemia was almost similar in all groups suggests that
had a negative effect.16 effects of modulation of NO synthesis are car-
The role of different NOS isoforms in ried out primarily by eNOS mediated NO-
reperfusion injury may also explain at least synthesis in this setting. However, under cer-
partly the conflicting results. While eNOS is tain circumstances, when eNOS activity or
calcium dependent and is usually constitu- L-arginine concentration is low, the blocking
tively present in predominantly in endothelial of NO-synthesis may be beneficial by
cells but also in myocardial cells, iNOS is cal- inhibiting iNOS mediated NO-synthesis and
cium-independent and is expressed in various subsequent free radical generation. eNOS also
types of cells including vascular smooth muscle may generate free radicals if its substrate-L-
cells, myocardial cells and macrophages after arginine has a low concentration. Therefore,
stimulation with cytokines and polysaccha- at low concentrations of L-arginine, blockers
rides.43 Maintained production of NO by of NO-synthesis may reduce free radical gen-
eNOS may be protective, whereas overproduc- eration by eNOS itself.45
tion of NO by activated iNOS may induce The preservation of eNOS activity after
tissue damage. In a former study,44 we observed L-arginine treatment may be also an explana-
The NO-Donor L-Arginine Reduces the Reperfusion Injury after Heart Transplantation 67

tion for the persisting beneficial effects of NO- 3. Galinanes M, Saldanha C, Kato H et al. Vas-
synthesis on endothelial function after 24 cular and contractile function and tissue me-
tabolites after prolonged hypothermic is-
hours. A rapid infusion of L-arginine during chemia and reperfusion: Comparison of
the first minutes of reperfusion may not only single- versus multi-dose infusions with two
increase NO-synthesis by increasing concen- cardioplegic solutions in blood-perfused neo-
trations of its substrate but by preserving natal pig hearts. J Mol Cell Cardiol 1995;
27:1915-30.
eNOS activity. Even if no differences were 4. Cocks TM, Angus JA, Campbell GR. Release
found between the groups regarding eNOS and properties of endothelium-derived relax-
expression by semiquantitative immunohistol- ing factor (EDRF) from endothelial cells in
ogy, preserved eNOS activity during the early culture. J Cell Physiol 1985; 123:310-20.
reperfusion phase may be responsible for bet- 5. Schulz R, Nava E, Moncada S. Introduction
and potential relevance of a Ca2+-indepen-
ter endothelial function after 24 hours of dent nitric oxide synthase in the myocardium.
reperfusion. Schnabel et al46 showed in a recent Br J Pharmacol 1992; 105:575-80.
ultrastructural study in human transplant 6. Schulz R, Smith JA, Lewis MJ et al. Nitric
biopsy specimens that while myocyte ultra- oxide synthase in cultured endocardial cells
of the pig. Br J Pharmacol 1991; 104:21-24.
structural integrity recovers within 60 minutes 7. Kelm M, Schrader J. Control of coronary
of reperfusion, ultrastructural regeneration of tone by nitric oxide. Circ Res 1990; 66:
the endothelium lasts days up to one week. 1561-75.
Under these aspects, endothelial protection by 8. Pohl U, Lamontagne D. Impaired tissue per-
substituting NO during reperfusion may be fusion after inhibition of endothelium derived
nitric oxide. Basic Res Cardiol 1991; 86:
essential for preserving endothelial function. (Suppl 2) 97-105.
In summary, substitution of NO-donors 9. Lefer AM, Tsao PS, Lefer DJ et al. Role of
or -precursors presents a new concept of endothelial dysfunction in the pathogenesis of
cardioprotection against ischemia-reperfusion reperfusion injury after myocardial ischemia.
FASEB J 1991; 5:2029-34.
injury during cardiac surgery in two terms. 10. Kurose I, Wolf R, Grisham MB et al. Modu-
First, NO-donors act beneficially primarily lation of ischemia/reperfusion-induced mi-
during the reperfusion phase14,16 especially in crovascular dysfunction by nitric oxide. Circ
case of hypothermic ischemia. In contrast to Res 1994; 74:376-82.
other cardioprotection concepts which focus 11. Pabla R, Buda AJ, Flynn DM et al. Intra-
coronary nitric oxide improves postischemic
on ischemic preservation, the use of NO- coronary blood flow and myocardial contrac-
donors aims to reduce reperfusion injury. Sec- tile function. Am J Physiol 1995; 269:
ond, this concept provides a complex integra- H1113-1121.
tive cardioprotection at both the myocardial 12. Adams MR, Jessup W, Hailstones D et al.
L-arginine reduces human monocyte adhesion
and the vascular level. The latter seems to be to vascular endothelium and endothelial ex-
very important, since intact vascular function pression of cell adhesion molecules. Circula-
may play a key role in adequate myocardial tion 1997; 95:662-8.
recovery during reperfusion. 13. Siegfried MR, Erhardt J, Rider T et al.
Cardioprotection and attenuation of endo-
thelial dysfunction by organic nitric oxide
References donors in myocardial ischemia reperfusion. J
1. Argano V, Galinanes M, Edmondson S. Pharm Exp Ther 1992; 260:668-75.
Effects of cardioplegia on vascular function 14. Schulz R, Wambolt R. Inhibition of nitric
and the ‘no reflow’ phenomenon after is- oxide synthesis protects the isolated working
chemia and reperfusion: Studies in the iso- rabbit heart from ischemia-reperfusion injury
lated blood perfused rat heart. J Thorac Cardiovasc Res 1995; 30:432-9.
Cardiovasc Surg 1996; 111:432-42. 15. Naseem SA, Kontos MC, Rao PS et al. Sus-
2. Murphy CO, Pan-Chih, Gott JP et al. Coro- tained Inhibition of nitric oxide by NG-ni-
nary microvascular reactivity after ischemic tro-L-arginine improves myocardial function
cold storage and reperfusion. Ann Thorac following ischemia/reperfusion in isolated
Surg 1997; 63:20-7. perfused rat heart. J Mol Cell Cardiol 1995;
27:419-26.
68 Ischemia-Reperfusion Injury in Cardiac Surgery

16. Depré C, Vanoverschelde JL, Goudemant JF 31. Pabla R, Buda AJ, Flynn DM et al. Nitric
et al. Protection against ischemic injury by oxide attenuates neutrophil-mediated contrac-
nonvasoactive concentrations of nitric oxide tile dysfunction after ischemia and reper-
synthase inhibitors in the perfused rabbit fusion. Circ Res 1996; 78:65-72.
heart. Circulation 1995; 92:1911-18. 32. Gauthier TW, Davenpeck KL, Lefer AM.
17. Aukland K, Bower BF, Berliner RW. Mea- Nitric oxide attenuates leukocyte-endothelial
surement of local blood flow with hydrogen interaction via P-selectin in splanchnic is-
gas. Circ Res 1964; 14:164-87. chemia reperfusion. Am J Physiol 1994; 267
18. Hamano K, Ohmi M, Esato K et al. Myo- G562-8.
cardial tissue blood flow in allotransplanted 33. Gryglewski RJ, Palmer RMJ, Moncada S.
rat heart with a special reference to acute re- Superoxide anion is involved in the break-
jection. J Heart Transplant 1989; 8:48-52. down of endothelium-derived relaxing factor.
19. Hiramatsu T, Forbess JM, Miura T et al. Nature 1986; 320:454-6.
Effects of L-arginine and L-nitro-arginine- 34. Rubanyi GM, Vanhoutte PM. Superoxide
methyl ester on recovery of neonatal lamb anions and hypoxia inactivate endothelium-
hearts after cold ischemia. J Thorac Cardio- derived relaxing factor. Am J Physiol 1986;
vasc Surg 1995; 109:81-7. 250:H822-7.
20. Furchgott RF. Role of endothelium in re- 35. Rubanyi GM, Ho EH, Cantor EH et al.
sponse of vascular smooth muscle. Circ Res Cytoprotective function of nitric oxide: In-
1983; 53:557-73. activation of superoxide radicals produces by
21. Amezcua JL, Palmer RMJ, De Souza BM et human leukocytes. Biochem Biophys Res
al. Nitric oxide synthesized from L-arginine Commun 1991; 181:1392-7.
regulates vascular tone in the coronary circu- 36. Patel VC, Yellon DM, Singh KJ et al. Inhi-
lation of the rabbit. Br J Pharmacol 1990; bition of nitric oxide limits infarct size in the
97:1119-24. in situ rabbit heart. Biochem Biophys Res
22. Moncada S, Palmer RMJ, Higgs EA. Nitric Commun 1993; 194:234-8.
oxide: Physiology, pathophysiology and phar- 37. Craven PA, DeRubertis FR. Restoration of
macology. Pharmacol Rev 1991; 43:109-42. the responsiveness of purified guanylate cy-
23. Amrani M, Chester AH, Jakyakumar CJ et clase to nitrosoguanidine, nitric oxide and
al. L-arginine reverses low coronary reflow related activators by heme and heme proteins:
and enhances postischemic recovery of car- Evidence for the involvement of the paramag-
diac mechanical function. Cardiovasc Res netic nitrosyl-heme complex in enzyme acti-
1995; 30:200-4. vation. J Biol Chem 1978; 253:8433-43.
24. Engelman DT, Watanabe M, Maulik N et 38. Beckman JS, Ischiropoulos H, Zhu L et al.
al. L-arginine reduces endothelial inflamma- Kinetics of SOD and iron catalyzed nitration
tion and myocardial stunning during is- of phenolics by peroxynitrite. Arch Biochem
chemia/reperfusion. Ann Thorac Surg 1995; Biophys 1992; 298:438-45.
60:1275-81. 39. Maulik N, Engelmann DT, Watanabe M et
25. Pinsky DJ, Oz MC, Koga S et al. Cardiac al. Nitric oxide signaling in ischemic heart.
preservation is enhanced in a heterotopic rat Cardiovasc Res 1995; 30:593-601.
transplant model by supplementing the ni- 40. Szabó G, Bährle S, Dengler TJ et al. Reduc-
tric oxide pathway. J Clin Invest 1994; tion of reperfusion injury after heart trans-
93:2291-97. plantation by the nitric oxide donor L-argin-
26. Kubes P, Suzuki M, Granger DN. Nitric ine. Langebecks Arch Chir I Forumband
oxide: An endogenous modulator of leuko- 1997:7-10.
cyte adhesion. Proc Natl Acad Sci USA. 41. Beresewitz A, Karwatowska-Prokopzuk E,
1991;88:4651-5. Lewartowski B et al. A protective role of ni-
27. Bath PMW, MacGregor GA. Spontaneous tric oxide in isolated ischemic/reperfused rat
nitric oxide donors inhibit monocyte chemo- heart. Cardiovasc Res 1995; 30:1001-8.
taxis and increase intracellular cGMP concen- 42. Takeuchi K, McGowan FX, Danh HC et al.
tration. J Vasc Res 1992; 29:170. Direct detrimental effects of L-arginine upon
28. Moilanen E, Vuorinen P, Kankaareranta H ischemia-reperfusion injury to the myocar-
et al. NO donors inhibit human neutrophil dium. J Mol Cell Cardiol 1995; 27:1405-14.
activation in vivo [Abstract], J Vasc Res 1992; 43. Ursell PC, Mayes M. Anatomic distribution
29:170. of nitric oxide synthase in the heart. Int J
29. Cooke JP, Tsao PS. Cytoprotective effects of Cardiol 1995; 50:217-23.
nitric oxide Circulation 1993; 88:2451-4. 44. Szabó G, Bährle S, Dengler TJ et al. L-argi-
30. MA XL, Weyrich AS, Lefer DJ et al. Dimin- nine reduces reperfusion injury and coronary
ished basal nitric oxide release after myocar- endothelial dysfunction in a rat heart trans-
dial ischemia and reperfusion promotes neu- plantation model. [Abstract] J Mol Cell
trophil adherence to coronary endothelium Cardiol 1997.
Circ Res 1993; 72:403-12.
The NO-Donor L-Arginine Reduces the Reperfusion Injury after Heart Transplantation 69

45. Wang QD, Morcos E, Wiklund P et al. L- 46. Schnabel PA, Lange R, Amann K et al. Ul-
arginine enhances functional recovery and trastructural integrity predicts the incidence
Ca2+-dependent nitric oxide synthase activity of acute rejection during the first year after
after ischemia and reperfusion in the heart. J HTX. [Abstract] J Heart Lung Transplant
Cardiovasc Pharmacol 1997; 29:291-6. 1997; 16:S45.
CHAPTER 6

Sodium-Proton Exchange
Inhibition as a Novel Strategy
for Myocardial Protection
Willem Flameng and Wolfgang Scholz

O
ngoing developments in cardiac sodium-proton exchange inhibitor with good
surgery, like surgery on the beating, tolerability and kinetic properties was chosen.
warm heart in minimal invasive First of all we will describe more in detail
coronary bypass grafting, the increasing inci- the theoretical background of sodium-proton
dence of cardiac surgery in the elderly as well exchange inhibition in myocardial ischemia-
as in neonates and the increasing number of reperfusion. Then we will provide experimen-
complex interventions, imply a continuous tal evidence for the protective properties of
request for better myocardial protective tech- inhibiting this exchange system during and
niques. The common challenge for these tech- after ischemia.
niques however remains the damaging effects The drug HOE 642 however was never
of acute myocardial ischemia. Because the tested clinically so that a preliminary clinical
sequence of events during acute myocardial trial was mandatory. We performed a small
ischemia had been the subject of many stud- prospective randomized double blind study,
ies in the past, it became more and more clear first of all to assess safety and secondly to search
that a disturbance of cellular ionic homeosta- for some efficacy in terms of improved recov-
sis is the initial trigger of the ultimate damage. ery of function after complex cardiac surgery
One of the most important early mecha- in man. We will present these data here, bear-
nisms of ionic disturbances is the activation ing clearly in mind that efficacy of sodium-
of the sodium-proton exchange system.1 The proton exchange inhibition in the clinical set-
inhibition of this system during ischemia and ting still needs to be proven.
reperfusion diminishes both Na+ and Ca++
overload and potentially prevents or delays Sodium-Proton Exchange
myocardial damage.2-9 Inhibition in Cardiac Ischemia-
In this chapter, the idea to use a selective
inhibition of the sodium-proton exchange sys- Reperfusion
tem is propagated as a novel approach for pro- In the last decade it has become clear, that
tection against ischemia-induced myocardial most pathophysiological processes in cardiac
damage. For this purpose, HOE642 (4-iso- ischemia and reperfusion are connected to a
propyl-3-methylsulfonyl-benzoylguanidin- derangement of cellular ion homeostasis. The
methanesulfonat) (Hoechst, Frankfurt, characterization of various ion transport sys-
Germany), a new, selective, and potent tems has led to the identification of three
effects, which seem to play key roles in the

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Sodium-Proton Exchange Inhibition as a Novel Strategy for Myocardial Protection 71

impairment of ischemic and reperfused tissue. There are at present at least five different
These interdependent effects are decreased NHE subtypes (NHE 1-5).14,15 NHE inhibi-
intracellular pH (pHi), intracellular Na+ (Na+i) tors as amiloride derivatives, as well as
overload and intracellular Ca ++ (Ca ++ i ) HOE 694 and HOE 642 (cariporide) are to
overload. different degrees preferential inhibitors of the
The interdependence between intracellu- NHE subtype 1, the so called housekeeper,
lar acidosis and elevated Na+i and Ca++i con- which seems to be predominant in cardiac tis-
centrations is based on the activity of certain sue. Whenever the NHE is mentioned in the
ion transport systems. Increased activity of the following, the involvement of this subtype is
Na+ coupled Cl–/HCO3– exchange and an implicated.
excessive activation of Na +/H + exchange
(NHE) by a decrease of pHi lead to a signifi- Effects on Intracellular Na+
cant elevation of Na+ influx into the endan- An increase of Na+i is well documented in
gered tissue. As long as sufficient ATP is avail- “chemically ischemic” or hypoxic myocardial
able, the intruding Na+ ions can be transported cells and in ischemic or hypoxic hearts. It has
against the Na+ gradient into the extracellular become clear, that the increase of Na+i results
space by the Na+/K+ ATPase. But eventually, from an imbalance between Na+ influx and
decreasing energy stores and elevated Na+ Na+ extrusion in ischemic cells. On the one
influx result in a marked increase of the con- hand, Na+ influx is increased by an activation
centration of Na+i. Since intracellular Na+ and of pH regulating ion transport systems as the
Ca++ concentrations are linked by a 3Na+/Ca++ NHE and possibly the Na+-HCO3– symport
exchange, the elevation of Na+i finally causes and the Na+ dependent Cl-/HCO3– exchange.
intracellular Ca++ overload, which induces car- On the other hand, the extrusion of Na+ by
diac arrhythmias,10,11 contracture, stunning the Na+/K+ ATPase decreases according to
and necrosis. energy depletion. It has been claimed that the
Already in 1984 it has been suggested by NHE is responsible for a substantial part of
Frelin and co-workers,12 that the NHE could Na+ influx under these conditions, while
play an important role in this chain of patho- another part of Na+ influx goes through the
physiological events. While the regulation of HCO3– dependent systems and Na+ channels.
pHi seems to be dominated by HCO3– depen- Recent results indicate that the balance be-
dent systems under normal conditions, it has tween NHE activity and Na+-HCO3– symport
been claimed that the NHE causes a major activity seems influenced by alpha adrenergic
part of the Na+ influx in cardiac ischemia and receptors.16 The release of alpha adrenergic
reperfusion. More recently, this point of view agents, as it occurs in myocardial ischemia, is
has been supported by studies showing a sig- expected to shift transport activity towards the
nificant reduction of Na+ and Ca++ overload NHE system. This would mean that Na+ influx
and impressive cardioprotective effects with through the NHE might be even more im-
inhibitors of the Na+/H+ exchanger.9,13 Alto- portant in the living organism than in isolated
gether this points to a crucial involvement of heart models, which are removed from their
this ion transport system in the mechanisms normal systemic neuronal and hormonal
of ischemic and reperfusion injuries. influences.
In the following, this chapter surveys the Whatever the balance between different
observed effects of NHE inhibitors on intrac- Na+ transport systems in the ischemic myo-
ellular Na+ (Na+i), Ca++ (Ca++i), pH (pHi) and cardium might be, it has been demonstrated
metabolism in cardiac tissue, as well as on that Na+ overload is reduced by NHE inhibi-
ischemic and reperfusion injuries. Further- tors. Observations made in hypoxic or “chemi-
more it tries to evaluate the potential clinical cally ischemic” myocardial cells have been con-
use of specific NHE inhibitors in indications firmed by a growing number of Na+ NMR
related to cardiac ischemia and reperfusion. measurements in ischemic hearts.
72 Ischemia-Reperfusion Injury in Cardiac Surgery

While this effect of NHE inhibitors in car- These detrimental Ca++ oscillations can be
diac ischemia seems quite clear, it is more dif- reduced by NHE inhibitors.17
ficult to judge their influence on Na+i during
reperfusion. In reperfusion, after a transient Effects on Intracellular pH
increase, Na+i rapidly decreases, possibly as fast and Ischemic Metabolism
as the cells begin to recover from energy deple-
In ischemia, anaerobic metabolism and
tion. The rate of Na+i decrease depends on the
hydrolysis of ATP cause an accumulation of
relation between Na+ influx and Na+ extrusion,
intracellular protons, and thus, an activation
mainly by Na+/K+ ATPase. Since the NHE is
of pH regulating ion transport systems as the
excessively activated in reperfusion, it might
NHE. In lack of proton clearance the extra-
be expected that NHE inhibitors would
cellular pH (pHo) tends to drop in parallel with
facilitate the recovery from Na+i overload. Up
pHi in the ischemic tissue. It has been dem-
to now, this has been shown in isolated myo-
onstrated that NHE activity is decreased by
cardial cells under rather artificial conditions
extracellular acidosis. At a pHo of about 6.3,
and in the reperfused heart. In general it has
which can be reached within 10-15 min in
been claimed that protective effects of NHE
ischemic hearts, the NHE seems to be quies-
inhibitors in cardiac reperfusion result from a
cent. The activity of the NHE in myocardial
reduction of Na+i overload.
ischemia might be limited to this first period.
Effects on Intracellular Ca++ Since protons are extruded by the active
It is the connection between Na+i levels and NHE, it could be assumed, that the decrease
++
Ca i levels which is thought to make Na+i of pHi was accelerated by NHE inhibitors.
overload harmful for cardiac tissue. In cardiac This has not, as yet, been confirmed by NMR
cells, Ca++i is significantly affected by the studies in isolated hearts. In general the drop
activity of a 3Na + /Ca ++ exchanger. This of pHi was not influenced, or only slightly
exchanger depends on the Na+ gradient to influenced by NHE inhibitors. However, it has
extrude Ca++ from the cell. If this gradient is been demonstrated that in the presence of the
diminished by increasing Na+i, the 3Na+/Ca++ !-1 agonist phenylephrine NHE inhibition
exchanger fails and Ca++i is going to rise. It caused a faster drop of pHi in isolated rat
has been shown in numerous studies with hearts. Since there is a massive release of cat-
myocardial cells and isolated hearts that in echolamines in cardiac ischemia/reperfusion
ischemia the increase of Na+i is paralleled by in animals and humans, it might well be that
an increase Ca++i. As it would be expected, not this study was more significant for the situa-
only Na+i overload but also ischemic Ca++i tion in the living organism, than other experi-
overload is substantially reduced by NHE ments with isolated hearts which were lacking
inhibitors. catecholamins in the perfusion solution.
It has been observed in myocardial cells that In spite of a reduction of anaerobic me-
there is a rapid and transient decrease of free tabolism by NHE inhibitors, high energy
Ca++i at the onset of reperfusion. It is assumed phosphates (HEP) have been found conserved
that this fast decrease is due to an uptake of or unchanged in ischemic cardiac tissue. This
Ca++ ions in intracellular stores. The overall strongly suggests, that the consumption of
cellular Ca++ content is reduced more slowly HEP has been decreased. NHE inhibitors
via the 3Na+/Ca++ exchanger after restoration might cause this effect by diminishing is-
of the Na+ gradient. If the restoration of the chemic Na+i overload and thereby reducing
gradient fails because of an imbalance between Na+/K+ATPase activity.
Na+ influx and Na+ extrusion, Ca++i stores are In cardiac reperfusion, a rapid restoration
kept in an overloaded unstable state. They of pHi to normal values has been observed.
release Ca++ ions and cause repetitive Ca++ os- Together with elevated Ca++i or with Ca++
cillations, which finally kill the reperfused cells oscillations this fast increase of pHi might be
the major cause of reperfusion damage. Since
Sodium-Proton Exchange Inhibition as a Novel Strategy for Myocardial Protection 73

the coupling of Ca++ ions to contractile ele- in rat hearts. 20 In these experiments, the
ments and other Ca++ induced reactions is sus- ischemic insult was so mild, that there was
pended under acidotic conditions, elevated hardly any decrease of ATP and pHi and con-
Ca++i becomes especially serious when pHi is sequently no activation of the exchanger. Duff
restored. The fate of reperfused myocardium and coworkers21 did not find antiarrhythmic
might be decided by the balance between effects of EIPA in a postinfarction model in
decreasing Ca++i and increasing pHi. It seems dogs under conditions where the exchanger
quite clear that the crucial period affecting the should be quiescent.
ultimate course of these events is within the However, in acute ischemia and reperfusion
first minutes or even seconds of reperfusion. where the NHE is excessively activated, it
Acidic reperfusion solutions showed marked seems that all the typical injuries as arrhyth-
protective effects during the first two minutes mias, contracture, stunning, ultrastructural
of reperfusion. Thereafter, perfusion at nor- damage and cardiac necrosis are ameliorated
mal pH was no longer harmful.18 or prevented by NHE inhibitors.
The NHE system, which is excessively A prevention of arrhythmias has been
activated in reperfusion, seems to have a sig- observed in isolated hearts and in animals of
nificant part in pH recovery. Measurements different species with amiloride derivatives,
of pH i with fluorescent dyes in isolated HOE 694 and cariporide. Ventricular fibril-
myocytes have shown that pH recovery is lation, which seems connected to Ca++i over-
delayed by NHE inhibitors. In most NMR load, can be abolished by NHE inhibitors in
studies there are rather large intervals of two ischemia and reperfusion. This has also been
or three minutes between pH measurements shown in pigs, which are known to be highly
which are not suitable to evaluate rapid pH vulnerable to rhythm disturbances and sud-
changes during reperfusion. Nevertheless, den cardiac death.
there are studies showing that pH recovery is Numerous investigators report a substan-
slowed down by NHE inhibitors in reperfused tial reduction of ischemic and postischemic
hearts. It is probable, that this effect of NHE contracture, an improvement of cardiac per-
inhibitors adds to the protective reduction of formance, and an amelioration of myocardial
Ca++i overload in ischemia and reperfusion, by stunning caused by NHE inhibitors. In sev-
making elevated Ca++i less harmful. eral studies, a corresponding prevention of
ultrastructural cellular damage was also
Cardioprotective Effects observed. Other experiments show a dimin-
The reduction of Ca++i overload and the ished release of intracellular enzymes in car-
desensitization of cardiac cells to elevated Ca++i diac ischemia and reperfusion, indicating a re-
are paralleled by outstanding cardioprotective duction of cellular necrosis. A very substantial
effects of NHE inhibitors. These have been reduction of infarct size has been demonstrated
demonstrated with amiloride and numerous in rats, rabbits and pigs.
derivatives including ethylisopropyl amiloride For some time, there has been some con-
(EIPA), dimethyl amiloride (DMA), methyl- troversy as to whether cardioprotection by
isobutyl amiloride (MIBA), hexamethylene NHE inhibitors was limited to reperfusion or
amiloride (HMA) and with the more specific would also occur under ischemic conditions
NHE 1 inhibitors HOE 694 8 and cari- where the NHE might be blocked by low pHo.
poride.19 There are only a few studies which It has become clear, however, that the reported
did not demonstrate protective effects of NHE reduction of ischemic Ca++i overload is accom-
inhibitors. When looked at in detail, it can be panied by a prevention of arrhythmias, con-
seen that they were possibly performed under tracture and myocardial damage during
conditions where the NHE system was not ischemia. It has been shown that there are pro-
significantly activated. Imai and coworkers did tective effects of NHE-inhibitors, if the com-
not see any effect of EIPA in low flow ischemia pound is given only during reperfusion.
74 Ischemia-Reperfusion Injury in Cardiac Surgery

In our laboratory a study was performed Myocardial stiffness is known to be influ-


on isolated blood perfused rabbit hearts7 that enced by myocardial contracture and/or
gives an example for the full scope of edema.23-28 In this study, significantly differ-
cardioprotection caused by NHE inhibition ent stiffness coefficients without difference in
in ischemia/reperfusion. In this study, the myocardial water content among the groups
hearts were subjected to 45min of warm suggest that the improvement in myocardial
ischemia followed by 1h of reperfusion. They compliance in the treated groups is mainly
served either as controls or were treated with affected by myocardial contracture.
the NHE-1 inhibitor HOE 694 during All groups showed successful weaning from
ischemia and reperfusion or only during CPB without inotropic support such as
reperfusion. The pretreatment prevented the dopamine and/or dobutamine and well-pre-
contracture which was observed in controls served ultrastructure without irreversible
nearly totally and markedly improved the per- damage.
formance of the hearts on reperfusion. The In a subsequent experimental study in dogs
reperfusion treatment reduced contracture on we extended the donor heart storage time to
reperfusion and slightly improved the perfor- 24 hours (Flameng et al, unpublished results).
mance of the rabbit hearts (see Fig. 6.1). In Then the hearts were orthotopically trans-
addition, in this study, biopsies were taken planted and reperfused for one hour on CPB.
from the hearts and evaluated by electron mi- Pressure volume relations were studied dur-
croscopy. The results showed that pretreatment ing CPB using a balloon inserted in the left
with the NHE inhibitor nearly totally pre- ventricle. It was found that, when the donor
vented ultrastructural damage, accumulation as well as the recipient was treated with HOE
of Ca++ in the mitochondria and mitochon- 642, myocardial compliance was significantly
drial damage. Treatment on reperfusion only improved and ultrastructure of the myocar-
was still protective, but not as effective as pre- dium remained intact. All hearts stored cold
treatment. for 24 hours after hyperkalemic cardioplegic
In another experimental study from our arrest could be weaned from CPB with stable
laboratory,22 we assessed the effects of HOE hemodynamics and a normal cardiac index,
642, on myocardial function after transplan- provided donor and recipient were treated with
tation of canine brain-dead and nonbrain-dead the sodium proton exchange inhibitor (HOE
donor hearts preserved for 4 hours. Four 642 at a dose of 2 mg/kg body weight). Com-
groups were studied: brain-dead donors; pliance, myocardial structure and functional
nonbrain-dead donors; brain-dead donors and outcome after transplantation were signifi-
recipients treated with HOE 642 (2 mg/kg); cantly better than those in control experiments
and treated nonbrain-dead donors and recipi- without drug treatment.
ents. Donor hearts were stored in hyperkalemic We concluded that the use of this inhibi-
cardioplegic solution. At the end of 60 min- tor might improve the current myocardial pres-
utes reperfusion after transplantation, pressure- ervation technique for transplantation.
volume curves were constructed. Biopsies were
analyzed histologically and ultrastructurally. Preliminary Clinical Experience
Afterwards, weaning from CPB was with Sodium-Proton Exchange
accomplished.
When both donor and recipient are treated Inhibition
with this inhibitor both in the brain death and In this pilot study, a relatively small num-
nonbrain death group, significantly lower ber of patients is included. Nevertheless the
myocardial stiffness is shown after transplan- group of patients represents a rather uniform
tation than in the untreated group (see pathology : all patients had a combination of
Fig. 6.2). However, there were no significant aortic valve disease and coronary artery dis-
differences for the LV systolic performance and ease of such an extent that surgical correction
myocardial water content among the groups. is required. This implies a combination of left
Sodium-Proton Exchange Inhibition as a Novel Strategy for Myocardial Protection 75

Fig. 6.1. Line plot shows effect of (3-methylsulfonyl-4-piperidinobenzoyl)guanidine methanesulfonate (HOE


694) on left ventricular end-diastolic pressure (LVEDP) during 45 minutes of ischemia and 1 hour of reperfusion.
LVEDP is plotted as a function of time. Zero time is the beginning of reperfusion. Bars represent SEM. *p < .05,
**p < .01, ***p < .001 control vs HOE 694 pretreatment, +p < .05, ++ p < .01 control vs HOE 694 reperfusion.
This phenomenon is largely suppressed by HOE 694, even when given only during reperfusion. (Reprinted with
permission from: Circulation 1994; 89:2787-2798).

Fig. 6.2. LV stiffness coefficients (a); There were significantly different SC’s between group 1 and group 3 (0.127
± 0.006 vs 0.072 ± 0.016; p = 0.02), and between group 2 and group 4 (0.152 ± 0.013 vs 0.096 ± 0.019; p = 0.015).
Data are means with SEM as error bar (Reprinted with permission from: Cardiovascular Surgery 1998; (6)67-75.
76 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 6.3. Effect of HOE 642 on the need for positive inotropic support after weaning from cardiopulmonary
bypass. The right panel represents the median values of the total dose of dopamine (in mg) given during the first
36 hours after weaning and during the whole hospital stay. In the left panel the total dose of positive inotropic
medication is represented during these time intervals. (ns: p > 0.05, placebo versus HOE 642).

ventricular hypertrophy and impaired myo- From the point of safety assessment, the
cardial blood supply. On top of this cardiac frequency of possibly treatment related adverse
pathology, a period of aortic cross clamping effects until 8 weeks after discharge was regis-
and consequently of acute ischemia renders tered. The investigators mentioned 5 features
these hearts more vulnerable and make them as possibly related to treatment but none of
a good model to test myocardial protection them however was statistically significantly
techniques. related to treatment (p > 0.05).
Twenty patients undergoing aortic valve Efficacy assessment was done by evaluation
replacement combined with coronary artery of hemodynamic parameters during weaning
bypass grafting were included in this prospec- from cardiopulmonary bypass, parameters of
tive randomized double blind study. The myocardial damage, clinical outcome and the
patients were either pretreated with HOE 642 need for positive inotropic support.
(40 mg I.V. over 10 minutes) or placebo in- Hemodynamic parameters were studied
travenously before going on cardiopulmonary 5 minutes before starting cardiopulmonary
bypass. The study end-points were 1) safety bypass and every 5 minutes after weaning, up
assessment and 2) efficacy of treatment in to 45 minutes after cessation of bypass. None
terms of improvement of hemodynamics after of these parameters showed any difference
weaning from bypass, the need for positive between placebo and HOE 642 treatment.
inotropic support, release of cardiac enzymes Also transesophageal echocardiography
and clinical outcome. The results of the study revealed no differences in the value for end
were published previously in detail.29 The diastolic and end systolic dimensions between
twenty patients were prospectively randomized groups (p > 0.05). Also pulmonary and sys-
into 2 groups of 10 patients each. They were temic vascular resistance did not differ between
13 males and 7 females. Mean age was 67 years groups (p > 0.05).
with a range of 44-77 years. The dose of positive inotropic medication
was calculated in mg for each drug (dopam-
Sodium-Proton Exchange Inhibition as a Novel Strategy for Myocardial Protection 77

ine, dobutamine or noradrenaline) during the 4. Karmazyn M. Na+/H+ exchange inhibitors


first 36 hours and during the total hospital stay. reverse lactate-induced depression in postis-
chemic ventricular recovery. Br J Pharmacol
The dose of noradrenaline was multiplied by 1993; 108:50-56.
20 to obtain an equal reflection of drug 5. Hata K, Takasago T, Saeki A et al. Stunned
potency when the total dose of positive ino- myocardium after rapid correction of acido-
tropic medication was calculated. The results sis-increased oxygen cost of contractility and
are shown in Figure 6.3. In the right panel the role of the Na+/H+ exchange system. Circ
Res 1994; 74:794-805.
only the median dose of dopamine is given as 6. Harada K, Johnson RG, Grossman W et al.
well for the first 36 hours period as for the Acidemia and hypernatremia enhance postis-
total duration of hospital stay. The need for chemic recovery of excitation-contraction cou-
dopamine is lower in the HOE 642 treated pling. Circ Res 1994; 74:1197-1209.
7. Hendrikx M, Mubagwa K, Verdonck F et al.
group, although statistical significance is not New Na+/H+ exchange inhibitor HOE 694
reached (p > 0.05). The total dose of positive improves postischemic function and high-en-
inotropic drugs (left panel) provides approxi- ergy phosphate resynthesis and reduces Ca++
mately the same distribution, indicating that the overload in isolated perfused rabbit heart.
majority of patients received only dopamine. Circulation 1994; 89:2787-2798.
8. Scholz W, Albus U, Lang J et al. HOE 694,
None of the patients showed any evidence a new Na+/H+ exchange inhibitor and its
of myocardial infarction nor life threatening effects in cardiac ischemia. Br J Pharmacol
arrhythmias during the intra- or postopera- 1993; 109:562-568.
tive phase. The release of CK-MB during the 9. Scholz W, Albus U, Linz W et al. Effects of
Na+/H + exchange inhibitors in cardiac is-
first 24 hours postoperatively is very small and chemia. J Moll Cell Cardiol 1992; 24:
did not differ significantly (p > 0.05) between 731-740.
treatment groups. 10. Coetzee WA, Opie LH. Effects of compo-
There was no hospital mortality in the nents of ischemia and metabolic inhibition
study population. The length of stay on the on delayed afterpolarization in guinea pig
papillary muscle. Circulat Res 1987; 61:
intensive care unit and the hospital stay was 157-165.
not different between the two groups 11. Shen AC, Jennings RB. Myocardial calcium
(p > 0.05). and magnesium in acute ischemic injury. Am
This preliminary study showed that the J Pathol 1972; 67:417-440.
12. Frelin C, Vigne P, Ladzdunski M. The role
drug has no adverse effects at the dose used of the Na+/H+ exchange system in cardiac
and that the need for positive inotropic sup- cells in relation to the control of internal Na+
port was less in the group of patients receiv- concentration. J Biol Chem 1984; 259:
ing the Na+/H+ exchange inhibitor. 8880-8885.
These data suggest that further clinical 13. Anderson SE, Murphy E, Steenbergen C et
al. Na-H exchange in myocardium: effects of
evaluation of this type of pharmacological pro- hypoxia and acidification Na and Ca. Am J
tection is meaningful. Physiol 1990; 259:C940-C948.
14. Orlowsky J, Kandasamy RA, Shull GE. Mo-
References lecular cloning of putative members of the
Na/H exchanger gene family. J Biol Chem
1. Karmazyn M, Moffat MP. Role of Na+/H+
1992; 267:9331-9339.
exchange in cardiac physiology and patho-
15. Klanke CA, Su YR, Callen DF et al. Mo-
physiology; mediation of myocardial reper-
lecular cloning and physical and genetic map-
fusion injury by the pH paradox. Cardiovasc
ping of a novel human Na+/H+ exchanger
Res 1993; 27:915-924.
(NHE5/SLC9A5) to chromosome 16q22.1.
2. Scholz W, Albus U. Na+/H+ exchange and Genomics 1995; 25:615-622.
its inhibition in cardiac ischemia and reper- 16. Lagadic Gossmann D, Vaughn-Johns RD
fusion. Basic Res Cardiol 1993; 88:443-455. Coupling of dual acid extrusion in the guinea
3. Karmazyn M, Ray M, Haist JV. Compara- pig isolated ventricular myocyte to alpha 1
tive effects of Na+/H+ exchange inhibitors and beta adreno receptors. J Physiol London
against cardiac injury produced by ischemia/ 1993; 464:49-73.
reperfusion hypoxia/reoxygenation, and the
calcium paradox. J Cardiovasc Pharmacol
1993; 21:172-178.
78 Ischemia-Reperfusion Injury in Cardiac Surgery

17. Ladilov Y, Siegmund B, Piper HM. Block- 25. Cross CE, Rieben PA, Salisbury PF. Influ-
ade of the Na+/H+ exchanger protects against ence of coronary perfusion and myocardial
reoxygenation induced hypercontracture in edema on pressure-volume diagram of left
Ca ++ overloaded cardiomyocytes. Europ J ventricle. Am J Physiol 1961; 201:102-108.
Physiol 1994; 426:R117. 26. Hsu DT, Weng Z-C, Nicolosi AC et al.
18. Ibuki C, Hearse DJ, Avkiran M. Mechanisms Quantitative effects of myocardial edema on
of antifibrillatory effect of acidic reperfusion: the left ventricular pressure-volume relation.
role of perfusate bicarbonate concentration. Influence of cardioplegia osmolarity over two
Am J Physiol 1993; 264:H783-H790. hours of ischemic arrest. J Thorac Cardiovasc
19. Scholz W, Albus U, Lang HJ et al. Protec- Surg 1993; 106:651-657.
tive effects of HOE 642, a selective sodium- 27. Lazar HL, Haasler GB, Sponitz WD et al.
hydrogen exchange subtype 1 inhibitor, on Compliance, mass, and shape of the canine
cardiac ischaemia and reperfusion. Cardiovas- left ventricle after global ischemia analyzed
cular Research 1995; 29:260-268. with two-dimensional echocardiography. J
20. Imai S, Shi AY, Ishibashi T, Nakazawa N. Surg Res 1985; 39:199-208.
Na+/H+ exchange is not operative under low 28. Amirhamzeh MMR, Jia CX, Starr JP et al.
flow ischemic conditions. J. Mol Cell Cardiol Diastolic function in the heterotopic rat heart
1991; 23:505-517. transplant model-Effect of edema, ischemia,
21. Duff HJ, Lester WM, Rahmberg M. Amil- and rejection. J Thorac Cardiovasc Surg
oride, antiarrhythmic and electrophysiologi- 1994; 108:928-937.
cal activity in the dog. Circulation 1988; 29. Flameng W. Clinical experience with
78:1469-1477. HOE 642. A pilot study. Proceedings
22. Kim Y-I, Herijgers P, Van Lommel A et al. Symposium “Organ Preservation in Heart
Na+/H+ Exchange Inhibition Improves Post- Transplantation”. Berlin: Deutsches
transplant Myocardial Compliance in 4-Hour Herzzentrum 1996; in press.
Stored Donor Hearts. Cardiovascular Surgery
1997; (6):67-75.
23. Bethencourt D, Laks H. Importance of edema
and compliance changes during 24 hours of
preservation of the dog heart. J Thorac
Cardiovasc Surg 1981; 81:440-449.
24. Burt JM, Copeland JG. Myocardial function
after preservation for 24 hours. J Thorac
Cardiovasc Surg 1986; 92:238-246.
CHAPTER 7

Metabolic Support for the Heart


During Ischemia and Reperfusion:
Role of Amino Acids
Heinrich Taegtmeyer and Torsten Doenst

I
n reviewing amino acid metabolism of the of endogenous substrates. These substrates are
heart during ischemia and reperfusion it glycogen and, as will be discussed below, cer-
is important to address a few principles of tain amino acids. The breakdown of glycogen
energy substrate metabolism first. The healthy provides glycolytic substrate for essential cell
human heart has a very high rate of energy functions, especially for support of ion
turnover. Like all living cells, cardiac myocytes pumps.5,6 The role of the amino acids in the
need the energy captured in ATP to power metabolic response to myocardial ischemia7,8
their essential functions. The bulk of it is de- is still not very well defined. There is, how-
rived from oxidative phosphorylation of ADP, ever, intriguing evidence that amino acids
which, in turn, is fueled by oxidation of sub- modify the response to myocardial ischemia
strates such as fatty acids, glucose, glycogen, and improve the recovery of function.9,10 The
lactate, and certain amino acids. The normal biochemical basis for this phenomenon can
human heart, which weighs about 300 g, con- only be understood in the wider context of
sumes about 5 kg of ATP in the course of 24 some of the principles of energy transfer.
hours, i.e., 17 times its own weight!1 Under
normal conditions most of the energy for con- Energy for Contraction: Cycles
traction is derived from the oxidation of long- Improve Efficiency
chain fatty acids,2 but inotropic stimulation
The term “oxidative phosphorylation of
results in the preferential oxidation of glyco-
ADP” is an insufficient description of the pro-
gen.3 The heart oxidizes amino acids only
cess of myocardial energy production. Linked
when their plasma concentrations are very
to oxidative metabolism is not only the pro-
high.4
duction of protons (or reducing equivalents)
Ischemia alters substrate metabolism and
in the citric acid cycle and other oxidative path-
impairs energy transfer. With severe ischemia
ways, but also the maze of highways and by-
fatty acid oxidation ceases, while glucose me-
ways of intermediary metabolism.
tabolism continues. Because heart muscle, like
all living tissues, contains only very small Intermediary metabolism of energy provid-
amounts of ATP and phosphocreatine, this ing substrates forms the link between gene
“energy reserve” is exhausted within seconds expression on the one hand and contractile
after the interruption of blood supply. This function of the heart on the other hand.11
extreme situation is modified by substrate level Energy substrate metabolism is complex and
phosphorylation of ADP from the metabolism highly regulated.12 Heart muscle is capable of

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


80 Ischemia-Reperfusion Injury in Cardiac Surgery

using energy-providing substrates interchange- requires the restoration of moiety-conserved


ably and has therefore been called an “omni- cycles, a process termed “anaplerosis”. The
vore”.13 There is growing recognition for the amino acids glutamate, aspartate, isoleucine,
concepts that the regulation of substrate flux and valine are able to function as anaplerotic
is shared by several of the pathway enzymes14,15 substrates for the citric acid cycle (see below).
and that contractile function is optimal only
when fatty acids and carbohydrates are oxi- Amino Acids: Cycling In
dized simultaneously.2 and Out of Proteins
We have recently drawn attention to the
Although this review emphasizes the me-
importance of moiety-conserved cycles for the
tabolism of amino acids, it is important to re-
efficient transfer of energy in heart muscle.16
member that amino acids are first and fore-
A moiety-conserved cycle is defined as a regu-
most the building blocks for proteins, as amino
larly returning sequence of reactions, in which
acids cycle in and out of proteins (Fig. 7.1).
the concentrations of the key components
The incorporation of specific amino acids into
(moieties) neither increase nor decrease.
specific proteins of the heart is governed by
Amino acids play an important role in either
the transcription and translation of specific
maintaining or restoring moiety-conserved
genes on the chromosomes of the nucleus.
cycles.
While the details of gene expression and pro-
The rhythmic nature of contraction and
tein synthesis are increasingly well understood,
relaxation of heart muscle is a palpable mani-
the same cannot be said for protein degrada-
festation of cyclic processes as integral part of
tion.12 Both processes require energy in the
normal physiology. Cycles can be grouped into
form of ATP18 and oxygen deprivation inhib-
cycles of energy provision (e.g., the circula-
its both protein synthesis and degradation.7
tion itself ) and into cycles of energy consump-
Studies with tracers and inhibitors of either
tion (e.g., the actin-myosin crossbridge for-
protein synthesis or degradation have shown
mation and its release). Many intermediary
that under steady state conditions a large por-
metabolites that serve as vehicles for energy
tion of the amino acids is recycled between
transfer are cycled and recycled as well. Best
synthesis and degradation. Furthermore,
known among them are the intermediates of
branched chain amino acids (e.g., leucine) are
the Krebs citric acid cycle. As it has been
thought to regulate protein turnover.19
pointed out by Krebs himself, the evolution-
Some amino acids are neither synthesized
ary advantage of such cycles is that they make
nor degraded by heart muscle. These com-
efficient use of resources.17 The principle is
pounds are frequently used as tracers for pro-
easily understood if one compares two differ-
tein turnover.19 The rates of protein turnover
ent types of locomotion.16 A bicyclist covers
in the heart are often underestimated. The
nearly four times the distance of a runner for
principle of the “dynamic nature of body con-
the same amount of energy expended. The
stituents”20 is perhaps best illustrated in the
only difference between the two is the inter-
heart, which regenerates itself completely once
position of two wheels (i.e., two moiety-con-
every thirty days.21
served cycles) between the body and the
ground.
Myocardial ischemia results in a collapse Amino Acid Metabolism
of most moiety-conserved cycles, and the heart in Normal Heart
has to rely on less efficient linear pathways for The important role of amino acids in en-
energy production. A frequently quoted ex- ergy substrate metabolism is also often over-
ample is the energy yield of 36 moles of ATP looked. Heart muscle contains the enzymes
per mole of glucose oxidized compared to 2 required for the synthesis and degradation of
moles of ATP per mole of glucose converted most, but not all, amino acids. Oxidation of
to lactate. As we will show below, restoration branched chain amino acids proceeds rapidly
of oxidative metabolism with reperfusion also
Metabolic Support for the Heart During Ischemia and Reperfusion 81

Fig. 7.1. A simple scheme which emphasizes the importance of amino acids as building blocks for myocardial
proteins. Gene transcription in the nucleus is followed by translation of activated amino acids (tRNA-amino acid)
into protein. Sets of three nucleotides (codons) in an mRNA are translated into amino acids in the course of protein
synthesis. Major steps include RNA transcription from DNA, RNA processing in the nucleus (RNA splicing) and
RNA translation on ribosomes. The “dynamic nature of body constituents” is best illustrated by the cycling of
amino acids between protein synthesis and degradation. The cycling of amino acids in and out of proteins requires
biological energy in the form of ATP.

(and in preference to other substrates) when malate-aspartate shuttle involves the concerted
their plasma level is high.22,23 activity of a unidirectional glutamate-aspar-
Furthermore, the amino acids aspartate and tate exchanger and a reversible alpha-ketoglu-
glutamate also constitute an integral part in tarate-malate exchanger.25
the transport of reducing equivalents across The dynamic nature of amino acid metabo-
the inner mitochondrial membrane for the oxi- lism and the turnover of amino acids is
dation of cytosolic NADH by the mitochon- underscored by the participation of amino
drial electron transport chain.24 In the malate- acids as intermediates of many metabolic path-
aspartate shuttle cytosolic NADH is oxidized ways. Experimentally, isotopic enrichment of
by the reduction of oxaloacetate to malate. the amino acid glutamate from 13C-labeled
Malate enters the mitochondrial matrix, where exogenous substrates is used as a “lap counter”
it is oxidized to oxaloacetate, which is tran- for the citric acid cycle26 and sophisticated
saminated with glutamate to form alpha-ke- models have been developed to determine the
toglutarate and aspartate. Aspartate and alpha- origin and fate of specific substrates by NMR
ketoglutarate leave the mitochondrion. spectroscopy.27,28 The same technique lends
Transamination of these metabolites itself to the analysis of the fate of dicarboxy-
regenerates oxaloacetate and glutamate in the acids during ischemia and reperfusion.29
cytosol, and the net effect is the transfer of The main features of myocardial amino
hydrogen ions across the mitochondrial acid metabolism are listed in Table 7.1 and
membrane. Net forward flux through the illustrated in Figure 7.2. First, we draw
82 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 7.1. Main features of myocardial amino acid metabolism

1. Amino acids and myocardial energy metabolism


Source of acetyl-CoA
Precursors of citric acid cycle intermediates
Shuttle of reducing equivalents across the inner mitochondrial membrane
2. Amino acids and myocardial ammonia metabolism
Glutamine synthesis
Glutamate synthesis

Fig. 7.2. Salient features of anaerobic amino acid metabolism in heart muscle. Enhanced rates of glycolysis result
in the accumulation of lactate, alanine, succinate, and CO2, as well as the depletion of glutamate and alpha-
ketoglutarate. Substrate level phosphorylation occurs at two sites by mechanisms indicated on the scheme. See text
for further discussion.

attention to amino acids as intermediates of strates for the fuel of respiration. A case in
myocardial energy metabolism. Secondly, point is the amino acid leucine. Leucine is tran-
we draw attention to amino acids as an saminated, decarboxylated, and further me-
integral part of myocardial ammonia tabolized in heart muscle to yield both acetyl-
metabolism, as it has been demonstrated in CoA and acetoacetyl-CoA. The high Km of
studies using [13N]ammonia, which is avidly branched-chain amino transferase for leucine
extracted by the myocardium and retained as assures that leucine is oxidized only when
glutamine.30 The positron-emitting tracer present in high plasma concentrations
[13N]ammonia is commonly used as tracer for (∋ 2.5 ∝mol/L), as it is the case after a pro-
myocardial blood flow. tein-rich meal. Early studies of cardiac sub-
Amino acids which are metabolized to strate metabolism, using coronary sinus cath-
acetyl-CoA (Fig. 7.2) may become an efficient eterization to measure A-V differences, suggest
source of energy and compete with other sub- that as much as 40% of the fuel for respira-
Metabolic Support for the Heart During Ischemia and Reperfusion 83

tion can be supplied by amino acids.4 Leucine An important aspect of anaerobic amino
oxidation by heart muscle has been demon- acid metabolism is the conversion of glutamate
strated by several investigators.22,31,32 The to succinate.8 As already shown, this pathway
physiologic importance of the control of is linked to substrate-level phosphorylation in
amino acid metabolism by Km seems evident. part of the citric acid cycle. Unlike glycolysis,
Since dietary protein and amino acids cannot glutamate utilization can be augmented by a
be stored in major quantities, amino acids simple mass action effect and this pathway may
ingested in the form of protein are distributed represent a source of energy during O2 depri-
in the blood stream, and degradation of any vation. Sanborn et al38 studied the mechanism
excess amino acids takes precedence over the of glutamate conversion to succinate and
degradation of carbohydrate and fat.33 found that in oxygen deprived papillary
Amino acids which may be metabolized to muscles of the rabbit decarboxylation of alpha-
citric cycle intermediates are, in addition to ketoglutarate continued under hypoxic con-
leucine, alanine, glutamate, aspartate, valine, ditions, whereas complete oxidation of alpha-
and isoleucine (Fig. 7.2). The oxidation of ketoglutarate was inhibited. More recent work
glutamate by heart muscle homogenates and by Lazar et al9 and by Bittle and Shine39 dem-
mitochondria34,35 is thought to be of particu- onstrated the beneficial effect of glutamate on
lar importance. Because heart muscle does not cardiac function when glutamate was added
contain the enzyme glutamate dehydrogenase, to the perfusion medium of reversibly
glutamate is transaminated to alanine and ischemic, reperfused rabbit heart. In an effort
alpha-ketoglutarate in an anaplerotic reaction to quantitate the amount of ATP production
of the citric acid cycle. A similar transamina- from the conversion of glutamate to succinate,
tion process occurs with the conversion of Wiesner et al40 measured succinate production
aspartate to alanine and oxaloacetate. from other amino acids in regional ischemia
in dog hearts. The investigators calculated that
Amino Acid Metabolism glutamate accounted for 20% of the ATP gen-
in Oxygen-Deprived Heart erated during the ischemic period. Although
exogenous supplementation of glutamate leads
Ischemia and reperfusion alter the pattern
to an increased uptake,41 the amount of ATP
of myocardial amino acid metabolism in sev-
production from glutamate metabolism dur-
eral ways. First, the efficient transfer of energy
ing ischemia when glutamate was supple-
through moiety-conserved cycles is replaced
mented is not known.
by less efficient linear pathways. Secondly,
A second aspect of anaerobic amino acid
amino acids are no longer oxidized to acetyl-
metabolism is the conversion of aspartate to
CoA. Third, alanine and succinate accumu-
fumarate. Fumarate may be metabolized to
late, while glutamate is catabolized (Fig. 7.3).
succinate by the enzyme fumarate reductase
This short pathway supports the substrate-level
regenerating FAD+ and ATP in the process.42
phosphorylation of GDP.
However, the concept of anaerobic ATP pro-
When we discovered alanine accumulation
duction through reduction of fumarate to suc-
in ischemic heart muscle, we postulated that
cinate has been questioned based on insignifi-
alanine was, like lactate, a product of anaero-
cant reduction of fumarate during ischemia.43
bic glucose metabolism.36 Indeed, in patients
Other concepts on the mechanism of pro-
with coronary artery disease alanine release
tection against ischemic damage by amino
paralleled the release of lactate and was accom-
acids include the maintenance of glycolytic
panied by increased utilization of glutamate.37
activity during ischemia by regeneration of
We thought of alanine as a marker for
cytosolic NAD+ through the malate aspartate
ischemia, however, quantitatively much less
shuttle or by the reduction of feedback inhi-
pyruvate is converted to alanine than to lac-
bition of glycolysis by lactate since part of the
tate,7 and rigorous testing of this hypothesis
pyruvate is transaminated to alanine and does
has never been carried out.
not enter the lactate pool.44
84 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 7.3. Amino acids as precursors or products of citric acid cycle intermediates. The amino acids glutamate, valine,
isoleucine, and aspartate may be important carbon sources for the replenishment of the citric acid cycle upon
reperfusion after ischemia. See text for further discussion.

All findings indicate that carbohydrate and be observed when the perfusion medium is
amino acid metabolism in oxygen-deprived supplemented before, during, and after
heart muscle are linked through the transami- ischemia39,44,46 or during reperfusion alone.9,46
nation of pyruvate with glutamate and the Most of the studies have focused on aspartate
resultant formation of alanine and succinate. and glutamate, because these substrates are the
Since the conversion of glutamate to succinate key amino acids of the malate-aspartate shuttle,
occurs in the mitochondrial matrix, it appears an important transport mechanism for the
that the matrix enzymes still function during transfer of cytosolic reducing equivalents in
brief periods of oxygen deprivation. As will be to the mitochondrial matrix, and because
discussed further below, the advanced hypoth- glutamate is readily transaminated to alpha-
eses have to be viewed with caution because ketoglutarate, a citric acid cycle intermediate
they depend on proper functioning of the (see “anaplerosis” above).
malate-aspartate shuttle which may be Several concepts have been advanced on
impaired in its activity during ischemia and the mechanism of the protective effects of
reperfusion.45 amino acids and their metabolites during
reperfusion. First, as already described above,
Amino Acids as Substrates maintenance of anaerobic glycolysis and ATP
for Metabolic Support generation through substrate level phospho-
rylation in the formation of succinate reduce
During Reperfusion injury during ischemia thereby improving
There is good experimental evidence that recovery during reperfusion.8,9,40 Secondly, the
amino acids are capable of decreasing the replenishment of the intermediates of the
amount of injury caused by ischemia or malate-aspartate shuttle (e.g., glutamate,
reperfusion or both, although the results are which is depleted during ischemia) during
not all consistent.32 Some of the discrepancies reperfusion promotes oxidative metabolism,
may be explained by the different models used specifically glucose oxidation.46 Third, aspar-
by different investigators. Beneficial effects can tate gives rise to ATP when converted to suc-
Metabolic Support for the Heart During Ischemia and Reperfusion 85

cinate.42 Fourth, fumarate, the integral inter- effect of glutamate may limit the clinical use
mediate in the conversion of aspartate to suc- of this amino acid to the enrichment
cinate during ischemia, may also function as cardioplegia.
free radical scavenger during reperfusion and
contributes to Ca2+ transport.47 The first three Substrate Enhancement
concepts reflect the rationale behind the of Cardioplegic Solutions
addition of aspartate and glutamate to car-
dioplegia (see below). The fourth concept was by Amino Acids
the basis for a study on neonate piglets, where The observation that amino acids reduce
the hearts were arrested for 120 min with ischemic injury in vitro has led to the next
blood cardioplegia supplemented with fuma- logical step, the addition of various amino ac-
rate.47 The investigators found full recovery ids to cardioplegic solutions for the protection
of contractile function during reperfusion in of ischemic heart muscle in vivo. Myocardial
the group with fumarate supplementation utilization of amino acids, especially of
compared to approximately 70% of recovery glutamate, has been demonstrated clinically
of function in the hearts without fumarate in patients during aorto-coronary bypass sur-
addition. gery. 51 Most studies showed evidence for
However, all the suggested mechanisms reduced ischemic injury with amino acid
depend more or less on the exchange of supplementation in cardioplegia (mainly blood
metabolites across the inner mitochondrial cardioplegia).39,47,52,53 Rosenkranz et al54 dem-
membrane through the malate-aspartate onstrated an additive benefit of glutamate and
shuttle. This aspect may render the advanced aspartate supplementation of blood cardiople-
mechanisms only hypothetical, since gia on postreperfusion ventricular function in
Lewandowski et al45 demonstrated that the bi- adults (Fig. 7.4). These findings are consistent
directional part of the malate-aspartate shuttle with the concepts of anaerobic ATP genera-
(malate/alpha-ketoglutarate exchanger) is tion through two different pathways, both
reduced in its activity during ischemia and resulting in the production of succinate (see
reperfusion in rabbit heart. This decreased Fig. 7.3 and the above). However, the impor-
activity of the malate-aspartate shuttle may tance of these pathways has never been dem-
contribute to a persisting proton accumula- onstrated under the conditions tested, and like
tion in the reperfused, “stunned” myocar- in the experimental data in vitro, the results
dium48 and may explain why in our hands of other studies argue against a benefit of
glutamate failed to improve contractile func- amino acids in cardioplegic solutions.55,56
tion during and following ischemia in isolated Perhaps the most striking observations in
rabbit hearts perfused with glucose.32 Thus, the clinical setting of myocardial stunning after
while some experimental results suggest pro- cardioplegia are those of Beyersdorf et al.57 The
tective effects of amino acids against ischemic investigators used glutamate/aspartate-en-
injury, the mechanisms for this protective riched blood cardioplegia during the coronary
effect are not yet understood. An important revascularization procedure in patients with
step in the validation of the above discussed perioperative sudden death. Despite the grim
mechanisms would be the demonstration of prognosis in this group of patients, 78% of
normal or increased activity of the malate-as- them recovered completely and were dis-
partate shuttle in the setting where amino acid charged with significantly improved ejection
supplementation leads to reduction in fractions (Fig. 7.5). Still, the results need to
ischemic injury and improvement of postis- be viewed with caution. The obvious difficul-
chemic function. For the systemic application ties in obtaining a control group for this
of glutamate in patients to improve recovery complex clinical study did not allow conclu-
in postischemic cardiac function, one has to sions whether the excellent outcome was due
bear in mind that high concentrations of to or at least influenced by amino acid
glutamate result in neurotoxicity.49,50 This addition.
86 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 7.4. Benefit of glutamate and aspartate enrichment of blood cardioplegia on postreperfusion ventricular
function in adults. LAP, left atrial pressure; SWI, stroke work index. Reprinted with permission from Rosenkranz
ER, Okamoto F, Buckberg GD et al. Safety of prolonged aortic clamping with blood cardioplegia: Aspartate
enrichment of glutamate-blood cardioplegia in energy-depleted hearts after ischemic and reperfusion injury. J
Thorac Cardiovasc Surg 1986; 91:428-435.

Fig. 7.5. Global ejection fraction (EF) of the left ventricle before (Pre-Op) and after (Post-Op) operation and
during cardiopulmonary resuscitation (CPR). Note a) the moderate reduction of EF before operation and before
intractable ventricular fibrillation occurred (open bar), b) the significant reduction of EF in those patients with
preoperative or postoperative arrest (filled bar), and c) significant improvement (p<0.05) of EF postoperatively
(hatched bar). Reprinted with permission from Beyersdorf F, Kirsh M, Allen BS. Warm glutamate/aspartate-
enriched blood cardioplegic solution for perioperative sudden death. J Thorac Cardiovasc Surg 1992; 104:1141-1147.
Metabolic Support for the Heart During Ischemia and Reperfusion 87

It is possible that the inconsistencies 3. Goodwin GW, Ahmad F, Doenst T et al. En-
observed in the different studies are due to dif- ergy provision from glycogen, glucose and
fatty acids upon adrenergic stimulation of iso-
ferent experimental designs. It appears neces- lated working rat heart. Am J Physiol 1998;
sary to further define indications for amino in press.
acid enrichment of cardioplegic solutions for 4. Bing RJ. The metabolism of the heart.
the use in human heart. A better understand- Harvey Lect 1955; 50:27-70.
5. McDonald T, MacLeod D. Metabolism and
ing of the underlying mechanisms is therefore the electrical activity of ventricular muscle. J
of paramount importance. It should, however, Physiol (Lond) 1973; 229:559-582.
be noted that none of the studies demonstrated 6. Xu KY, Zweier JL, Becker LC. Functional
any detrimental effects of amino acid enrich- coupling between glycolysis and sarcoplasmic
ment of cardioplegia. reticulum Ca2+ transport. Circ Res 1995;
77:88-97.
7. Taegtmeyer H, Ferguson A, Lesch M. Pro-
Conclusions and Outlook tein degradation and amino acid metabolism
Amino acids are the building blocks of in autolyzing rabbit myocardium. Exp Mol
Pathol 1977; 26:52.
myocardial proteins. Certain amino acids are 8. Taegtmeyer H. Metabolic responses to car-
also important intermediates of energy sub- diac hypoxia: Increased production of succi-
strate metabolism in aerobic and anaerobic nate by rabbit papillary muscles. Circ Res
heart. The conversion of glutamate to succi- 1978; 43:808-815.
nate during oxygen deprivation is coupled to 9. Lazar HL, Buckberg GD, Manganaro AJ et
al. Reversal of ischemic damage with amino
substrate-level phosphorylation of GDP to acid substrate enhancement during reper-
GTP. The conversion of aspartate to succinate fusion. Surgery 1980; 88:702-709.
via fumarate reductase has also been suggested 10. Pisarenko O, Solomatina E, Studneva I et al.
as a source of anaerobic energy. Because amino Protective effect of glutamic acid on cardiac
function and metabolism during cardioplegia
acids appear to improve ischemia tolerance in and reperfusion. Basic Res Cardiol 1983;
vitro, solutions fortified with amino acids have 78:534-543.
been successfully employed for cardioplegia 11. Taegtmeyer H, Goodwin GW, Doenst T et
and reperfusion in vivo. Although some reports al. Substrate metabolism as a determinant for
record spectacular results and no adverse effects postischemic functional recovery of the heart.
Am J Cardiol 1997; 80:3A-10A.
of amino acid infusions have been observed, 12. Taegtmeyer H. Energy metabolism of the
the well known neurotoxicity of glutamate heart: From basic concepts to clinical appli-
may limit the unrestricted use of this amino cations. Curr Prob Cardiol 1994; 19:57-116.
acid in patients with ischemic heart disease. 13. Taegtmeyer H, Roberts AFC, Rayne AEG.
Energy metabolism in reperfused rat heart:
Return of function before normalization of
ATP content. J Am Coll Cardiol 1985;
Acknowledgments 6:864-870.
Work from the authors’ laboratory was 14. Kashiwaya Y, Sato K, Tsuchiya N et al. Con-
supported by US Public Health Service trol of glucose utilization in working perfused
rat heart. J Biol Chem 1994; 269(41):25502-
Grant No. RO1-HL43133. Torsten Doenst 25514.
was the recipient of a research fellowship 15. Srere P. Complexities of metabolic regulation.
from the German Research Foundation Trends in Biochem Sci 1994; 19:519-520.
(Deutsche Forschungsgemeinschaft, DFG). 16. Taegtmeyer H, Villalobos DH. Metabolic
support for the postischaemic heart. Lancet
1995; 345:1552-1555.
17. Baldwin JE, Krebs HA. The evolution of
References metabolic cycles. Nature 1981; 291:381-382.
1. Apstein CS, Taegtmeyer H. Glucose-insulin- 18. Lesch M, Taegtmeyer H, Peterson M et al.
potassium in acute myocardial infarction. Cir- Studies on the mechanism of the inhibition
culation 1997; 96:1074-1077. of myocardial protein synthesis during oxy-
2. Taegtmeyer H, Hems R, Krebs HA. Utiliza- gen deprivation. Am J Physiol 1976; 230:
tion of energy providing substrates in the iso- 120-126.
lated working rat heart. Biochem J 1980;
186:701-711.
88 Ischemia-Reperfusion Injury in Cardiac Surgery

19. Morgan H, Rannels D, McKee E. Protein 34. Borst P, Slater E. The oxidation of glutamate
Metabolism of the Heart. In: Berne R, ed. by rat heart sarcosomes. Biochim Biophys
Handbook of Physiology: The Cardiovascu- Acta 1960; 41:170-171.
lar System: The Heart. Washington, D.C.: 35. Krebs H, Bellamy D. The interconversion of
American Physiology Society, 1979:845-871. glutamic acid and aspartic acid in respiring
20. Schoenheimer R. The Dynamic State of Body tissues. Biochem J 1960; 75:523-529.
Constituents. Cambridge, MA: Harvard Uni- 36. Taegtmeyer H, Peterson MB, Ragavan VV et
versity Press, 1942. al. De novo alanine synthesis in isolated oxy-
21. Morgan H, Neely J. Metabolic Regulation gen-deprived rabbit myocardium. J Biol
and Myocardial Function. In: Hurst JW, ed. Chem 1977; 252:5010-5018.
The Heart. 6th ed. New York: McGraw-Hill, 37. Mudge G, Mills R, Taegtmeyer H et al. Al-
1986:85-100. terations of myocardial amino acid metabo-
22. Ichihara K, Neely J, Siehl D et al. Utiliza- lism in chronic ischemic heart disease. J Clin
tion of leucine by working rat heart. Am J Invest 1976; 58:1185-1192.
Physiol 1980; 239:E430-E436. 38. Sanborn T, Gavin W, Berkowitz S et al. Aug-
23. Bing R, Siegel A, Ungar I et al. Metabolism mented conversion of aspartate and glutamate
of the human heart. 2. Studies on fat, ke- to succinate during anoxia in rabbit heart. Am
tone and amino acid metabolism. Am J Med J Physiol 1979; 237:H535-H541.
1954; 16:504-515. 39. Bittl J, Shine K. Protection of ischemic rab-
24. Safer B, Smith C, Williamson J. Control of bit myocardium by glutamic acid. Am J
the transport of reducing equivalents cross the Physiol 1983; 245:H406-H412.
mitochondrial membrane in perfused rat 40. Wiesner R, Deussen A, Borst M et al.
heart. J Mol Cell Cardiol 1971; 2:111-124. Glutamate degradation in the ischemic dog
25. LaNoue K, Schoolwerth A. Metabolite trans- heart: Contribution to anaerobic energy pro-
port in mitochondria. Annu Rev Biochem duction. J Mol Cell Cardiol 1989; 21:49-59.
1979; 48:871-922. 41. Dinkelborg LM, Kinne RKH, Grieshaber
26. Malloy C, Sherry A, Jeffrey F. Carbon flux MK. Transport and metabolism of L-gluta-
through citric acid cycle pathways in perfused mate during oxygenation, anoxia, and reoxy-
heart by 13C NMR spectroscopy. FEBS Lett genation of rat cardiomyocytes. Am J Physiol
1987; 212:58-62. 1996; 270:H1825-H1832.
27. Chance E, Seeholzer S, Kobayashi K et al. 42. Wilson MA, Cascarano J. The energy yield-
Mathematical analysis of isotope labeling in ing oxidation of NADH by fumarate in sub-
the citric acid cycle with applications to 13C mitochondrial particles of rat tissues. Biochim
NMR studies in perfused rat hearts. J Biol Biophys Acta 1970; 216:54-62.
Chem 1983; 258:13785-13794. 43. Peuhkurinen K, Takala T, Nuutinen E et al.
28. Jeffrey F, Diczku V, Sherry A et al. Substrate Tricarboxylic acid cycle metabolites during
selection in the isolated working rat heart: ischemia in isolated perfused rat heart. Am J
Effects of reperfusion, afterload, and concen- Physiol 1983; 244:H281-H288.
tration. Basic Res Cardiol 1995; 90:388-396. 44. Pisarenko OI, Studneva IM, Shulzhenko VS
29. Brainard JR, Hutson JY, Hoekenga DE et al. et al. Substrate accessibility to cytosolic as-
Ordered synthesis and mobilization of glyco- partate aminotransferase improves posthypoxic
gen in the perfused heart. Biochem J 1989; recovery of isolated rat heart. Biochem Mol
28:9766-9772. Medicine 1995; 55:138-148.
30. Schelbert H, Phelps M, Hoffman E. Regional 45. Lewandowski ED, Yu X, LaNoue KF et al.
myocardium blood flow, metabolism, and Altered metabolite exchange between subcel-
function assessed noninvasively with positron lular compartments in intact postischemic
emission tomography. Am J Cardiol 1980; rabbit hearts. Circ Res 1997; 81:165-175.
46:1269-1276. 46. Shug AL, Madsen D, Dobbie R et al. Pro-
31. Tischler ME, Goldberg AL. Leucine degra- tection of mitochondrial and heart function
dation and release of glutamine and alanine by amino acids after ischemia and cardiople-
by adipose tissue. J Biol Chem 1980; 255: gia. Life Sci 1994; 54:557-567.
8074-8081. 47. Pearl JM, Hiramoto J, Laks H et al. Fuma-
32. Taegtmeyer H, Russell R. Glutamate metabo- rate-enriched blood cardioplegia results in
lism in rabbit heart: Augmentation by is- complete functional recovery of immature
chemia and inhibition with acetoacetate. J myocardium. Ann Thorac Surg 1994; 57:
Appl Cardiol 1987; 2:231-249. 1636-1641.
33. Krebs HA. Some aspects of the regulation of
fuel supply in omnivorous animals. Adv En-
zyme Regul 1972; 10:397-420.
Metabolic Support for the Heart During Ischemia and Reperfusion 89

48. Marban E, Kitakaze M, Koretsune Y et al. 54. Rosenkranz ER, Okamoto F, Buckberg GD
Quantification of [Ca2+]i in perfused hearts. et al. Safety of prolonged aortic clamping
Critical evaluation of the 5F-BAPTA and with blood cardioplegia: Aspartate enrichment
nuclear magnetic response method as applied of glutamate-blood cardioplegia in energy-de-
to the study of ischemia and reperfusion. Circ pleted hearts after ischemic and reperfusion
Res 1990; 66:1255-1267. injury. J Thorac Cardiovasc Surg 1986;
49. Bittigau P, Ikonomidou C. Glutamate in neu- 91:428-435.
rologic diseases. J Child Neurol 1997; 12: 55. Asai T, Grossi EA, LeBoutillier MR et al. Re-
471-485. suscitative retrograde blood cardioplegia. Are
50. Olney JW. Brain lesions, obesity, and other amino acids or continuous warm techniques
disturbances in mice treated with monoso- necessary? J Thorac Cardiovasc Surg 1995;
dium glutamate. Science 1969; 164:719-721. 109:242-248.
51. Svedjeholm R, Ekroth R, Joachimsson PO et 56. Crooke GA, Harris LJ, Grossi EA et al. Role
al. Myocardial uptake of amino acids and of amino acids and enhancement. Cardiople-
other substrates in relation to myocardial oxy- gia in routine myocardial protection. J Thorac
gen consumption four hours after cardiac Cardiovasc Surg 1993; 106:497-501.
operations. J Thorac Cardiovasc Surg 1991; 57. Beyersdorf F, Kirsh M, Allen BS. Warm
101:688-694. glutamate/aspartate-enriched blood cardio-
52. Svedjeholm R, Hakanson E, Vanhanen I. Ra- plegic solution for perioperative sudden
tionale for metabolic support with amino ac- death. J Thorac Cardiovasc Surg 1992;
ids and glucose-insulin-potassium (GIK) in 104:1141-1147.
cardiac surgery. Ann Thor Surg 1995; 59
(Suppl 2):S15-S22.
53. Kjellman U, Björk K, Ekroth R et al. Alpha-
ketoglutarate for myocardial protection in
heart surgery. Lancet 1995; 345:552-553.
CHAPTER 8

Metabolic and Antioxidant Support


with Amino Acids
Oleg I. Pisarenko

H
ypothermic hyperkalemic cardiople- extracted more L-glutamate than did hearts
gia is currently the preferred method of individuals with normal coronary arteries.3
of myocardial preservation for the Later enhanced myocardial extraction of
performance of cardiac operations. At present L-13N-Glu in the areas with a reduced blood
research efforts of many laboratories focus on flow was confirmed in human heart by
approaches to enhanced myocardial protection positron-emission tomography.4 Together with
by various pharmacological agents. During the Asp, myocardial Glu content dramatically
past 15 years amino acids are being extensively decreases during periods of ischemia and
studied among other natural metabolites hypoxia. It was shown that catabolism of both
potentially capable to attenuate ischemic and amino acids may be coupled with anaerobic
reperfusion injury. A minor significance of and aerobic energy formation.
amino acids as energy producing substrates is
generally accepted since less than 5% of oxy- Anaerobic Action
gen consumed by the heart is used for their The key enzymes controlling the entry of
oxidation.1 Although amino acids are involved these amino acids into the energy generating
in intracellular metabolism and operating spe- pathways are cytosolic alanine and aspartate
cific transport systems of the plasma mem- aminotransferases, leading to formation of
brane, they do not affect cardiac function un- 2-oxoglutarate and oxaloacetate.5 Oxidation
der normal conditions.2 However, there is a of 2-oxoglutarate to succinate in mitochon-
growing body of evidence that certain of them dria leads to substrate phosphorylation of
may be vital for myocardial function and sur- GDP in the succinic thiokinase reaction and
vival during ischemia/reperfusion stress. In this then formation of ATP by the nucleoside
respect glutamate (Glu), aspartate (Asp), tau- diphosphate kinase. Oxaloacetate can be con-
rine (Tau), branched chain amino acids verted to fumarate in successive reactions cata-
(BCAA), histidine (His) and methionine (Met) lyzed by cytosolic malate dehydrogenase and
seem to be the most important. Mechanisms mitochondrial fumarase. Further NADH-de-
of their action, in conditions relevant to cardiac pendent reduction of fumarate to succinate is
surgery, are briefly discussed in this chapter. coupled with ATP formation in complex 1 of
the respiratory chain. Probably, electron trans-
Glutamate and Aspartate fer from NADH to fumarate occurs from
The first evidence of Glu importance for NADH dehydrogenase through ubiquinone
ischemic myocardium was observed more than to succinate dehydrogenase.6 Both succinate-
20 years ago in clinical studies showing that producing routes may operate with the com-
hearts of patients with coronary artery disease mon mitochondrial NAD+/NADH pair to

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Metabolic and Antioxidant Support with Amino Acids 91

maintain the redox potential of the cell loid cardioplegia.13-16 Moreover, aspartate en-
unchanged.7 Succinate is the end product of richment of Glu-blood cardioplegia further
anaerobic metabolism of Glu and Asp and does improves myocardial protection.17 Beneficial
not undergo further conversions in the absence effects of Glu/Asp-blood cardioplegia are not
of oxygen. confined to adult hearts. Abundant experi-
Glu and Asp may also exert stimulatory mental data obtained by Buckberg and col-
effects on anaerobic glycolysis. Conversion of leagues clearly demonstrate advantages of
Asp to oxaloacetate and then to malate con- amino acid-enriched blood cardioplegia in
sumes cytosolic NADH in the malate dehy- immature hearts.18 Addition of Glu or Asp to
drogenase reaction, thus, relieving glycolytic crystalloid cardioplegic solutions can also pro-
flux through the glyceraldehyde 3-phosphate long the safe limits for cold storage techniques
dehydrogenase.8 Additionally, transamination used in cardiac transplantation.19-21
of glycolytic pyruvate with glutamate may
reduce lactate accumulation in myocardial tis- Aerobic Action
sue and diminish inhibition of glycolysis at Noteworthy, exogenous Glu and Asp pro-
the lactate dehydrogenase step. Intracellular mote recovery of oxidative metabolism after
acidosis favors this reaction, since Km of ala- periods of ischemic arrest restoring depleted
nine aminotransferase for pyruvate decreases intracellular pools of tricarboxylic acid cycle
with decreasing pH.9 Alternatively, transport intermediates and amino acids.11,12,15,19,22
of Glu and malate into mitochondria may Replenishment of these reactants is of critical
stimulate operation of mitochondrial part of importance for operation of the tricarboxylic
the malate-aspartate shuttle. Dismutation of acid cycle23 and malate-aspartate shuttle,
malate to oxaloacetate and succinate in oxy- which transfers reducing equivalents of
gen-deprived rat heart mitochondria7 provides NADH from the cytosol to the respiratory
the formation of reducing equivalents and chain of mitochondria.24 Further, these amino
their prompt removal by the mitochondrial acids take part in binding of ammonia excess
fumarate reductase. 10 According to this in glutamine and asparagine synthesis. Cyto-
hypothesis, dehydration of malate to fumarate toxic effects of ammonia, a by-product of
may serve as an alternative electron acceptor adenine nucleotide degradation, are associated
promoting deinhibition of glycolysis. with the inhibition of isocitrate dehydrogenase
Coupling of Glu and Asp degradation with and activation of mitochondrial pyrophos-
the energy-producing pathways in the cytosol phatase catalyzing degradation of NADPH to
and mitochondria is well-documented in stud- nicotinamide.25 They may impede coordina-
ies with labeled amino acids and intermedi- tion of tricarboxylic acid cycle activity with
ates of the tricarboxylic acid cycle and electron transfer in mitochondrial respiratory
presented in Figure 8.1. Importance of tran- chain.23 Correction of these metabolic disor-
samination, as a necessary step for further ders may reduce the impact of reperfusion
energy production, was justified by elimina- injury after cardiac surgery and, therefore,
tion of the protective effect of Glu by adding incidences of postoperative myocardial stun-
aminooxyacetate, an inhibitor of mitochon- ning. In turn, providing higher cytosolic con-
drial and cytosolic transaminases.11,12 Numer- centrations of ATP and ATP/ADP ratios in
ous animal experiments testify that exogenous compartments close to ion pumps and
Glu and Asp improve myocardial tolerance to myofibrils, Glu and Asp supplementation
ischemia and enhance postischemic functional favors cellular membrane integrity, mainte-
recovery via stimulation of anaerobic and aero- nance of ionic gradients, and removal and
bic energy formation. These observations pro- binding of Ca2+ by myofibrils.26 Recent stud-
vided a solid rationale for the use of these com- ies suggest that supplementing the prime
pounds as beneficial additives to cardioplegic of cardiopulmonary bypass circuit with Glu
solutions. Both amino acids separately enhance and Asp attenuates lipid peroxidation and
the protection afforded by blood and crystal- myocardial dysfunction after reoxygenation.
92 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 8.1. Stimulation of anaerobic energy formation in cytosol (A) and mitochondria (B) by Glu and Asp supple-
mentation. G-3P, glyceraldehyde 3-phosphate; 1,3-DG, 1,3-diphosphoglycerate; Mal, malate; Fum, fumarate; FP,
flavoprotein of the succinate dehydrogenase complex in the oxidized (ox) or reduced (red) form.

This effect may be attributed to amino acid strating superior recovery of high-energy
inhibition of L-arginine uptake by myocardial phosphates, stroke volume index and compli-
cells leading to subsequent reduction of •NO ance after cardioplegic arrest followed by reper-
formation.27 fusion or reoxygenation with Glu and
In agreement with these findings, there are Asp.16-18,28,29 In experimental coronary artery
a number of experimental reports demon- occlusion, the intravenous infusion of Glu and
Metabolic and Antioxidant Support with Amino Acids 93

Asp may reduce infarct size due to improve- metabolism were revealed in patients treated
ment of the myocardial metabolism stimulat- with dopamine because of cardiac failure. They
ing reparative processes.30,31 Additionally, the were associated with an increased Glu myo-
intravenous compositions containing both cardial uptake, a change from lactate release
amino acids exhibit salutary effects on the con- to its uptake, a cessation of ammonia release
tractility of viable remote myocardium in coro- and simultaneous improvement of hemody-
nary occlusion32 implying that metabolic sup- namic performance.39
port of cardiomyocytes can be helpful before
coronary artery bypass grafting. Other Amino Acids
Clinical Experience Histidine
In routine cardiac surgical practice, the The idea for using His as a protective agent
main interest concerning Glu and Asp supple- against myocardial ischemia/reperfusion injury
mentation is focused on enhancement of myo- was developed from several studies demon-
cardial protection during the periods of aortic strating a lesser degradation of Ca2+-ATP
cross-clamping. As a rule, separate or com- activity of sarcoplasmic reticulum, reduction
bined enrichment of blood and crystalloid car- of sarcolemmal Na+-K+-ATPase activity, inhi-
dioplegia with these amino acids improves bition of lipid peroxidation and delayed gen-
postoperative hemodynamic recovery even in esis of arrhythmias in isolated hearts challenged
high-risk coronary patients and enhances with singlet oxygen generation in the presence
restoration of myocardial energy metabolism of this amino acid.40-42 Additionally, His is a
in congenital surgical patients.13,33,34 Anti- well-known scavenger of OH• radicals.43
ischemic effects of Glu were also observed in The use of high His concentrations
the preoperative setting. Thus, intravenous (150-200 mM) in cardioplegic solutions pro-
administration of Glu to patients with coro- vides superior intracellular buffer capacity to
nary artery disease was associated with stimulate anaerobic energy production during
increased tolerance to ischemia in regard to oxygen deprivation. His-buffered hearts dis-
chest pain and electrographic changes during play effective recovery of cardiac function
exercise testing and pacing.35 coincident with more normal pH and PCO2 in
In the early postoperative period, blood- the coronary sinus effluent at the termination
born amino acids and endogenous intracellu- of ischemia.44 The rationale for this type of
lar constituents are appeared to be the prefer- solution is 2-fold. His exerts the maximal buff-
able substrates oxidized by the heart, while ering near the pH of neutrality, since pKa of
uptake of free fatty acids and carbohydrates is its imidasole group is 7.0 at 25oC. In contrast
significantly compromised.36 It is very likely to bicarbonate buffer solutions, the buffering
that under these conditions, augmented myo- capacity of His does not depend upon the par-
cardial consumption of amino acids, and first tial pressure of CO2 which may dramatically
of all Glu, precedes an adequate recovery of change during global myocardial ischemia.
aerobic metabolism. The high fractional Due to these cardioprotective effects His con-
extraction of Glu exhibits a consistent corre- taining Bretschneider’s cardioplegic solutions
lation to its arterial plasma level.37 These have a sound reputation in clinical and experi-
interesting observations suggest that a relative mental practice for long years.45
shortage of myocardial glutamate following
cardiac arrest38 persists during the first hours Branched Chain Amino Acids
after unclamping of the aorta and, therefore, A mixture of leucine, isoleucine and valine
substantiate administration of exogenous Glu added to crystalloid cardioplegic solutions
after operations. Accordingly, positive effects maintains myocardial ATP at higher levels
of postoperative Glu infusion on myocardial during experimental ischemia.46 This effect is
accompanied by a reduced release of adenine
94 Ischemia-Reperfusion Injury in Cardiac Surgery

nucleotide catabolites from the heart and bet- modifier of Ca2+ fluxes through the sarco-
ter sarcolemmal integrity during the early lemma55 and a membrane stabilizer.56
phase of reperfusion.47 The mechanisms of The normal heart maintains Tau content
BCAA action on myocardial energetics remain in cytosol by several selective transport mecha-
obscure. nisms, so that intracellular concentration of
Increased extracellular concentrations of this free amino acid is extremely high
these amino acids facilitate their uptake by (20-40 mM).57 However, lowered Tau con-
cardiomyocytes.48 There are experimental facts tents have been reported in cardiomyopathies
suggesting that the reduced intracellular pH induced by hypoxia and ischemia58 and in
during myocardial ischemia favors the hearts subjected to calcium paradox.59 Recent
transport of BCAA from cytosol into the assessment of Tau release into the interstitial
mitochondrial matrix, where they are tran- fluid by microdialysis technique revealed its
saminated.49 These reactions yield branched potential significance as a marker of myocar-
chain 2-oxoacids and Glu, which is a power- dial cell injury in experimental coronary
ful antiischemic agent itself. However, further occlusion.60 Profound Tau losses were associ-
catabolism of BCAA is probably inhibited, ated with irreversible damage of myocytes and
since it involves steps catalyzed by NAD+ and accompanied by increased plasma levels of cre-
FAD-dependent dehydrogenases and by ATP- atine phosphokinase MB fraction. The loss of
linked carboxylases which are blocked under myocardial Tau and other !-amino acids was
ischemia.50 also observed in patients undergoing cold car-
It has been claimed that after cardioplegic dioplegia with St. Thomas’ Hospital cardio-
arrest, increased intracellular concentrations of plegic solution.61 Accordingly, postoperatively
BCAA stimulate formation of acetyl-CoA and increased Tau levels were found in the blood
succinyl-CoA depleted during ischemia, thus of patients subjected to major surgical proce-
enhancing recovery of oxidative phosphoryla- dures.62 As a rule, correction of Tau depletion
tion on reperfusion.47 As Glu and Asp, BCAA results in improvement of myocardial dam-
are the only exogenous substrates extracted by age.59 Franconi et al63 found a good positive
the human heart early after cardiac surgery. relation between intracellular and extracellu-
Myocardial uptake of these amino acids cor- lar concentrations of Tau, suggesting that
relates positively with their arterial concentra- administration of high doses of Tau increases
tions and oxygen consumption.37,51 This sug- its content in the myocardium. In this regard,
gests that the accessibility of these substrates the addition of Tau to cardioplegic and
to myocytes may be one of the factors deter- reperfusate solutions or in organ storage media
mining postischemic recovery of energy may be a worthwhile measure to prevent the
metabolism. Other potential benefits of loss of !-amino acid pool and disturbances of
increased intracellular levels of BCAA are usu- energy metabolism.64,65
ally attributed to the promotion of protein Tau has been shown to possess antioxidant
synthesis and reduction of proteolysis.52 Fur- activity suppressing lipoperoxide formation
ther clinical studies investigating the metabolic and membrane phospholipid degradation,
and functional effects of BCAA in postopera- which could also account for its cardio-
tive settings are, therefore, warranted. protective action. This “membrane-stabilizing”
effect is accompanied by a lesser release of
Taurine myocardial reduced glutathione, creatine
This is the most abundant amino acid of phosphokinase and diminished depletion of
the mammalian heart comprising about 50% ATP.66,67 A free-radical scavenging action of
of the total free amino acid pool.53 Although Tau was observed in patients during myocar-
its physiological function remains undefined, dial revascularization. Reduction of reper-
Tau exhibits an extensive cardiovascular phar- fusion injury due to preoperative intravenous
macology as an antiarrhythmic agent,54 a infusion of Tau was associated with reduced
lipoperoxidation and significantly lesser dam-
Metabolic and Antioxidant Support with Amino Acids 95

age of cardiomyocyte ultrastructure assessed Like Met, Cys potentiates vasodilating


in biopsy samples.68 Surprisingly, despite abun- effect of nitroglycerin through formation of
dance of promising experimental data the rela- S-nitrosothiols which activate guanilate
tive benefit of myocardial support with Tau cyclase.78 Cys reverses structural changes of mi-
has not been properly evaluated in cardiac tochondrial membranes and restores the aor-
surgery. tic flow in rat heart perfused with uncouplers
of oxidative phosphorylation.79 This effect is
Methionine and Cysteine attributed to preventing interaction of
Met deficiency is mainly associated with uncouplers with SH and amino groups at the
disturbances in lipid metabolism and myocar- inner mitochondrial membrane which results
dial lesions, since this essential amino acid is in reduction of a depletion of high energy
required for transmethylation of membrane phosphates. There were no attempts to
phospholipids.69,70 In patients with coronary enhance postischemic recovery of cardiac func-
artery disease, Met potentiates the hemody- tion and metabolism using Cys as a donor of
namic effects of acutely administrated intra- SH groups in cardioplegic and reperfusate
venous nitroglycerin.71 This observation sup- solutions.
ports the concept that nitroglycerin action
requires sulfhydril groups (SH) to form Conclusion
S-nitrosothiols in vascular smooth muscle. Under conditions relevant to cardiac sur-
These, in turn, activate guanilate cyclase, gery, mechanisms of action of the majority of
which stimulates the production of cyclic amino acids, except Glu and Asp, have not
GMP, which is known to mediate vasodilata- yet been thoroughly elucidated. Presumably,
tion.72 Met contains no SH groups, but it is they are closely linked with adaptive derange-
converted to Cys, SH-amino acid and, thus, ment of myocardial metabolism induced by
may provide additional formation of S-nitro- ischemic and reperfusion injury. As seen,
sothiols leading to increased cyclic GMP levels. amino acid intervention may affect interme-
As a powerful OH• radical scavenger,73 Met diary and energy metabolism, ion homeosta-
is effective in reducing myocardial damage sis, scavenging oxygen-derived free radicals,
induced by oxygen free radical formation fol- and subsequently membrane integrity and
lowing ischemia and reperfusion. Thus, addi- myocardial performance. In view of such mul-
tion of Met to the St. Thomas’ Hospital tifactorial effect on the state of myocardial
cardioplegic solution enhances postischemic metabolism, it seems logical to use these natu-
functional recovery and cell membrane rally-occurring compounds in various set-
integrity in the isolated working heart model tings of cardiac surgery. Further studies are
of cardiopulmonary bypass and ischemic car- needed to delineate the optimal regimens of
diac arrest.74 As Met is not freely permeable myocardial treatment with individual amino
through the cell membrane,75 its scavenging acids or their combination.
effect may be mediated extracellularly, on cell
surfaces and in the capillary endothelium.
There are several reports of Met ability to pre- References
vent changes in myofibrillar ATPase activities 1. Neely JR, Morgan HE. Relationship between
induced by hypochlorous acid (HOCl) and carbohydrate and lipid metabolism and en-
ergy balance of heart muscle. Ann Rev Physiol
H2O2.76,77 This effect, associated with a lesser 1974; 36: 413-459.
oxidation of myofibrillar SH groups, suggests 2. Opie LH. Fuels. In: The Heart. Physiology,
that Met can also protect the basic contractile Metabolism, Pharmacology and Therapy.
machinery of the myocardial cell against the New York: Gruine & Stratton Ltd., 1984:
111-117.
adverse influence of oxygen free radicals. 3. Mudge GH, Mills RM, Taegtmeyer H et al.
Alterations of myocardial amino acid metabo-
lism in chronic ischemic heart disease. J Clin
Invest 1976; 58:1185-1192.
96 Ischemia-Reperfusion Injury in Cardiac Surgery

4. Knapp WH, Helus F, Ostertag H et al. Up- 17. Rosenkranz ER, Okamoto F, Buckberg GD
take and turnover of L-[13N]glutamate in the et al. Safety of prolong aortic clamping with
normal heart and in patients with coronary blood cardioplegia. III. Aspartate enrichment
artery disease. Eur J Nucl Med 1982; 7: of glutamate-blood cardioplegia in energy-
211-215. depleted hearts after ischemic and reperfusion
5. Sanborn T, Gavin W, Berkowitz S et al. injury. J Thorac Cardiovasc Surg 1986;
Augmented conversion of aspartate and 91:428-435.
glutamate to succinate during anoxia in rab- 18. Kofsky E, Julia P, Buckberg GD et al. Stud-
bit heart. Am J Physiol 1979; 273: ies of myocardial protection in the immature
H535-H541. heart. V. Safety of prolonged aortic clamp-
6. Haas DW. Phosphorylation coupled to the ing with hypocalcemic glutamate/aspartate
oxidation of NADH by fumarate in digito- blood cardioplegia. J Thorac Cardiovasc Surg
nin fragments of beef heart mitochondria. 1991; 101:33-43.
Biochim Biophys Acta 1964; 92: 433-439. 19. Pisarenko OI, Rosenfeldt FL, Lagley L et al.
7. Pisarenko OI, Khlopkov VN, Ruuge EK. A Differing protection with aspartate and
1H-NMR study of succinate synthesis from glutamate cardioplegia in the isolated rat
exogenous precursors in oxygen-deprived rat heart. Ann Thorac Surg 1995; 59:1541-1548.
heart mitochondria. Biochem Internat 1986; 20. Haan CK, Lazar HL, Rivers S et al. Improved
12:145-153. myocardial preservation during cold storage
8. Wright G. Amino acids in the treatment of using substrate enhancement. Ann Thorac
ischemic heart disease. J Mol Cell Cardiol Surg 1990; 50:80-85.
1985; 17:441-443. 21. Choong YS, Gavin JB. L-Aspartate improves
9. Owen TG, Hochachka PW. Purification and the functional recovery of explanted hearts
properties of Dolphin muscle aspartate and stored in St. Thomas’ Hospital cardioplegic
alanine transaminases and their possible roles solution at 40oC. J Thorac Cardiovasc Surg
in the energy metabolism of diving mammals. 1990; 99:510-517.
Biochem J 1974; 143:541-553. 22. Bittl JA, Shine K. Protection of ischemic rab-
10. Hohl CH, Oestreich R, Rosen P et al. Evi- bit myocardium by glutamic acid. Am J
dence for succinate production by reduction Physiol 1983; 245:H406-412.
of fumarate during hypoxia in isolated adult 23. Williamson JR, Ford C, Illingworth J et al.
rat heart cells. Arch Biochem Biophys 1987; Coordination of citric acid cycle activity with
259:527-535. electron transport flux. Circ Res 1976; 38
11. Rau EE, Shine KI, Douglas AM et al. En- (Suppl 1):39-48.
hanced mechanic recovery of anoxic and is- 24. Digerness SB, Reddy W. The malate-aspar-
chemic myocardium by amino acid reper- tate shuttle in heart mitochondria. J Mol Cell
fusion. Am J Physiol 1979; 236:H873-879. Cardiol 1976; 8:779-785.
12. Pisarenko OI, Solomatina ES, Studneva IM 25. Katunuma N, Okada M, Nishii Y. Regula-
et al. Protective effect of glutamic acid on tion of the urea cycle and TCA cycle by
cardiac function and metabolism during car- ammonia. Adv Enzyme Reg 1966; 4:317-336.
dioplegia and reperfusion. Basic Res Cardiol 26. Humphrey SM, Holliss DG, Seelye RN. Ad-
1983; 78:534-543. enine pool catabolism in the ischemic, the
13. Pisarenko OI, Portnoy VF, Studneva IM et calcium-depleted and substrate-free anoxic
al. Glutamate-blood cardioplegia improves isolated rat heart: Relationship to contracture
ATP preservation in human myocardium. development. J Mol Cell Cardiol. 1984;
Biomed Biochim Acta 1987; 46:499-504. 16:1127-1136.
14. Weldner PW, Myers JL, Miller CA et al. 27. Morita K, Ihnken K, Buckberg GD et al.
Improved recovery of immature myocardium Studies of hypoxemic/reoxygenation injury:
with L-glutamate blood cardioplegia. Ann Without aortic clamping. VIII. Counteraction
Thorac Surg 1993; 55:102-105. of oxidant damage by exogenous glutamate
15. Engelman RM, Dodds WA, Rousou JH et and aspartate. J Thorac Cardiovasc Surg
al. Myocardial high-energy phosphate replen- 1995; 110:1228-1234.
ishment during ischemic arrest: Aerobic ver- 28. Haas GS, DeBoer LWV, O’Keefe DD et al.
sus anaerobic metabolism. Ann Thorac Surg Reduction of postischemic myocardial dys-
1982; 33:453-458. function by substrate repletion during reper-
16. Lazar HL, Buckberg GD, Mangarano AJ et fusion. Circulation 1984; 70 (Suppl 1):65-74.
al. Myocardial energy replenishment and re- 29. Rousou JA, Engelman RM, Anisimowicz L
versal of ischemic damage by substrate en- et al. Metabolic enhancement of myocardial
hancement of secondary blood cardioplegia preservation during cardioplegic arrest. J
with amino acids during reperfusion. J Thorac Thorac Cardiovasc Surg 1986; 91:270-276.
Cardiovasc Surg 1980; 80:350-359.
Metabolic and Antioxidant Support with Amino Acids 97

30. Engelman RM, Rousou JA, Flack JE et al. 42. Misra BR, Misra HP. Vasoactive intestinal
Reduction of infarct size by systemic amino peptide, a singlet oxygen quencher. J Biol
acid supplementation during reperfusion. J Chem 1990; 265:15371-15374.
Thorac Cardiovasc Surg 1991; 101:855-859. 43. Zs-Nagy I, Floyd RA. Hydroxyl free radical
31. Singh J, Garg D, Chugh K et al. Effect of reactions with amino acids and proteins:
aspartate and glutamate on experimental myo- Studies by electron spin resonance spectros-
cardial infarction in rats. Indian J Exper Biol copy and spin-trapping. Biochim Biophys
1989; 27:621-624. Acta 1984; 790:238-250.
32. Beyersdorf F, Acar C, Buckberg GD et al. 44. Heinemeyer D, Belles G, Stapenhorst K.
Studies on prolonged acute regional ischemia. Intracellular pH measurement during car-
V. Metabolic support of remote myocardium diac arrest in ventricular myocardium by
during left ventricular power failure. J Thorac Bretschneider’s cardioplegic solution HTK
Cardiovasc Surg 1989; 98:567-579. and St. Thomas’ Hospital solution with and
33. Beyersdorf F, Kirsh M, Buckberg GD et al. without procaine. Thorac Cardiovasc Surg
Warm glutamate/aspartate-enriched blood 1987; 35:48-52.
cardioplegic solution for perioperative sudden 45. Bretschneider HJ. Myocardial Protection.
death. J Thorac Cardiovasc Surg 1992; Thorac Cardiovasc Surg 1980; 28:295-302.
104:1141-1147. 46. Schwalb H, Kushnir T, Navon G et al. The
34. Rosenkranz ER, Buckberg GD, Laks H et protective effect of enriched branched chain
al. Warm induction of cardioplegia with amino acid formulation in the ischemic heart:
glutamate-enriched blood in coronary artery A phosphorus-31 nuclear magnetic resonance
patients with cardiogenic shock who are de- study. J Mol Cell Cardiol 1987; 19:991-998.
pendent on inotropic drugs and intra-aortic 47. Schwalb H, Izhar U, Yaroslavsky E et al. The
balloon support. J Thorac Cardiovasc Surg effect of amino acids on the ischemic heart.
1983; 86:507-518. J Thorac Cardiovasc Surg 1989; 98:551-556.
35. Thomassen A, Nielsen TT, Bagger JP et al. 48. Schwartz RG, Barret EJ, Francis CK et al.
Antiischemic and metabolic effects of gluta- Regulation of myocardial amino acid balance
mate during pacing in patients with stable in conscious dog. J Clin Invest 1985; 75:
angina pectoris secondary to either coronary 1204-1211.
artery disease or syndrome X. Am J Cardiol 49. McMillin JB. Branched chain amino acid
1991; 68:291-295. perfusion. J Thorac Cardiovasc Surg 1989;
36. Svedjeholm R, Hakason E, Vanhanen I. Ra- 98:632-638.
tionale for metabolic support with amino ac- 50. Pisarenko OI, Solomatina ES, Studneva IM.
ids and glucose-insulin-potassium (GIK) in The role of amino acid catabolism in the for-
cardiac surgery. Ann Thorac Surg 1995; mation of the tricarboxylic acid cycle inter-
59:S15-S22. mediates and ammonia in anoxic rat heart.
37. Svedjeholm R, Ekroth R, Joachimsson PO et Biochim Biophys Acta 1986; 885:154-161.
al. Myocardial uptake of amino acids and 51. Hallhagen S, Svedjeholm R, Ekroth R et al.
other substrates in relation to myocardial oxy- Effects of insulin on myocardial uptake of
gen consumption four hours after cardiac branched chain amino acids soon after car-
surgery. J Thorac Cardiovasc Surg 1991; diac operations. J Thorac Cardiovasc Surg
101:688-694. 1992; 103:98-107.
38. Pisarenko OI, Studneva IM, Portnoy VF et 52. Tischler ME, Cammisa H. Metabolism of
al. Ischemic heart arrest: Nitrogenous metabo- protein, amino acids, and glucose and their
lism in energy-depleted human myocardium. response to insulin in atria and cardiac
Eur Surg Res 1987; 19:329-336. myocytes of traumatized rats. Metabolism
39. Pisarenko OI, Lepilin MG, Ivanov VE. Car- 1984;33:515-520.
diac metabolism and performance during L- 53. Jacobsen JG, Smith LH. Biochemistry and
glutamic acid infusion in postoperative car- physiology of taurine and taurine derivatives.
diac failure. Clin Sci 1986; 70:7-12. Physiol Rev 1968; 48:421-511.
40. Kusama, Bernier M, Hearse DJ. Photo- 54. Huxtable RJ, Sebring LA. Cardiovascular ac-
activation of porphyrins: Studies of reactive tion of taurine. In: Kiriyama K, Huxtable RJ,
oxygen intermediates and arrhythmogenesis in Iwata H, ed. Sulfur amino acids: Biochemi-
the aerobic rat heart. Cardiovasc Res 1990; cal and clinical aspects. New York: Alan R.
24:676-682. Liss, 1983: 5-37.
41. Kukreja RC, Kearns AA, Zweier JL et al. Sin- 55. Chovan J, Kulakowski E, Benson B et al.
glet oxygen interaction with Ca2+-ATPase of Taurine enhancement of calcium binding to
cardiac sarcoplasmic reticulum. Circ Res rat heart sarcolemma. Biochim Biophys Acta
1991; 69:1003-1014. 1979; 551:129-136.
98 Ischemia-Reperfusion Injury in Cardiac Surgery

56. Huxtable RJ, Sebring LA. Towards an unify- 70. Panagia V, Okumura K, Makino N. et al.
ing theory for the action of taurine. Trends Stimulation of Ca2+ pump in heart sarco-
Pharm Sci 1986; 7:481-485. lemma by phosphatidylethanolamine N-me-
57. Huxtable RJ, Bressler R. Elevation of taurine thylation. Biochim Biophys Acta 1986;
in human congestive heart failure. Life Sci- 856:383-387.
ence 1974; 4:1353-1359. 71. Levi WS, Katz RJ, Ruffalo RL et al. Poten-
58. Crass MF, Lombardini JB. Release of taurine tiation of the hemodynamic effects of acutely
from the oxygen-deficient perfused heart. Proc administered nitroglycerin by methionine.
Soc Exper Biol Med 1978; 157:486-488. Circulation 1988; 78:640-645.
59. Kramer J, Chovan J, Schapper S. The effects 72. Ignarro LJ, Lippton H, Edwards JC et al.
of taurine on calcium paradox and ischemic Mechanisms of vascular smooth muscle relax-
heart failure. Am J Physiol 1981; 240: ation by organic nitrates, nitrites, nitroprus-
H238-246. side and nitric oxide: Evidence for the in-
60. Holmgren A, Anderson B, Haggmark S et al. volvement of S-nitrosothiols as active inter-
Monitoring of myocardial ischemic and mediates. J Pharmacol Exp Ther 1981;
reperfusion damage. A microdialysis study 218:739-745.
with taurine as a marker. J Mol Cell Cardiol 73. Blasig IE. Effects of troxerutin and methion-
1997; 29:A89 (Abstract). ine on spin trapping of free oxy-radicals.
61. Chapman RA, Suleiman MS, Rodrigo GS. Biomed Biochim Acta 1988; 47:252-255.
Intracellular taurine, intracellular sodium and 74. Chambers DJ, Astras G, Takahashi A et al.
defense against cellular damage. In: Earm, Free radicals and cardioplegia: organic anti-
Y.E. & Noble, D. eds, Cardiac Ion Chan- oxidants as additives to the St. Thomas’ Hos-
nels and Effects of Taurine on the Heart. pital cardioplegic solution. Cardiovasc Res
Boston: Kluwer Academic Publishers, 1993: 1989; 23:351-358.
73-92. 75. Halliwell B. Free radicals, oxygen toxicity and
62. Lombardini JB, Bricker DL. Effects of car- aging. In: Age Pigments, Shoal, RS, ed.
diovascular surgery on blood concentrations Amsterdam: Elsevier/North Holland Biomedi-
of taurine and amino acids. Proc Soc Exper cal Press 1981: 1-62 .
Biol Med 1981; 167:498-505. 76. Kukreja RC, Weaver AB, Hess ML. Stimu-
63. Franconi F, Martini F, Stendardi I et al. Ef- lated human neutrophils damage cardiac sar-
fect of taurine on calcium level and contrac- coplasmic reticulum function by generation
tility in guinea pig ventricular strips. Biochem of oxidants. Biochim Biophys Acta 1989;
Pharmacol 1982; 31:3181-3186. 990:198-205.
64. Earm YE, Ho WK, So IS et al. Effect of tau- 77. Suzuki S, Kaneko M, Chapman DC et al.
rine on the activation of background current Alterations in cardiac contractile proteins due
in cardiac myocytes. In: Earm YE and Noble to oxygen free radicals. Biochim Biophys Acta
D eds. Cardiac Ion Channels and Effects of 1991; 1074:95-100.
Taurine on the Heart. Boston: Kluwer Aca- 78. Laustiola K, Vuorinen P, Vapaatalo H et al.
demic Publishers 1993: 132-140. Effects of cysteine and nitroglycerin on bo-
65. Kapelko VI, Pukhova TM, Lakomkin VL et vine heart guanilate cyclase and on tissue cy-
al. Modification of reperfusate formulae for clic GMP and lactate of rat atria. Eur J
ischemic heart. Cardiology (Russ) 1993; Pharmacol 1983; 91:301-304.
7:246-251. 79. Viet P, Fuchs J, Zimmer G. Uncoupler- and
66. Pasantes-Morales H, Wrigh CE, Gaull GE. hypoxia-induced damage in the working rat
Taurine protects lymphoblastoid cells from heart and its treatment. I. Observation with
iron-ascorbate induced damage. Biochem uncouplers of oxidative phosphorylation. Ba-
Pharmacol 1985; 34:2205-2207. sic Res Cardiol 1985; 80:107-115.
67. Ohta H, Azuma J, Awata N et al. Mecha-
nism of the protective action of taurine
against isoprenaline induced myocardial dam-
age. Cardiovasc Res 1988; 22:407-413.
68. Milei J, Ferreira R, Llesuy S et al. Reduction
of reperfusion injury with preoperative rapid
intravenous infusion of taurine during myo-
cardial revascularization. Am Heart J 1992;
123:339-345.
69. Lovenberg W, Yomori Y. Nutritional factors
and cardiovascular disease. Clin Exp Hyperten
1984; A6:417-426.
CHAPTER 9

Methods to Reduce Ischemia/


Reperfusion Injury—PICSO
Günter Steurer, Katharina Palisek and Werner Mohl

Ischemia/Reperfusion Injury

E
normous advances in surgical,
pharmacological, and interventional vessels, and hemodynamic complications.
techniques resulting in early restoration Early reperfusion damages vascular endothe-
of infarct artery patency significantly improved lium and capillary structures of the coronary
outcome in patients with acute coronary syn- microcirculation by formation of oxygen-de-
dromes.1 However, time-consuming prepara- rived free radicals, activation of the comple-
tions in patients with acute myocardial ment system and disturbance of the calcium
ischemia scheduled for emergency bypass sur- homeostasis.7 Increasing evidence of reper-
gery, prolonged interventional procedures in fusion-induced myocardial damage in routine
patients with complex coronary lesions, and revascularization therapy of infarct patients
early re-establishment of coronary blood flow stimulated interest in therapy strategies to
in acute myocardial infarction by means of attenuate myocardial ischemia/reperfusion
thrombolytic agents or primary angioplasty injury in acute myocardial ischemia.
may result in myocardial tissue damage partly As a rule the more time elapsed the higher
related to reperfusion of oxygenated blood into is the danger of formation of oxygen-derived
ischemia-damaged myocardial areas.2 Myocar- radicals leading to changes in microcirculation
dial stunning,3 ventricular arrhythmias,4 or as shown by Komamura et al.8 In this trial a
even lethal cell injury in the sequel of success- decrease of coronary vein flow during
ful reperfusion therapy5 were observed and reperfusion after myocardial infarction was
confirmed the clinical relevance of reperfusion shown, demonstrating again that arterial early
injury in acute myocardial infarction. reperfusion does not necessarily salvage myo-
cardium. As a conclusion, time should be the
Coronary reperfusion within a critical pe- determining factor whether to choose arterial
riod is essential for survival of ischemic myo- reperfusion, as after acute vessel occlusion or
cardium. In experimental conditions coronary ischemia in the cath lab, or venous retrograde
occlusion followed by reperfusion revealed reperfusion when the exact time of the occlu-
progression of irreversible injury of ischemic sion is unknown.
myocardium from the endocardium toward
the subepicardium with potentially salvageable Coronary Sinus Interventions
myocardium for up to 3-6 hours.6 The course Coronary sinus techniques comprising the
of events is similar in man depending on degree methods of retroinfusion, 9 synchronized
of coronary stenosis, size of the ischemic area, retroperfusion (SRP),10-13 selective suction and
size and number of pre-existing collateral retroperfusion (SSR),14 intermittent coronary

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


100 Ischemia-Reperfusion Injury in Cardiac Surgery

sinus occlusion (ICSO),15-17 and pressure-con- (CAN). The knot “user interface” is a stan-
trolled intermittent coronary sinus occlusion dard industry personal computer, the knot
(PICSO)18-30 are based on the experimental “master” is a microcontroller system connected
studies of Pratt in the isolated heart31 to main- to an instrumentation amplifier linked to a
tain residual myocardial blood flow via the pressure sensor, the knot “electrocardiogram
coronary venous system. The coronary venous (ECG)” is a serial linked I/O (SLIO) control-
vasculature, a dense meshwork with numer- ler connected to an ECG amplifier, and the
ous interconnections, has three different drain- knot “balloon” is a SLIO controller connected
age systems, the coronary sinus, the lymphatic to an amplifier linked to a sensor measuring
system, and the Thebesian system. The differ- the balloon pressure and to a motor balloon
ent compartments are connected with each pump.
other and create a vast and normally unused The PICSO system redistributes coronary
volume capacity. This capacitance and the venous blood flow by changes in pressure gra-
numerous interconnections such as arterio- dients throughout the coronary venous sys-
venous, lymphatic, and venovenous anasto- tem in the setting of coronary artery occlu-
moses provide the essential basis and the sion (Fig. 9.2). The pump automatically
mechanism of all coronary sinus interventions. inflates/deflates the balloon according to the
Evidence of beneficial effects of coronary occlusion/release timing obtained from the
sinus interventions in experimental ischemia closed-loop feedback algorithm of the system
and clinical studies renewed interest in coro- (Fig. 9.3). During balloon inflation there is a
nary sinus interventions to protect ischemic slow increase in systolic and diastolic coronary
myocardium from irreversible damage due to sinus pressure with the effect of redistribution
ischemia/reperfusion injury in patients with of the coronary venous blood flow from nor-
acute ischemia by extension of the filling mal to underperfused, ischemic areas (redis-
capacity of the smallest cardiac veins (SCV) tribution period). Gradual rise of systolic coro-
and through improvement of myocardial per- nary sinus pressure to a plateau, and a
fusion. Today’s role of coronary sinus inter- concomitant increase of systolic pressure in the
vention in the therapy of coronary artery dis- perfusion area of the infarct-related coronary
ease is based on the theory of temporary artery, results in exchange of toxic metabolic
protection of jeopardized myocardium in products (equilibrium period). The occlusion
patients with acute coronary syndromes period is automatically terminated when the
undergoing early revascularization procedures. exponential curve fitted to the systolic coro-
nary sinus pressure peaks 95% of its predicted
Pressure-Controlled Intermittent plateau value. In healthy controls, plateau val-
Coronary/Sinus Occlusion ues of 60-80 mmHg are usually observed, and
occlusion times approximately range from 6-
(PICSO) 10 seconds. In patients with myocardial inf-
The technique of PICSO developed by arction, due to the loss of myocardial contrac-
Mohl and coworkers19 is based on a forced tility, however, slope of the peak values will be
redistribution of venous blood flow into the drastically reduced resulting in decreased pla-
coronary beds in the setting of coronary artery teau values and substantially prolonged occlu-
occlusion to reduce infarct size, improve sion times. Following termination of the
regional myocardial function, and enhance PICSO cycle by deflation of the balloon, coro-
washout of ischemic metabolic byproducts nary sinus pressure abruptly decreases and
(Fig. 9.1). The PICSO device is composed of coronary sinus flow is released, facilitating an
a balloon-tipped catheter positioned in the ori- enhanced drainage of the venous system and
fice of the coronary sinus by means of fluo- reestablishing flow currents that promote
roscopy, a pneumatic pump device, and a washout of accumulated toxic wastes in the
decentral process computer connected by a ischemic area (washout phase). After a release
four knots Field Bus Controller Area Network period of approximately 6-7 heart beats and
Methods to Reduce Ischemia/Reperfusion Injury—PICSO 101

Fig. 9.1. Illustration of a thermographic study of retrograde myocardial blood supply during coronary sinus
intervention in experimental ischemia. Note the disappearance of the dark color during coronary sinus occlusion
(D) determining improved perfusion. During coronary sinus release (E) washout takes place (recurrence of dark
color).
102 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 9.2. Schematically illustration of myocardial blood supply in normal conditions, during acute ischemia, and
during coronary sinus intervention (PICSO).

complete return of coronary sinus pressure to cardium in the PICSO group compared to
baseline levels, the balloon is automatically controls. In two experimental occlusion/
inflated in order to initiate the next PICSO reperfusion models, however, PICSO15 and
cycle. The intermittent nature of the inflation/ ICSO,25 respectively, did not significantly re-
deflation cycle prevents potential complica- duce infarct size or afford myocardial protec-
tions of hemorrhage, edema, thrombosis, ar- tion. The negative results of these trials could
rhythmias, or conduction disturbances in spite be related to the insufficient increase of sys-
of an individual peak coronary sinus pressure tolic coronary sinus pressure during coronary
of 60 mmHg or more. sinus intervention and inadequate individual
adaptation of the PICSO algorithm.
Experimental Studies Recent experimental studies on pigs have
In experimental myocardial infarction shown that PICSO effectively reduced both,
ICSO and PICSO demonstrated a significant infarct size and myocardium at risk after coro-
reduction of infarct size compared to controls nary artery occlusion.27-29 In addition to
(Fig. 9.4). In 31 mongrel dogs with experi- reduction of infarct size in experimental myo-
mental infarction due to ligation of the left cardial infarction a significant improvement
anterior descending coronary artery (LAD) for of regional ischemic myocardial function due
6 hours Mohl et al20 showed a significant in- to temporary intravascular balloon occlusion
crease of myocardial salvage by PICSO com- of the proximal LAD by PICSO has been
pared to controls. In 41 dogs subject to a 3 demonstrated.21
hours occlusion of the proximal LAD Ciuffo
et al16 showed a significant reduction of is- Clinical Studies
chemic necrosis and significant increase of Safety and efficacy of PICSO was evalu-
myocardial perfusion in the ICSO therapy ated in a randomized trial of 30 patients with
group compared to controls. In 22 dogs sub- coronary artery disease undergoing bypass sur-
ject to 3 hours ligation of the LAD followed gery.30 To analyze changes of sectional and seg-
by 8-12 days or reperfusion Guerci et al26 mental wall motion during extracorporeal
showed a significant increase of myocardial circulation intraoperative two-dimensional
salvage in the ICSO therapy group compared echocardiography was performed from the
to controls. In 22 mongrel dogs with LAD short-axis cross-sectional view. PICSO was
occlusion for 3 hours followed by 3 hours of started after aortic declamping and continued
reperfusion Jacobs et al23 showed a significantly for one hour during early reperfusion.
enhanced myocardial salvage of ischemic myo- Although sectional wall motion did not sig-
Methods to Reduce Ischemia/Reperfusion Injury—PICSO 103

Fig. 9.3 Schematic illustration of a PICSO cycle during acute ischemia and depicts the increase in venous perfusion
in relation to the coronary sinus pressure.

Fig. 9.4. Summary of the effects of PICSO and ICSO in experimental reperfused myocardial infarction.
104 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 9.5. Illustration of the time course of the systolic (solid line) and diastolic (dashed line) coronary sinus pressure
(CSP) plateau values during intraoperative PICSO treatment in one patient.

nificantly change, hypokinetic segments were angina or emerging myocardial infarction,


preserved better in the PICSO therapy group during high-risk interventional procedures,
compared to placebo coronary sinus interven- and during cardiac arrest in bypass surgery.
tion. Coronary artery bypass grafting was per-
formed, and PICSO was applied after reopen- References
ing of the aorta while the patient was still on 1. Grover A, Rihal CS. The importance of early
cardio-pulmonary bypass. Note the sudden patency after acute myocardial infarction.
increase of the systolic plateau during open- Current Opinion in Cardiology 1995; 10:
361-366.
ing of the bypass graft (Fig. 9.5).
In patients with anterior acute myocardial 2. Braunwald E. Myocardial reperfusion, limi-
infarction treated with thrombolytic therapy tation of infarct size, reduction of left ven-
tricular dysfunction, and improved survival.
implementation of ICSO was obtained in 12 Should the paradigm be expanded? Circula-
patients and compared 12 patients treated with tion 1989; 79:441-444.
thrombolysis alone.18 In the absence of adverse 3. Bolli R. Myocardial stunning in man. Circu-
effects ICSO therapy slightly improved enzy- lation 1992; 86:1671-1691.
4. Kusama Y, Bernier M, Hearse DJ. Exacerba-
matic infarct size, myocardial lactate metabo- tion of reperfusion arrhythmias by sudden
lism, left ventricular wall motion, and oxidant stress. Circulation Research 1990;
scintigraphically documented perfusion 67:481-489.
defects compared to controls. 5. Ambrosio G, Becker LC, Hutchins GM et
al. Reduction in experimental infarct size
by recombinant human superoxide dis-
Future Directions mutase: Insights into the pathophysiology
Based on the positive effects of PICSO and of reperfusion injury. Circulation 1986;
ICSO in experimental myocardial ischemia 74:1424-1433.
6. Reimer KA, Lowe JE, Ramussen MM et al.
and clinical studies randomized clinical stud- The wave front phenomenon of ischemia cell
ies are elaborated to evaluate the safety and death: Myocardial infarct size vs. duration of
efficacy of PICSO in patients with unstable coronary occlusion in dogs. Circulation 1977;
56:786-794.
Methods to Reduce Ischemia/Reperfusion Injury—PICSO 105

7. Steurer G, Yang P, Rao V et al. Acute myo- 19. Mohl W. The development and rationale of
cardial infarction, reperfusion injury, and in- pressure controlled intermittent coronary si-
travenous magnesium therapy: Basic concepts nus occlusion: A new approach to protect is-
and clinical implications. Am Heart J 1996; chemic myocardium. Wi Kli Wo 1984;
132:478-482. 96:205-209.
8. Komamura K, Kitakaze M, Nishida K et al. 20. Mohl W, Glogar DH, Mary H et al. Reduc-
Progressive decreases in coronary vein flow tion of infarct size induced by pressure-con-
during reperfusion in acute myocardial inf- trolled intermittent coronary sinus occlusion.
arction: Clinical documentation of the no Am J Cardiol 1984; 53:923-928.
reflow phenomenon after successful throm- 21. Mohl W, Punzengruber C, Moser. Effects of
bolysis. J Am Coll Cardiol 1994; 24(2): pressure-controlled intermittent coronary si-
370-377. nus occlusion on regional ischemic myocar-
9. Bolling SF, Flaherty JF, Bulkley BH. Im- dial function. J Am Coll Cardiol 1985;
proved myocardial preservation during global 5:939-947.
ischemia by continuous retrograde coronary 22. Mohl W, Glogar D, Kenner T. Enhancement
sinus perfusion. J Thorac Cardiovasc Surg of washout induced by PICSO in the canine
1983; 86:659. and human heart. In: W Mohl, W Wolner,
10. Meerbaum S, Lang TW, Osher JV et al. and D Glogar, eds. The coronary sinus. New
Diastolic retroperfusion of acutely ischemic York: Springer-Verlag; 1984:537-548.
myocardium. Am J Cardiol 1976; 37: 23. Jacobs SK, Faxon DP, Coats WD et al. Pres-
588-598. sure-controlled, intermittent coronary sinus
11. Farcot JC, Meerbaum S, Lang TW et al. Am occlusion (PICSO) during reperfusion mark-
J Cardiol 1978; 41:1191-1201. edly reduces infarct size. Clin Res 1985;
12. Gore JM, Weiner BH, Benotti JR et al. Pre- 33:197A.
liminary experience with synchronized coro- 24. Jacobs SK, Faxon DP, Coats WD et al. The
nary sinus retroperfusion in humans. Circu- effect of pressure controlled intermittent coro-
lation 1986; 74:381-388. nary sinus occlusion during reperfusion. In:
13. Nanto S, Nishida K, Hirayama A et al. Sup- W Mohl, ed. Clinics of CSI. Darmstadt:
ported angioplasty with synchronized retro- Steinkopff; 1986:345-347.
perfusion in high risk patients with left main 25. Diltz EA, Mames RN, Lee JW et al. Inter-
trunk or near left main trunk obstruction. Am mittent coronary sinus occlusion does not
Heart J 1993; 125:301-309. reduce infarct size or ischemic dysfunction in
14. Boekstegers P, Peter W, Degenfeld G et al. an occlusion/reperfusion model. Circulation
Preservation of regional myocardial function 1988; 72(supp III):120.
and myocardial oxygen tension during acute 26. Guerci AD, Ciuffo AA, DiPaula AF et al.
ischemia in pigs: Comparison of selective syn- Intermittent coronary sinus occlusion in dogs:
chronized suction and retroinfusion of coro- reduction of infarct size 10 days after reper-
nary veins to synchronized coronary venous fusion. JACC 1987; 9:1075-1081.
retroperfusion. J Am Coll Cardiol 1994; 27. Lazar HL, Rajaii A, Roberts AJ. Reversal of
23:459-469. reperfusion injury after ischemic arrest with
15. Zalewski A, Goldberg S, Slysh S et al. Myo- pressure-controlled intermittent coronary si-
cardial protection via coronary sinus interven- nus occlusion. J Thorac Cardiovasc Surgery
tion: Superior effects of arterialization com- 1988; 95:637-42.
pared with intermittent occlusion. Circulation 28. Lazar HL, Haan CK, Yang X et al. Reduc-
1985; 71:1215-1223. tion of infarct size with coronary venous
16. Ciuffo AA, Guerci AD, Halperin G et al. retroperfusion. Circulation 1992; 96(suppII):
Intermittent obstruction of the coronary sinus 352-357.
following coronary ligation in dogs reduces 29. Lazar HL, Trenor P, Rivers S et al. Combin-
ischemic necrosis and increases myocardial ing percutaneous bypass with coronary retro-
perfusion. The coronary sinus. Steinkopff, perfusion limits myocardial necrosis. Ann
Darmstadt 1984; 454-64. Thorac Surg 1995; 59:373-378.
17. Komamura K. Human experience with inter- 30. Mohl W, Simon P, Neumann F et al. Clini-
mittent coronary sinus occlusion in acute cal evaluation of pressure-controlled intermit-
myocardial infarction. Int Working Group on tent coronary sinus occlusion: Randomized
CSI, Newsletter 1987; 1:9-10. trial during coronary artery surgery. Ann
18. Komamura K, Mishima M, Kodama K. Pre- Thorac Surg 1988; 46:192-201.
liminary clinical experience with intermittent 31. Pratt FH. The nutrition of the heart through
coronary sinus occlusion in combination with the vessels of Thebesius and coronary veins.
thrombolytic therapy in acute myocardial in- Am J Physiol 1893; 1:86-103.
farction. Jap Circ J 1989; 53:1152-1163.
CHAPTER 10

Ischemic Preconditioning—
from Bench to Bedside
Torsten Doenst and Heinrich Taegtmeyer

Discovery, Definition, vitro preparations, and with global as well as


and Capabilities regional ischemia. Most studies have used as

T
experimental models, dogs,1,13,14 pigs,15-17 rab-
raditional ways to improve ischemia
bits,2,18-21 or rats,4,7,10,22 preconditioning effects
tolerance in patients with obstructive
in man.22-26,27 It is of interest that the protec-
coronary artery disease include
tive effects of ischemic preconditioning can
pharmacological and mechanical interven-
also be induced by nonischemic interventions
tions. Recently, ischemic preconditioning has
such as hypoxia,28 rapid pacing,29 or pharma-
emerged as a third modality. Observations in
cological agents30,31 e.g., adenosine agonists32
the laboratory have developed into a strategy
(Table 10.1). Ischemic preconditioning has
for the protection of the ischemic heart which
also been observed in patients undergoing
may prove to be of clinical usefulness. Ischemic
angioplasty. It was noted that balloon infla-
preconditioning was first described more than
tions with occlusion of the vessel subsequent
a decade ago as a phenomenon that increases
to the first occlusion were associated with a
myocardial tolerance against ischemic injury
reduction of ST-segment elevation, with re-
by pre-exposure of the heart to one or more
duced lactate release into the coronary sinus,
brief episodes of ischemia.1 In the original
and with less severe pain of angina pecto-
work, Murry et al1 found a 75% reduction in
ris.22,23,33 More recent reports suggest that
infarct size caused by a 40 min occlusion of a
preinfarction angina may have a protective
coronary artery when the occlusion was pre-
effect.26,34-36 The suggested presence of is-
ceded by four episodes of 5 min ischemia and
chemic preconditioning in patients with coro-
5 min of reperfusion (Fig. 10.1). In the fol-
nary artery disease is also supported by the
lowing years the use of the term ischemic pre-
observation that patients with a transmural
conditioning has been extended to include
myocardial infarction and a history of angina
endpoints other than reduction of infarct size.
within 7 days prior to infarction have a lower
Ischemic preconditioning was shown to pro-
morbidity and a better clinical outcome than
tect against postischemic contractile dysfunc-
patients without preinfarction angina.36
tion2-4 (Fig. 10.2), postischemic dysrhyth-
mia,5,6 autonomic nerve dysfunction,7 vascular
dysfunction,8,9 generation of free oxygen Limitations of Ischemic
radicals, 10,11 and myocardial stunning 12 Preconditioning
(Table 10.1). Despite the described pluripotency of
ischemic preconditioning, the efficacy of the
The phenomenon could be observed in all
intervention is limited by a number of factors.
animal species studied in both in vivo and in

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Ischemic Preconditioning—from Bench to Bedside 107

Fig. 10.1. Reduction of infarct size by ischemic preconditioning without a change in collateral flow. Ischemic
preconditioning with four cycles of 5 min ischemia followed by 5 min of reperfusion ( ) resulted in a 75%
reduction of infarct size after a 40 min sustained occlusion of the circumflex artery compared to nonpreconditioned
control hearts ( , panel A). At the same time collateral flow was unaltered (panel B). Mean ± SEM, *
p < 0.001 compared to Control. Reproduced, with permission from Murry et al. Circulation 1986; 74:1124-1136.

The adaptive response of the heart to the pre- preconditioning ischemia/reperfusion are nec-
conditioning stimulus occurs quickly (within essary to reduce infarct size,45 while others have
2-15 min) and is relatively short-lived. If the shown before that one cycle protects as good
duration of reperfusion between precondition- against infarction as 12 cycles.46 Liu and
ing and sustained ischemia exceeds 120 min Downey45 compared a protocol with three
(in dogs, 60 min in rats) the protective effects cycles of 5 min ischemia and 5 min of
are largely lost. Furthermore, if the duration reperfusion with a protocol with one cycle of
of sustained ischemia exceeds 60-180 min no 5 min ischemia and 10 min of reperfusion and
protective effects can be detected.6,37-39 concluded that the rat heart has a higher
Recent evidence suggests the existence of a threshold of preconditioning than other ani-
“second window of protection”, a slower adap- mal species. On the contrary, we4,47 and oth-
tive response where protective effects are ers48 have found that only one cycle of ischemic
detectable 24h after the preconditioning preconditioning (5 min of ischemia with
stimulus.40 The mechanisms involved in this 10 min of reperfusion) is necessary in the iso-
late protection appear to involve the regula- lated rat heart to improve postischemic recov-
tion of gene expression, especially the expres- ery of contractile function4,47 or to reduce in-
sion of a group of stress proteins termed heat farct size.48 Thus, it seems difficult to establish
shock proteins.40,41 thresholds of ischemic preconditioning for
Another limitation is the poor reproduc- individual animal species, although species
ibility of results. Despite keeping to the differences may be of importance in situations
described time frames several investigators did where several mediators elicit the same
not find protection by ischemic precondition- response (see section 4a).
ing with protocols that have been shown as The literature also contains controversy
protective. 42-44 Different protocols also about the response of the contractile apparatus
produce conflicting results. It has been of preconditioned hearts during ischemia.
reported that in the rat heart three cycles of Both the delay and early onset of ischemic
108 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 10.2. Effects of 15 min ischemia on the recovery of contractile function of isolated working rat hearts perfused
with Krebs-Henseleit buffer containing glucose (10 mM). Control hearts (❍), ischemically preconditioned hearts
( ▼ ), hearts from fasted animals where lactate and insulin were added to the perfusate ( ● ). Note, that both
ischemic preconditioning as well as the metabolic interventions resulted in an almost complete recovery of func-
tion. Mean ± SEM, * p < 0.05 compared with last measurement before ischemia. Reproduced, with permission
from Doenst et al. Am. J. Physiol. 1996; 270:H1607-H1615.

Table 10.1. Ischemic preconditioning

A) Ways to assess the effects on the myocardium

- Reduction in infarct size1,48,134


- Improvement in recovery of postischemic contractile function2,4,42,50,135
- Reduction of postischemic dysrhythmia5
- Reduction in nerve dysfunction7,136
- Reduction of the generation of free oxygen radicals10
- Reduction of the endothelial damage8
- Reduction of stunning12
B) Ways to induce the effects

- Ischemia1,4,38
- Hypoxia28
- Rapid pacing29
- Transient substrate withdrawal95,137
- Pharmacological interventions30,32
- Cyclic variations of coronary flow138
Ischemic Preconditioning—from Bench to Bedside 109

contracture as well as both decreased and If hearts were subjected to 40 min of total
increased end diastolic stiffening during ischemia in 8 episodes of 5 min ischemia,
ischemia have been reported.4,49-52 In contrast, interrupted by short reperfusion periods, the
there is no disagreement on the ability of pre- tissue ATP content at the end of ischemia was
conditioning to improve postischemic cardiac significantly higher than in hearts subjected
function. to one episode of 40 min ischemia. Five years
Similar discrepancies can be found later the same laboratory demonstrated that
throughout the literature on precondition- this strategy was also capable of reducing inf-
ing. These discrepancies may possibly be arct size.1
explained by the different animal models and The preservation of ATP during ischemia
animal species used as well as the different in preconditioned hearts has since been con-
degrees of “inadvertent” preconditioning35 firmed by many others.50,57,58 The mechanism
during the preparation of the model (cannu- for the “ATP-sparing effect”57,59 is still a mat-
lation of hearts,53,54 surgery,35 or cardiopulmo- ter of debate. A reduction in energy demand
nary bypass54). or an inhibition of the mitochondrial F0F1
ATPase, which hydrolyzes ATP during
Effects of Ischemic ischemia,60 have been proposed as plausible
Preconditioning on Cardiac mechanisms. However, it is important to
remember that contractile function is sup-
Metabolism ported by the turnover and not the tissue con-
Since the protection afforded by ischemic tent of ATP. Myocardial ATP and phospho-
preconditioning is short-lived it seems reason- creatine stores would be used up within
able to assume that acute adaptive responses seconds if regeneration did not occur. There-
of the metabolism, or of the regulation of ion fore, it is reasonable to ask whether the term
homeostasis, or of both, are involved in the “ATP-sparing” is not misleading, and whether
mechanism of ischemic preconditioning. the estimation of ATP turnover from measure-
Quite a few such responses have been identi- ments of tissue ATP content at selected time
fied and are summarized in Table 10.2. Three points61 is reliable. The preserved ATP levels
metabolic concepts emerged from these find- at the end of ischemia may either reflect a high
ings. Those are described in detail below and degree of ATP production or a decrease in ATP
they are included in Table 10.3 and the sche- demand. The contradictory findings on the
matic of Figure 10.3. glycolytic rate during ischemia (the main pro-
vider of ATP in the absence of oxygen) in pre-
Preservation of ATP—Conservation conditioned hearts make it impossible to reach
of the Moieties a firm conclusion at this time.
The first difference observed between the We have demonstrated that the depletion
ischemic metabolism of preconditioned and of ATP during ischemia is independent of
nonpreconditioned hearts was the preservation postischemic functional recovery as long as the
of the ATP pool in preconditioned hearts.55-57 adenine nucleotide pool remains large enough
This discovery was already made before to maintain sufficient ATP turnover upon
ischemic preconditioning got its name.56 The reoxygenation.62,63 The preservation of the key
authors proposed that the tissue ATP content components of metabolism (moieties), e.g.,
could be further depleted if an ischemic epi- ATP, and adenine nucleotides is a fundamen-
sode was divided into several short periods of tal principle of metabolism63 which can also
ischemia interrupted by a short episode of be incorporated into the concepts governing
reperfusion. The hypothesis was based on the the mechanism of ischemic preconditioning.
observation that during a long period of
ischemia ATP depletion occurred in an expo-
nential fashion. The results were unexpected.
110 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 10.2. Documented effects of ischemic preconditioning on metabolism,


contractile function, and ion homeostasis

Preservation of ATP1,139
Inhibition of mitochondrial F0F1-ATPase60
Depletion of glycogen48,65
Reduction in lactate accumulation48,50,139
Attenuation of acidosis48,57,67,73
Decreased accumulation of long chain acyl-carnitine138
Decreased generation of oxygen-derived free radicals10
Increased glycolytic rate18
Decreased glycolytic rate48,50,57,73,140
Decreased and delayed ischemic contracture10,50
Increased and early onset of ischemic contracture4,49,51
Protection against stunning12,141
Reduction of Ca2+ and Na+ overloading91,92
Inhibition of Na+/H+ exchange91,92

Table 10.3. Suggested mechanisms of ischemic preconditioning: metabolic concepts

Approach Mechanism of Action Pro Con

- ATP-preservation preservation Murry et al 1986,19901,57


of energy charge Jennings et al 1991142
Yellon et al 199324
Abd-Elfattah et al 199555

- due to inhibition Vuorinen et al 199560 Kobara et al 1996140


of F0F1 ATPase

- Glycogen depletion reduction of H+ and Asimakis et al 199250 Schaefer et al 199567


lactate accumulation Wolfe et al 199348 Asimakis 199674
Reimer et al 199461 Doenst et al 1996,19974,47
Barbosa et al 199665 King and Opie 1996143

- Reduced anaerobic reduction of H+ and Murry et al 199057 Fralix 1992144


glycolysis lactate accumulation Asimakis et al 199250 Janier et al 199418
Wolfe et al 199348 Doenst et al 1996,19974,47
Finegan et al 199573
Schaefer et al 199567

- Inhibition of Ca2+ avoid Ca2+ overloading Steenbergen et al 199391


channels Zucchi et al 1995145

- Inhibition of Na+/H+ avoid Na+/Ca2+ Fralix et al 1993146 Ramasamy et al 199592


exchange overloading Steenbergen et al 199391
Bugge and Ytrehus 1995147
Ischemic Preconditioning—from Bench to Bedside 111

Fig. 10.3. Schematic of the signal transduction cascades possibly involved in the mechanism of ischemic precon-
ditioning. Several sequences are included: preconditioning ischemia leads to the release of adenosine, bradykinin,
noradrenaline, and acetylcholine. Most of these mediators activate a Gi-protein which can have several effects.
Adenosine A1-receptor activation leads to activation of phospholipase D (PLD) which gives rise to phosphatidic
acid (PA) from phosphatidylcholine (PCh). PA is metabolized to diacyl-glycerol (DAG), which is the primary
activator of protein kinase C (PKC). The muscarinic M2-receptors and the alpha-adrenergic receptor lead to the
activation of phospholipase C (PLC) which gives rise to DAG and inositol-1,4,5,-trisphosphate (IP3) from
phosphinositolphosphate (PIP2). IP3 leads to the release of Ca2+ from the sarcoplasmic reticulum (SR) which also
activates PKC. G-proteins also reduce the sensitivity of K+-channels for ATP. A reduction in cellular ATP would
result in the opening of the channel and a shortening of the action potential. Finally, Gi proteins reduce the activity
of adenylate cyclase activity which suppresses cAMP production. Stimulation of the bradykinin receptor (B2-
receptor) causes an increase in intracellular cGMP which activates a cGMP-sensitive phosphodiesterase. Both the
reduction in cAMP and the increase in cGMP cause an inhibition of the L-type Ca2+-channel and thereby reduce
Ca2+ overloading. Preconditioning ischemia also leads to translocation of glucose transporters to the cell membrane
increasing the glucose transport capacity. Inhibition of the FoF1-ATPase which is responsible for a major part of
ischemic ATP breakdown may also account for the ATP-sparing effect.

The Glycogen Hypothesis— infarct size. They suggested that preischemic


An Epiphenomenon? glycogen depletion by preconditioning-is-
The observation that ischemic precondi- chemia reduced substrate availability for
tioning depletes preischemic glycogen content, anaerobic glycolysis during ischemia, thus re-
reduces lactate accumulation, and attenuates ducing lactate and H+ production, and thereby
intracellular acidosis led to a reexamination of attenuating acidosis. At least three observations
the hypothesis that H+ could replace Ca2+ on argue against this hypothesis.
the myofilaments64 which would result in First, we demonstrated in the isolated
impaired contractile function and ischemic working rat heart model that the protection
injury. Wolfe and his co-workers 48,65 afforded by ischemic preconditioning is
demonstrated a positive correlation between independent from the preischemic glycogen
preischemic tissue glycogen and postischemic content.4,47 We also demonstrated that lactate
accumulation does not affect postischemic
112 Ischemia-Reperfusion Injury in Cardiac Surgery

recovery of function. Secondly, if glycogen ischemia relies on indirect measures, such as


depletion were involved in the mechanism of glycogen depletion or lactate accumulation, or
ischemic preconditioning, depletion of both, and does not provide changes as a func-
preischemic glycogen without ischemia (e.g., tion of time or actual rates.
by anoxia, or by substrate-free perfusion) However, if the rate of anaerobic glycoly-
should also result in protection. We66 and oth- sis is reduced in preconditioned hearts, so is
ers67 were unable to confirm this hypothesis ATP production. Preservation of ATP is there-
in isolated rat hearts. fore only possible if energy consumption is
In contrast, we demonstrated that raising decreased before ATP is depleted. Possible
glycogen before the onset of ischemia was mechanisms could include an inhibition of the
beneficial for the return of postischemic con- mitochondrial F0F1 ATPase60 (a major culprit
tractile function4,68-70 and that the addition of for ischemic ATP hydrolysis) or an early
lactate or lactate and insulin to the perfusate reduction of contractile function. Another pos-
before or after ischemia improved postischemic sibility is an increased rate of anaerobic glyco-
recovery.4,66 The third argument against the lysis maintaining residual ATP generation.76
glycogen hypothesis is the fact that pharma- The latter proposal is opposed by the major-
cological preconditioning does not deplete ity of studies reporting decreased lactate pro-
preischemic glycogen. 71 We therefore con- duction and attenuation of acidosis during
clude that glycogen depletion through ischemia.35,50,67,72-74 Thus, there appears to be
ischemic preconditioning appears to be an some intervening influence to decrease con-
epiphenomenon. tractile function before total ATP depletion.
This influence is even more necessary in the
Glycolysis—A Major Player setting of ischemic preconditioning if anaero-
in Ischemic Preconditioning? bic glycolysis is indeed decreased (the major
The majority of the studies investigating source of ATP production during ischemia)
glucose metabolism in preconditioned hearts and ATP levels in the cell are maintained. In
suggest that the glycolytic rate during sustained this context, reduction of lactate accumula-
ischemia is decreased which results in less lac- tion and attenuation of acidosis may just be
tate and proton production implementing the consequence of reduced energy demand.
attenuation of acidosis for the protective ef-
fects of ischemic preconditioning.48,50,57,67,72-74 Delivery of Preconditioning—
In contrast, one group of investigators18 How Is Protection Mediated?
demonstrated increased glycolytic flux during Despite extensive investigation of signal
ischemia after preconditioning in a model of transduction pathways and other possible
low-flow ischemia, and another group reports mediators, the exact mechanisms underlying
an inverse relationship between the rate of gly- ischemic preconditioning remain elusive. Sev-
colysis and ischemic injury.75 The anaerobic eral hypotheses have been advanced.
production of cytosolic ATP during ischemia
has been linked to the maintenance of ion- At least one of the mechanisms of ischemic
pump activity in the sarcolemma for the pres- preconditioning can be triggered by the inter-
ervation of cellular integrity.76 In addition, action between endogenous factors produced
adenosine increases glycolytic flux through an within the heart and cardiac myocytes. Oth-
A1-receptor-dependent pathway.77 The same erwise, the efficacy of ischemic precondition-
receptor may be involved in the transfer of the ing could not be explained in isolated
ischemic preconditioning stimulus to the cel- hearts?4,19,47,73 In addition, it may well be that
lular level,43 (see below). remotely produced factors activate the same
It is difficult to resolve this controversy. or a similar response in the heart. 78, 79
Low-flow ischemia differs from total ischemia. McClanahan et al79 demonstrated that a prior
Assessment of glycolytic rates during total episode of renal ischemia and reperfusion
reduced myocardial infarct size in rabbits.
Ischemic Preconditioning—from Bench to Bedside 113

Several endogenous ischemic byproducts fects in several ways: 1) by activation of ATP


have been identified which may have the sensitive K+ channels98 (see below), 2) by trans-
potential to mediate protection against location of protein kinase C89 which is believed
ischemic injury of the heart, whether in a to be initiated by activation of phospholipase
paracrine or an endocrine fashion. These D99, 100 (see below), 3) by inhibition of cat-
mediators generally exert their effects through echolamine action through activation of Gi
the interaction with specific receptors and the proteins and reduction of adenylate cyclase
activation of transduction cascades. The most activity,101 4) by the reduction of norepineph-
widely discussed candidates are adenos- rine release from sympathetic nerve endings102
ine12,17,19,31,38,60,80 and noradrenaline.21,81-84 (in contrast to a1 adrenergic receptor hypoth-
Other mediators include bradykinin,85,86 esis, see below), 5) by blockade of cardio-
prostanoids, 5,29 endorphins, 87 acetylcho- myocyte L-type Ca2+ channels,93,94 6) by the
line,30,88 and oxygen derived free radicals.20,57 stimulation of glucose uptake and utilization.77
The “delivery systems” associated with the At this time, it is unclear which of these mecha-
individual receptors are believed to be protein nisms are predominant. Since adenosine re-
kinase C,89 the ATP sensitive K+ channel,90 the lease during reperfusion ceases after the first
Na+H+ exchanger,91,92 and the L-type Ca2+ cycle, a logical problem emerges when adenos-
channel.93,94 Figure 10.3 summarizes the most ine release is implicated in the mechanism of
commonly advanced hypotheses into a simple preconditioning in a model where several
schematic. In this scheme adenosine and cycles of preconditioning/reperfusion are nec-
noradrenaline are endogenous mediators. Pro- essary for protection. This effect can be ob-
tein kinase C and K+ ATP channels are pos- served in pigs103 and rats.45 Hence, it is not
sible effectors distal to the receptor. The con- surprising that certain studies were not able
cepts are discussed in detail in the following to confirm the adenosine hypothesis,104-106 and
section. Please refer to Table 10.4 and Fig- some investigators even question whether
ure 10.3 for summary information. adenosine is involved in the mechanism of
ischemic preconditioning.107,108
Two Possible Endogenous Mediators An explanation for these discrepancies may
of Protection come from the different animal species used
or may be provided by the threshold hypoth-
Adenosine—Sufficient but not esis of Downey and his colleagues.45,71 Accord-
Mandatory for Preconditioning ing to this hypothesis, the stimulus provided
The increase in tissue AMP during by adenosine release may not be sufficient to
ischemia causes an increased production of induce the protective effects and may require
adenosine through 5' nucleotidase. The gen- supplementation by other mediators. Vice
eration of adenosine takes place within min- versa, if the adenosine stimulus is blocked, e.g.,
utes and its release during preconditioning through an A1-receptor blocker, the stimula-
reperfusion with subsequent activation of A1 tion of the protective effects through alterna-
or A3 or both adenosine receptors is thought tive mediators has to be increased or protec-
to be involved in the mechanism of ischemic tion will not be detectable. In conclusion,
preconditioning.4,15,19,95,96 Evidence for this endogenous adenosine appears to be sufficient
hypothesis was provided by experiments where but not mandatory for the induction of
selective A1 receptor agonists (but not A2 ischemic preconditioning.
receptor agonists) could mimic the reduction
in infarct size,38,97 while the addition of Noradrenaline and the !1-Receptor—
adenosine receptor blockers (8-p-sulpho- Independent from Adenosine?
phenyl theophylline or PD115199) prevented Other investigators have demonstrated that
the infarct size reduction normally observed ischemic preconditioning may be mediated by
with ischemic preconditioning80 (Fig. 10.4). increased release of norepinephrine from
Adenosine may exert its cardioprotective ef- autonomic nerve terminals.21,81,82 On the ba-
sis of selective !1-adrenergic blockade and
114 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 10.4. Suggested mechanisms of ischemic preconditioning: Pharmacological agents,


receptors, and intracellular mediators

Pharmacological Receptor Mechanism Pro Con


Agent of Action

- Adenosine: A1 receptor activation of Gi protein Kirsch et al 199098 Cave et al 1993106


PKC-translocation Liu et al 1991-199419,80 Hale et al 1993105
A3 receptor K+ATP channel- Downey et al 199338 Hendrikx et al 1993104
opening Lasley, Mentzer 1993148 Li and Kloner 1993108
Burns et al 199554 Liu and Downey 1993149
Kerensky et al 199532 Armstrong et al 199495
Schulz et al 199531 Yabe et al 199528
Woolfson et al 199596
Auchampach, Gross 1993119

- Noradrenaline: !1 receptor translocation of PKC Banerjee et al 199381 Richardt et al 1987102


Mitchell et al 199583 Thornton et al 1993123
Toombs et al 199321 Bugge, Ytrehus 1995150
Tsuchida et al 199484 Weselcouch et al 1995151
Burns et al 199554
Kitakaze et al 199582

- Bradykinin: BK2 receptor translocation of PKC Vegh et al 1994, 19955,29 Bugge, Ytrehus 1996152
Nitric oxide Wall et al 199486

- Prostanoids: Nitric oxide Vegh et al 1990153 Weselcouch et al 1995154


Woolfson et al 199596

- Endothelin: translocation of PKC Wang et al 1996155

- Acetylcholine M2-receptor activation of Gi proteins Yao and Gross 199330


Hendrikx et al 1993156
Przyklenk et al 199588

- Morphine opiate activation of K+ATP Schultz et al 1995,199687,157


receptor channel

- Angiotensin II ATII receptor translocation of PKC Liu et al 1995158

- K+ATP channel- direct activa- Auchampach et al Kitzen et al 1992124


openers tion of K+ATP 1991,1992121,122 Grover et al 1993160
channel Cole et al 19933 Thornton et al 1993123
Steenbergen et al 199391 Yabe et al 199528
Schulz et al 199415
Tomai et al 199422
Mizumura et al 1995159
Hu et al 1996112

- PKC activators direct translo- Liu et al 1994161 Armstrong et al 199495


cation and Speechly-Dick et al 1994117 Vogt et al 1994118
activation of Ytrehus et al 199489 Przyklenk et al 199514
PKC Mitchell et al 199583
Hu et al 1996112
Kitakaze et al 1996162
Ischemic Preconditioning—from Bench to Bedside 115

Fig. 10.4. Prevention of the preconditioning-induced reduction in infarct size by the adenosine receptor antago-
nists 8-p-sulphophenyl theophylline (SPT) and PD115199 in rabbit hearts subjected to a 30 min occlusion of the
left coronary artery followed by three hours of reperfusion. Preconditioning was induced by a single cycle of 5 min
ischemia and 10 min of reperfusion. * p < 0.001 compared to Control. Reproduced with permission from Liu et
al. Circulation 1991; 84:350-356.

stimulation experiments it was concluded that transduction cascade but are able to acti-
ischemic preconditioning is mediated through vate them independently.
an !1-adrenergic mechanism.81 The activation
of !1-adrenergic receptors leads to the activa- Two Possible Effectors Distal
tion of phospholipase C, which in turn leads to the Receptors
to transient increases in diacylglycerol (DAG)
and inositol-1,4,5-trisphosphate (IP3).109,110 Protein Kinase C—
DAG is the primary physiological activator of The Common Denominator
protein kinase C, a serine-threonine kinase Protein kinase C (PKC) is a promiscuous
implicated in the preconditioning effects83,111 enzyme which is considered to be involved in
(see below and Fig. 10.3). most of the transduction cascades implicated
Some investigators suggest that the with ischemic preconditioning.83,89,112 PKC
!1-adrenoceptor is still capable of reducing
belongs to the serine threonine kinase family.
infarct size despite the presence of an adenos- Its 11 or more isoforms are associated with a
ine receptor blocker.84 Thus, it appears that variety of receptors and physiological effectors.113
both adenosine receptors and ! 1 - PKC participates in the regulation of ion
adrenoceptors share certain parts of the same homeostasis, vascular tone, myocyte contractility,
116 Ischemia-Reperfusion Injury in Cardiac Surgery

gene expression, hypertrophy, and in phospho- K+ATP—A Possible End Effector


rylation of specific cellular proteins.111,113 The of the Transduction Cascades
a, d, and e isoforms are predominant in the The ATP sensitive K+-channel is one of the
heart.83,114 Upon stimulation of e.g., adenos- most abundant ion channels on the sarco-
ine A1-receptors, !1-adrenoceptors, or during lemma. Studies on ischemic preconditioning
ischemia, DAG and IP3 are formed resulting suggest that activation of the K+ATP channel
in the activation and translocation of protein in myocardial fibers limits the detrimental
kinase C to the myocyte membrane.19,83,115 An effects of ischemia, probably because of the
as yet unidentified, membrane bound target shortening of the action potential duration,
protein has been proposed to undergo phos- which in turn results in a reduction in both
phorylation by PKC, thereby inducing the pro- Ca2+ inflow and ATP depletion.3,90,91,119,120
tective effects of ischemic preconditioning.19,89 The mechanism by which the activation of
However, support for the concept that PKC K+ATP-channels during a brief ischemic period
translocation by ischemic preconditioning is protects the myocardium during subsequent
responsible for the reduction of infarct size is ischemic episodes is unknown, but it has been
mainly indirect. In rat and rabbit hearts as well shown in a number of studies and animal spe-
as in rabbit myocyte preparations pretreatment cies that the inhibition of channel opening
with PKC blockers (staurosporin, polymyxin (e.g., with glibenclamide90,119) also abolished
B, or calphostin C) abolishes the protection the protective effects afforded by ischemic pre-
of ischemic preconditioning.19,83,89,95,116,117 conditioning. Vice versa, the stimulation of
Vice versa, the activation of PKC with potent the channel opening was able to mimic pre-
PKC activators (4b-phorbol, 12-myristate, or conditioning.121,122
13-acetate) mimics the cardioprotective effects Although several investigations do not sup-
of preconditioning.19,83,89,95,116,117 Mitchell et port the hypothesis of an involvement of the
al83 demonstrated, with the use of isoform K+ATP channel in the mechanism of ischemic
specific antibodies to PKC in the rat heart, preconditioning,123,124 recent studies suggest
that ischemic preconditioning causes a trans- a role for K+ATP channels in preconditioning
location of the d-isoform to the sarcolemma of human myocardium.22,112 Hu et al112 linked
and of the e-isoform to the nucleus. The the activation of PKC in human and rabbit
authors proposed the d-isoform to be involved myocytes to the opening of ATP-sensitive K+
in the acute protection afforded by ischemic channels. In their study, protein kinase C
preconditioning. reduced the channels’ sensitivity for ATP ren-
In dog hearts, a short period of ischemia dering it open under conditions of decreasing
induced no significant translocation of PKC.14 ATP.
In addition, not all the pharmacological evi-
dence has been supportive of the PKC trans- Preconditioning Without
location hypothesis.14,118 The issue is further
complicated by the demonstration that
Preconditioning?
The search for a common mechanism
blockers of protein kinase C (bisimdolyl,
underlying the phenomenon of ischemic and
maleimide, or staurosporin) elicit their own
pharmacological preconditioning is compli-
effects limiting infarct size.118
cated further by observations that simple meta-
Thus, if PKC is involved in precondition-
bolic or nutritional changes, such as fasting in
ing, the end effector is still unknown. Activa-
vivo, can increase ischemia tolerance in vitro
tion of the K+ ATP channel may be a possibility
to the same extent as ischemic precondition-
(see below). An alternative hypothesis has been
ing.4 As shown in Figure 10.2, the addition of
suggested by Kitakaze et al82 who suggested
lactate and insulin to the perfusion medium
that the activation of PKC precedes and is
of hearts from fasted animals improved post-
responsible for the release of adenosine
ischemic cardiac function to the same extent
through activation of ecto-5'nucleotidase and
as ischemic preconditioning. The two experi-
its translocation to the cell membrane.
Ischemic Preconditioning—from Bench to Bedside 117

mental groups shared two other findings which ing episode when patients were subjected to
were different from the control group. First, the two 3 min episodes of prior cross-clamp-
we observed a reduction of the release of the ing. See, however, our earlier reservations
ischemically accumulated lactate. We sug- about ATP levels as markers for ischemic
gested that the ability to oxidize lactate dur- injury.
ing the reperfusion period is a strong indica- The protective effects induced by ischemic
tor for postischemic return of function. preconditioning seem to be additive to those
Secondly, we observed a biphasic behavior of afforded by cardioplegia or hypothermia or
glucose uptake in the reperfusion period, mea- both.127-131 The additive effects rank from
sured with the positron-emitting glucose tracer overcoming the lack of protection by car-
analogue [ 18 F]-2-deoxy-2-fluoroglucose dioplegia due to impaired delivery of car-
(FDG). An initially high uptake rate was fol- dioplegia,130 through reversing the detrimen-
lowed by a suppression in the late phase. In tal effects of aprotinin on the ischemic
both, ischemically preconditioned hearts and myocardium,131 to the better recovery of the
hearts subjected to the metabolic manipula- implanted, ischemically preconditioned,
tions, there was an early increase in glucose donor hearts.128
uptake compared to control, suggesting an Some investigators, however, suggested
early need for glucose substrate. Other reports from their experiments on sheep hearts that
add support to this hypothesis. Insulin like the use of cardiopulmonary bypass alone is
growth factor (IGF2)114 as well as the calcito- stimulus enough to elicit the preconditioning
nin gene-related peptide125 mimic the protec- effects.54 The authors compared infarct size of
tive effects of ischemic preconditioning. Both preconditioned and nonpreconditioned hearts
agents are known for their stimulating effect with or without the use of cardiopulmonary
on glucose uptake. Ischemia also results in the bypass.
translocation of GLUT-1 and GLUT-4 trans- It is certainly desirable to “bottle” the dif-
porters to the plasma membrane.126 The pro- ferent variants of protection afforded by the
tective role of increased glucose uptake is not mechanisms of ischemic preconditioning. We
new, but it is often overlooked that glucose hope that further research in this field will lead
uptake, and not its supply, is the limiting factor. to the development of safe drugs mimicking
the effect of preconditioning.34,99 However,
Clinical Aspects—Implications characterization of the exact mechanism is a
for the Cardiac Surgeon prerequisite to achieve this goal.
Ischemic preconditioning has also been
documented in the human heart.22-26 It can Conclusions
be observed in infarct patients with preceding Ischemic preconditioning protects the
angina, in patients undergoing coronary myocardium from ischemic injury by prob-
angioplasty, as well as in patients undergoing ably more than one mechanism. One of the
surgery with cardiopulmonary bypass and mechanisms involves the release of adenosine
hypothermic ischemic arrest of the myocar- or noradrenaline during preconditioning
dium. One of the most persuasive evidences reperfusion and subsequent translocation of
for the occurrence of ischemic precondition- PKC and opening of K+ATP channels. It appears
ing in human heart is presented by Yellon et that simple metabolic interventions induce
al.24 In this study patients undergoing coro- protection of the myocardium similar to
nary artery bypass grafting were subjected to ischemic preconditioning. Conventional meta-
cross-clamp fibrillation for 10 min with or bolic interventions such as the administration
without two 3 minute episodes of prior aortic of glucose, insulin, and potassium may be
cross clamping. Biopsies taken during the sur- the most valuable for clinical use at the
gery showed significantly higher ATP tissue moment because their safety has been docu-
contents at the end of the 10 min cross-clamp- mented.132,133 The exploitation of ischemic
118 Ischemia-Reperfusion Injury in Cardiac Surgery

preconditioning for clinical practice will only 10. Tosaki A, Cordis GA, Szerdahelyi P et al.
be possible when the mechanisms of ischemic Effects of preconditioning on reperfusion ar-
rhythmias, myocardial functions, formation of
preconditioning are fully understood. free radicals, and ion shifts in isolated is-
chemic/reperfused rat hearts. J Cardiovasc
Acknowledgments Pharmacol 1994; 23:365-373.
Work from the authors’ laboratory was sup- 11. Turrens JF, Thornton J, Barnard ML et al.
Protection from reperfusion injury by precon-
ported by US Public Health Service Grant No. ditioning hearts does not involve increased
RO1-HL43133. Torsten Doenst was the antioxidant defenses. Am J Physiol 1992;
recipient of a research fellowship from the 262:H585-H589.
German Research Foundation (Deutsche 12. Ogawa T, Miura T, Shimamoto K et al. Ac-
tivation of adenosine receptors before is-
Forschungsgemeinschaft, DFG). We thank chemia enhances tolerance against myocardial
Sonya Carmical for help with the preparation stunning in the rabbit heart. J Am Coll
of the manuscript. Cardiol 1996; 27:225-233.
13. Murry CE, Richard VJ, Jennings RB et al.
Myocardial protection is lost before contrac-
tile function recovers from ischemic precon-
References ditioning. Am J Physiol 1991; 260:1796-
1. Murry CE, Jennings RB, Reimer KA. Pre- 1804.
conditioning with ischemia: A delay of lethal 14. Przyklenk K, Sussman MA, Simkhovich BZ
cell injury in ischemic myocardium. Circula- et al. Does ischemic preconditioning trigger
tion 1986; 74:1124-1136. translocation of protein kinase C in the ca-
2. Cohen MW, Liu GS, Downey JM. Precon- nine model? Circulation 1995; 92:1546-1557.
ditioning causes improved wall motion as well 15. Schulz R, Rose J, Heusch G. Involvement of
as smaller infarcts after transient coronary activation of ATP-dependent potassium chan-
occlusion in rabbits. Circulation 1991; nels in ischemic preconditioning in swine. Am
84:341-349. J Physiol 1994; 267:H1341-H1352.
3. Cole WC. ATP-sensitive K+ channels in car- 16. Schulz R, Post H, Sakka S et al. Intraischemic
diac ischemia: An endogenous mechanism for preconditioning. Increased tolerance to sus-
protection of the heart. Cardiovasc Drugs tained low-flow ischaemia by a brief episode
Ther 1993; 7 Suppl 3:527-537. of no-flow ischaemia without intermittent
4. Doenst T, Guthrie PH, Chemnitius J-M et reperfusion. Circ Res 1995; 76:942-950.
al. Fasting, lactate, and insulin improve is- 17. Schott RJ, Rohmans S, Brau ER et al. Is-
chemia tolerance: A comparison with ischemic chemic preconditioning reduces infarct size in
preconditioning. Am J Physiol 1996; 270: swine myocardium. Circ Res 1990; 66:
H1607-H1615. 1133-42.
5. Vegh A, Papp JG, Parratt J. Attenuation of 18. Janier MF, Vanoverschelde JL, Bergmann SR.
the antiarrhythmic effects of ischemic precon- Ischemic preconditioning stimulates anaero-
ditioning by blockade of bradykinin B2 re- bic glycolysis in the isolated rabbit heart. Am
ceptors. Br J Pharmacol 1994; 113:1167- J Physiol 1994; 267:H1353-H1360.
1172. 19. Liu GS, Richards SC, Olsson RA et al. Evi-
6. Parratt JR. Protection of the heart by is- dence that the adenosine A3 receptor may
chemic preconditioning: Mechanisms and mediate the protection afforded by pre-
possibilities for pharmacological exploitation. conditioning in the isolated rabbit heart.
Trends Pharmacol Sci 1994; 15:19-25. Cardiovasc Res 1994; 28:1057-1061.
7. Seyfarth M, Richardt G, Mizsnyak A et al. 20. Tanaka M, Fujiwara H, Yamasaki K et al.
Transient ischemia reduces norepinephrine Superoxide dismutase and N-2-mercap-
release during sustained ischemia. Neural pre- topropionyl glycine attenuate infarct size limi-
conditioning in isolated rat heart. Circ Res tation effect of ischemic preconditioning in
1996; 78:573-580. the rabbit. Cardiovasc Res 1994; 28:980-986.
8. DeFily DV, Chilian WM. Preconditioning 21. Toombs CF, Witte AL, Shebuski RJ. Is-
protects coronary arteriolar endothelium from chemic preconditioning fails to limit infarct
ischemia-reperfusion injury. Am J Physiol size in reserpinized rabbit myocardium: Im-
1993; 265:H700-H706. plications of norepinephrine release in the
9. Jerome SN, Akimitsu T, Gute DC et al. Is- preconditioning effect. Circulation 1993;
chemic preconditioning attenuates capillary 88:2351-2358.
no-reflow induced by prolonged ischemia and
reperfusion. Am J Physiol 1995; 268:H2063-
H2067.
Ischemic Preconditioning—from Bench to Bedside 119

22. Tomai F, Crea F, Gaspardone A et al. Is- 36. Anzai T, Yoshikawa T, Asakura Y et al.
chemic preconditioning during coronary Preinfarction angina as a major predictor of
angioplasty is prevented by glibenclamide, a left ventricular function and long-term prog-
selective ATP-sensitive K+ channel blocker. nosis after a first Q wave myocardial infarc-
Circulation 1994; 90:700-705. tion. J Am Coll Cardiol 1995; 26:319-27.
23. Deutsch E, Berger M, Kussmaul WG et al. 37. Alkhulaifi AM, Pugsley WB, Yellon DM. The
Adaptation to ischemia during percutaneous influence of the time period between precon-
transluminal coronary angioplasty: Clinical, ditioning ischemia and prolonged ischemia on
hemodynamic, and metabolic features. Cir- myocardial protection. Cardioscience 1993;
culation 1990; 82(2044-2051). 4:163-169.
24. Yellon DM, Alkhulaifi AM, Pugsley WB. 38. Downey JM, Liu GS, Thornton JD. Adenos-
Preconditioning the human myocardium. ine and the anti-infarct effects of precondi-
Lancet 1993; 342:276-277. tioning. Cardiovasc Res 1993; 27:3-8.
25. Vaitkus PT, Miller JM, Buxton AE et al. Is- 39. Van Winkle D, Thornton J, Downey D et
chemia-induced changes in human endocar- al. The natural history of preconditioning:
dial electrograms during percutaneous trans- Cardioprotection depends on the duration of
luminal coronary angioplasty. Am Heart J transient ischemia and time to subsequent
1994; 127:1481-1490. ischemia. Coron Art Dis 1991; 2:613-619.
26. Ottani F, Galvani M, Ferrini D et al. Pro- 40. Marber MS, Latchman DS, Walker JM et al.
dromal angina limits infarct size. A role for Cardiac stress protein elevation 24 hours af-
ischemic preconditioning. Circulation 1995; ter brief ischemia or heat stress is associated
91:1667-1668. with resistance to myocardial infarction. Cir-
27. Bolli R, Leesar M, Stoddard M. Ischemic culation 1993; 88:1264-1272.
preconditioning during coronary angioplasty. 41. Knowlton AA, Brecher P, Apstein CS. Rapid
J Am Col Cardiol 1997; 29(2):469. expression of heat shock protein in the rab-
28. Yabe K, Nasa Y, Takeo S. Hypoxic precon- bit after brief cardiac ischemia. J Clin Invest
ditioning in isolated rat hearts: Noninvolve- 1991; 87:139-147.
ment of activation of adenosine A1 receptor, 42. Volovsek A, Subramanian R, Reboussin D.
Gi protein, and ATP-sensitive K+ channel. Effects of duration of ischemia during pre-
Heart & Vessels 1995; 10:294-303. conditioning on mechanical function, enzyme
29. Vegh A, Kaszala K, Papp JG et al. Bradyki- release and energy production in the isolated
nin, a trigger for delayed pacing-induced pre- working rat heart. J Mol Cell Cardiol 1992;
conditioning (abstract). J Mol Cell Cardiol 24:1011-1019.
1995; 27:A279. 43. Lawson CS, Downey JM. Preconditioning:
30. Yao Z, Gross GJ. Role of nitric oxide, mus- State of the art myocardial protection.
carinic receptors, and the ATP-sensitive K+ Cardiovasc Res 1993; 27:542-550.
channel in mediating the effects of acetylcho- 44. Yang XM, Arnoult S, Tsuchida A et al. The
line to mimic preconditioning in dogs. Circ protection of ischemic preconditioning can be
Res 1993; 73:1193-1201. reinstated in the rabbit heart after the initial
31. Schulz R, Rose J, Post H et al. Involvement protection has waned. Cardiovasc Res 1993;
of endogenous adenosine in ischemic precon- 27:556-558.
ditioning in swine. Pflugers Arch-Eur J 45. Liu Y, Downey JM. Ischemic precondition-
Physiol 1995; 430:273-282. ing protects against infarction in rat heart.
32. Kerensky RA, Kutcher MA, Braden GA et Am J Physiol 1992; 263:H1107-H1112.
al. The effects of intracoronary adenosine on 46. Li GC, Vasquez JA, Gallagher KP et al.
preconditioning during coronary angioplasty. Myocardial protection with preconditioning.
Clin Cardiol 1995; 18:91-96. Circulation 1990; 82:609-619.
33. Heibig J, Bolli R, Harris S. Initial coronary 47. Doenst T, Guthrie PH, Taegtmeyer H. Is-
occlusion improves tolerance to subsequent chemic preconditioning in rat heart: No cor-
prolonged balloon inflations. Cath Cardiovasc relation between glycogen content and return
Diag 1989; 16:99-102. of function. J Mol Cell Biochem 1997:in
34. Kloner RA, Przyklenk K, Shook T et al. press.
Clinical aspects of preconditioning and im- 48. Wolfe CL, Sievers RE, Visseren FLJ et al.
plications for the cardiac surgeon. J Card Surg Loss of myocardial protection after precondi-
1995; 10 (4 Suppl):369-375. tioning correlates with the time course of gly-
35. Kloner RA, Przyklenk K, Whittaker P et al. cogen recovery within the preconditioned seg-
Preconditioning stimuli and inadvertent pre- ment. Circulation 1993; 87:881-892.
conditioning. J Mol Cell Cardiol 1995;
27:743-747.
120 Ischemia-Reperfusion Injury in Cardiac Surgery

49. Kolocassides KG, Galinanes M, Hearse DJ. 62. Taegtmeyer H, Roberts AFC, Rayne AEG.
Dichotomy of ischemic preconditioning: Im- Energy metabolism in reperfused rat heart:
proved postischemic contractile function de- Return of function before normalization of
spite intensification of ischemic contracture. ATP content. J Am Coll Cardiol 1985;
Circulation 1996; 93:1725-1733. 6:864-870.
50. Asimakis GK, Inners-McBride K, Medellin G 63. Taegtmeyer H, Goodwin GW, Doenst T et
et al. Ischemic preconditioning attenuates al. Substrate metabolism as a determinant for
acidosis and postischemic dysfunction in iso- postischemic functional recovery of the heart.
lated rat hearts. Am J Physiol 1992; 263: Am J Cardiol 1997:in press.
H887-H894. 64. Katz AM, Hecht HH. The early “pump” fail-
51. Cave AC, Horowitz GL, Apstein CS. Can ure of the ischemic heart. Am J Med 1969;
ischemic preconditioning protect against hy- 47:497-502.
poxia-induced damage? Studies of contractile 65. Barbosa V, Sievers RE, Zaugg CE et al. Pre-
function in isolated perfused rat hearts. J Mol conditioning ischemia time determines the
Cell Cardiol 1994; 26:1471-1486. degree of glycogen depletion and infarct size
52. Ladilov Y, Siegmund B, Balser C et al. Simu- reduction in rat hearts. Am Heart J 1996;
lated ischemia increases the susceptibility of 131:224-230.
rat cardiomyocytes to hypercontracture. Circ 66. Goodwin GW, Taegtmeyer H. Metabolic re-
Res 1997; 80(1):69-75. covery of the isolated working rat heart after
53. Minhaz U, Koide S, Shohtsu A et al. Perfu- brief global ischemia. Am J Physiol 1994;
sion delay causes unintentional ischemic pre- 267:H462-H470.
conditioning in isolated heart preparation. 67. Schaefer S, Carr LJ, Prussel E et al. Effects
Basic Res Cardiol 1995; 90:418-423. of glycogen depletion on ischemic injury in
54. Burns PG, Krukenkamp IB, Caldarone CA isolated rat hearts: Insights into precondition-
et al. Does cardiopulmonary bypass alone ing. Am J Physiol 1995; 268:H935-H944.
elicit myoprotective preconditioning? Circu- 68. Schneider CA, Nguyêñ VTB, Taegtmeyer H.
lation 1995; 92 (9 Suppl):II447-II451. Feeding and fasting determine postischemic
55. Abd-Elfattah AS, Ding M, Wechsler AS. In- glucose utilization in isolated working rat
termittent aortic crossclamping prevents cu- hearts. Am J Physiol 1991; 260:H542-H548.
mulative adenosine triphosphate depletion, 69. Schneider CA, Taegtmeyer H. Fasting in vivo
ventricular fibrillation, and dysfunction delays myocardial cell damage after brief pe-
(stunning): Is it preconditioning? J Thorac riods of ischemia in the isolated working rat
Cardiovasc Surg 1995; 110:328-339. heart. Circ Res 1991; 68:1045-1050.
56. Reimer KA, Hill ML, Jennings RB. Pro- 70. Lagerstrom CF, Walker WE, Taegtmeyer H.
longed depletion of ATP and adenine nucle- Failure of glycogen depletion to improve left
otide pool due to delayed resynthesis of ad- ventricular function of the rabbit heart after
enine nucleotides following reversible myocar- hypothermic ischemic arrest. Circ Res 1988;
dial ischemia injury in dogs. J Mol Cell 63:81-86.
Cardiol 1981; 13:229. 71. Downey JM, Cohen MV. Preconditioning:
57. Murry CE, Richard VJ, Reimer KA et al. Is- Markers vs. epiphenomena. In: Heusch G,
chemic preconditioning slows energy metabo- Schulz R, eds. New Paradigms of Coronary
lism and delays ultrastructural damage dur- Artery Disease. New York: Springer, 1996:
ing a sustained ischemic episode. Circ Res 153-155.
1990; 66:913-931. 72. Liedtke AJ, Renstrom B, Nellis SH et al.
58. Downey JM. Ischemic preconditioning: Mechanical and metabolic functions in pig
Nature’s own cardioprotective intervention. hearts after 4 days of chronic coronary steno-
Trends Cardiovasc Med 1992; 2:170-176. sis. J Am Col Cardiol 1995; 26:815-825.
59. Van der Heide R, Hill M, Steenbergen C et 73. Finegan BA, Lopaschuk GD, Gandhi M et
al. Effect of reversible ischemia on mitochon- al. Ischemic preconditioning inhibits glyco-
drial ATPase activity in canine myocardium. lysis and proton production in isolated work-
Circulation 1991; 84:192. ing rat hearts. Am J Physiol 1995; 269:
60. Vuorinen K, Ylitalo K, Peuhkurinen K et al. H1767-H1775.
Mechanisms of ischemic preconditioning in 74. Asimakis G. Myocardial glycogen depletion
rat myocardium. Roles of adenosine, cellular cannot explain the cardioprotective effects of
energy state, and mitochondrial F1F0-AT- ischemic preconditioning in the rat heart. J
Pase. Circulation 1995; 91:2810-2818. Mol Cell Cardiol 1996; 28:563-570.
61. Reimer KA, Vander Heide RS, Jennings RB. 75. Vanoverschelde JL, Janier MF, Bakke JE et
Ischemic preconditioning slows ischemic me- al. Rate of glycolysis during ischemia deter-
tabolism and limits myocardial infarct size. mines extent of ischemic injury and func-
Ann N Y Acad Sci 1994; 723:99-115. tional recovery after reperfusion. Am J Physiol
1994; 267:H1785-H1794.
Ischemic Preconditioning—from Bench to Bedside 121

76. Weiss J, Hiltbrand B. Functional compart- 89. Ytrehus K, Liu Y, Downey JM. Precondition-
mentation of glycolytic versus oxidative me- ing protects ischemic rabbit heart by protein
tabolism in isolated rabbit heart. J Clin In- kinase C activation. Am J Physiol 1994;
vest 1985; 75:436-447. 266:H1145-H1152.
77. Wyatt DA, Edmunds MC, Rubio R et al. 90. Gross GJ, Auchampach JA. Blockade of ATP-
Adenosine stimulates glycolytic flux in iso- sensitive potassium channels prevents myocar-
lated perfused rat hearts by A1 adenosine re- dial preconditioning in dogs. Circ Res 1992;
ceptors. Am J Physiol 1989; 257:H1952- 70:223-233.
H1957. 91. Steenbergen C, Perlman ME, London RE et
78. Przyklenk K, Bauer B, Ovize M et al. Re- al. Mechanisms of preconditioning: Ionic al-
gional ischemic ‘preconditioning’ protects re- terations. Circ Res 1993; 72:112-125.
mote virgin myocardium from subsequent 92. Ramasamy R, Liu H, Anderson S et al. Is-
sustained coronary occlusion. Circulation chemic preconditioning stimulates sodium
1993; 87:893-899. and proton transport in isolated rat hearts. J
79. McClanahan TB, Nao BS, Wolke LJ et al. Clin Invest 1995; 96:1464-1472.
Brief renal occlusion and reperfusion reduces 93. Eckert R, Utz J, Trautwein W et al. Involve-
myocardial infarct size in rabbits. The FASEB ment of intracellular Ca2+ release mechanism
Journal 1993; 7:682. in adenosine-induced cardiac Ca2+ current
80. Liu GS, Thornton J, Van Winkle DM et al. inhibition. Surgery 1993; 114:334-342.
Protection against infarction afforded by pre- 94. Schrader J, Rubio R, Berne RM. Inhibition
conditioning is mediated by A1 adenosine of slow action potentials of guinea pig atrial
receptors in rabbit heart. Circulation 1991; muscle by adenosine: A possible effect on
84:350-356. Ca2+ influx. J Mol Cell Cardiol 1975; 7:
81. Banerjee A, Locke-Winter C, Rogers KB et 427-433.
al. Preconditioning against myocardial dys- 95. Armstrong S, Downey JM, Ganote CE. Pre-
function after ischemia and reperfusion by an conditioning of isolated rabbit cardiomyo-
!1-adrenergic mechanism. Circ Res 1993; cytes: Induction by metabolic stress and
73:656-670. blockade by the adenosine antagonist SPT
82. Kitakaze M, Hori M, Morioka T et al. Al- and calphostin C, a protein kinase C inhibi-
pha 1-adrenoceptor activation increases ecto- tor. Cardiovasc Res 1994; 28:72-77.
5'-nucleotidase activity and adenosine release 96. Woolfson RG, Patel VC, Neild GH et al.
in rat cardiomyocytes by activating protein Inhibition of nitric oxide synthesis reduces
kinase. Circulation 1995; 91:2226-2234. infarct size by an adenosine-dependent
83. Mitchell MB, Meng X, Ao L et al. Precondi- mechanism. Circulation 1995; 91:1545-1551.
tioning of isolated rat heart is mediated by 97. Tsuchida A, Miura T, Miki T et al. Role of
protein kinase C. Circ Res 1995; 76:73-81. adenosine receptor activation in myocardial
84. Tsuchida A, Liu Y, Liu GS et al. alpha 1- infarct size limitation by ischemic precondi-
adrenergic agonists precondition rabbit is- tioning. Cardiovasc Res 1992; 26:456-461.
chemic myocardium independent of adenos- 98. Kirsch GE, Codina J, Birnbaumer L et al.
ine by direct activation of protein kinase C. Coupling of ATP-sensitive K+ channels to A1
Circ Res 1994; 75:576-585. receptors by G proteins in rat ventricular
85. Miura T. Adenosine and bradykinin: Are they myocytes. Am J Physiol 1990; 259:H820-
independent triggers of preconditioning. In: H826.
Heusch G, Schulz R, eds. New Paradigms 99. Cohen MV, Downey JM. Myocardial precon-
of Coronary Artery Disease. New York: ditioning promises to be a novel approach to
Springer, 1996:138-140. the treatment of ischemic heart disease. Ann
86. Wall TM, Sheehy R, Hartman JC. Role of Rev Medicine 1996; 47:21-29.
bradykinin in myocardial preconditioning. J 100. Cohen M, Liu Y, Liu G et al. Phospholipase
Pharmacol Exp Therapeutics 1994; 270: D plays arole in ischemic preconditioning in
681-689. rabbit heart. Circulation 1996; 94:1713-1718.
87. Schultz JE, Hsu AK, Gross GJ. Morphine 101. Dobson JGJ. Mechanism of adenosine inhi-
mimics the cardioprotective effect of ischemic bition of catecholamine-induced responses in
preconditioning via a glibenclamide-sensitive heart. Circ Res 1983; 52:151-160.
mechanism in the rat heart. Circ Res 1996; 102. Richardt G, Waas W, Kranzhofer R et al.
78:1100-1104. Adenosine inhibits exocytotic release of en-
88. Przyklenk K, Kloner RA. Low-dose i.v. ace- dogenous noradrenalin in rat heart: A pro-
tylcholine acts as a “preconditioning-mimetic” tective mechanism in early myocardial is-
in the canine model. J Card Surg 1995; 10 chemia. Circ Res 1987; 61:117-123.
(Suppl 4):389-395. 103. Sack S, Mohri M, Arras M et al. Ischemic
preconditioning—time course of renewal in
the pig. Cardiovasc Res 1993; 27:551-555.
122 Ischemia-Reperfusion Injury in Cardiac Surgery

104. Hendrikx M, Toshima Y, Mubagwa K et al. 118. Vogt AM, Barancik M, Weihrand D et al.
Improved functional recovery after ischemic Protein kinase C inhibitors reduce infarct size
preconditioning in the globally ischemic rab- in pig hearts in vivo. Circulation 1994;
bit heart is not mediated by adenosine A1 90:I647.
receptor activation. Bas Res Cardiol 1993; 119. Auchampach JA, Gross GJ. Adenosine A1
88:576-593. receptors, KATP channels, and ischemic pre-
105. Hale SL, Bellows SD, Hammerman H et al. conditioning in dogs. Am J Physiol 1993;
An adenosine A1 receptor agonist, R(-)-N-(2- 264:H1327-H1336.
phenylisopropyl)-adenosine (PIA), but not 120. Noma A. ATP-regulated K+ channels in car-
adenosine itself, acts as a therapeutic precon- diac muscle. Nature 1983; 305:147-148.
ditioning-mimetic agent in rabbits. Cardiovasc 121. Auchampach JA, Maruyama M, Cavero I et
Res 1993; 27:2140-2150. al. The new K+ channel opener Aprikalim (RP
106. Cave CC, Collis CS, Downey JM et al. Im- 52891) reduces experimental infarct size in
proved functional recovery by ischemic pre- dogs in the absence of hemodynamic changes.
conditioning is not mediated by adenosine in J Pharmacol Exp Ther 1991; 259:961-967.
the globally ischemic isolated rat heart. 122. Auchampach JA, Grover GJ, Gross GJ.
Cardiovasc Res 1993; 27:663-668. Blockade of ischemic preconditioning in dogs
107. Li Y, Kloner RA. Adenosine deaminase inhi- by the novel ATP dependent potassium chan-
bition is not cardioprotective in the rat. Am nel antagonist sodium 5-hydroxydecanoate.
Heart J 1993; 126:1293-1298. Cardiovasc Res 1992; 26:1054-1062.
108. Li Y, Kloner RA. The cardioprotective efforts 123. Thornton JD, Daly JF, Cohen MV et al.
of ischemic preconditioning are not mediated Catecholamines can induce adenosine recep-
by adenosine receptors in rat hearts. Circula- tor-mediated protection of the myocardium
tion 1993; 87:1642-1648. but do not participate in ischemic precondi-
109. Fedida D, Braun AP, Giles WR. Alpha 1- tioning in the rabbit. Circ Res 1993; 73:
adrenoceptors in myocardium: Functional as- 649-655.
pects and transmembranes signaling mecha- 124. Kitzen J, McCallum J, Harvey C et al. Po-
nisms. Physiol Rev 1993; 73:469-487. tassium channel activators cromakalim and
110. Terzic A, Puceat M, Vassort G et al. Car- celikalim (WAY-120-491) fail to decrease
diac alpha 1-adrenoceptors: An overview. myocardial infarct size in the anesthetized
Pharmacol Rev 1993; 45:147-175. canine. Pharmacology 1992; 45:71-82.
111. Nishizuka Y. Intracellular signaling by hy- 125. Li YJ, Xiao ZS, Peng CF et al. Calcitonin
drolysis of phospholipids and activation of gene-related peptide-induced preconditioning
protein kinase C. Science 1992; 1992: protects against ischemia-reperfusion injury in
607-614. isolated rat hearts. Eur J Pharmacol 1996;
112. Hu K, Duan D, Li GR et al. Protein kinase 311:163-167.
C activates ATP-sensitive K+ current in hu- 126. Young LH, Renfu Y, Russell R et al. Low-
man and rabbit ventricular myocytes. Circ flow ischemia leads to translocation of canine
Res 1996; 78:492-498. heart GLUT-4 and GLUT-1 glucose trans-
113. Johnson JA, Mochly-Rosen D. Inhibition of porters to the sarcolemma in vivo. Circula-
the spontaneous rate of contraction of neo- tion 1997; 95:415-422.
natal cardiac myocytes by protein kinase C 127. Ogino H, Smolenski RT, Zych M et al. In-
isozymes. A putative role for the epsilon fluence of preconditioning on rat heart sub-
isozyme. Circ Res 1995; 76:654-663. jected to prolonged cardioplegic arrest. Ann
114. Schaper W. Ischemic preconditioning, re- Thorac Surg 1996; 62:469-474.
membrances of things past and future. Bas 128. Karck M, Rahmanian P, Haverich A. Is-
Res Cardiol 1996; 91:8-11. chemic preconditioning enhances donor heart
115. Strasser RH, Braun-Dullaeus R, Walendzik H preservation. Transplantation 1996; 62:17-22.
et al. Alpha 1-receptor-independent activation 129. Menasche P, Kevelaitis E, Mouas C et al.
of protein kinase C in acute myocardial is- Preconditioning with potassium channel
chemia. Mechanisms for sensitization of the openers. A new concept for enhancing cardio-
adenylyl cyclase. Circ Res 1992; 70:1304- plegic protection? J Thor Cardiovasc Surg
1312. 1995; 110:1606-1613.
116. Li Y, Kloner RA. Cardioprotective effects of 130. Galinanes M, Argano V, Hearse DJ. Can is-
ischemic preconditioning can be recaptured chemic preconditioning ensure optimal myo-
after they are lost. J Am Coll Cardiol 1994; cardial protection when delivery of cardiople-
23:470-474. gia is impaired? Circulation 1995; 92 (Suppl):
117. Speechley-Dick ME, Mocanu MM, Yellon II389-II394.
DM. Protein kinase C. Its role in ischemic
preconditioning in the rat. Circ Res 1994;
75:586-590.
Ischemic Preconditioning—from Bench to Bedside 123

131. Bukhari EA, Krukenkamp IB, Burns PG et 145. Zucchi R, Ronca-Testoni S, Yu G et al. Pos-
al. Does aprotinin increase the myocardial tischemic changes in cardiac sarcoplasmic
damage in the setting of ischemia and pre- reticulum Ca channels. A possible mechanism
conditioning? Ann Thorac Surg 1995; 60: of ischemic preconditioning. Circ Res 1995;
301-310. 76:1049-1056.
132. Sodi-Pallares D, Bisteni A, Medrano G et al. 146. Fralix TA, Murphy E, London RE et al. Pro-
The polarizing treatment of acute myocardial tective effects of adenosine in the perfused
infarction. Dis Chest 1963; 43:424-432. rat heart: Changes in metabolism and ion
133. Gradinak S, Coleman GM, Taegtmeyer H et homeostasis. Am J Physiol 1993; 264:C986-
al. Improved cardiac function with glucose- C994.
insulin-potassium after coronary bypass sur- 147. Bugge E, Ytrehus K. Inhibition of sodium-
gery. Ann Thorac Surg 1989; 48:484-489. hydrogen exchange reduces infarct size in the
134. Iwamoto T, Bai X-J, Downey HF. Precondi- isolated rat heart—a protective additive to
tioning with supply-demand imbalance lim- ischemic preconditioning. Cardiovasc Res
its infarct size in dog heart. Cardiovasc Res 1995; 29:269-274.
1993; 27:2071-2076. 148. Lasley R, Mentzer R. Pertussis toxin blocks
135. Ovize M, Kloner RA, Przyklenk K. Stretch adenosine A1 receptor mediated protection of
preconditions canine myocardium. Am J the ischemic heart. J Mol Cell Cardiol 1993;
Physiol 1994; 266:H137-H146. 25:815-821.
136. Shiki K, Hearse DJ. Preconditioning of is- 149. Liu Y, Downey J. Preconditioning against
chemic myocardium: Reperfusion-induced infarction in the rat heart does not involve a
arrhythmias. Am J Physiol 1987; 253:H1470- pertussis toxin sensitive G protein. Cardiovasc
H1476. Res 1993; 27:608-611.
137. Goto M, Tsuchida A, Liu Y et al. Transient 150. Bugge E, Ytrehus K. Ischemic precondition-
inhibition of glucose uptake mimics ischemic ing is protein kinase C dependent but not
preconditioning by salvaging ischemic myo- through stimulation of alpha adrenergic or
cardium in the rabbit heart. J Mol Cell adenosine receptors in the isolated rat heart.
Cardiol 1995; 27:1883-1894. Cardiovasc Res 1995; 29:401-406.
138. Simkhovich BZ, Hale SL, Ovize M et al. Is- 151. Weselcouch E, Baird A, Sleph P et al. En-
chemic preconditioning and long-chain acyl dogenous catecholamines are not necessary for
carnitine in the canine heart. Cor Art Dis- ischemic preconditioning in the isolated per-
ease 1993; 4:387-392. fused rat heart. Cardiovasc Res 1995; 29:
139. Jennings RB, Murry CE, Reimer KA. Energy 126-132.
metabolism in preconditioned and control 152. Bugge E, Ytrehus K. Bradykinin protects
myocardium: Effect of total ischemia. J Mol against infarction but does not mediate is-
Cell Cardiol 1991; 23:1449-1458. chemic preconditioning in the isolated rat
140. Kobara M, Tatsumi T, Matoba S et al. Ef- heart. J Mol Cell Cardiol 1996; 28:2333-
fect of ischemic preconditioning on mito- 2341.
chondrial oxidative phosphorylation and high 153. Vegh A, Szekeres L, Parrat J. Protective ef-
energy phosphates in rat hearts. J Mol Cell fects of preconditioning on the ischemic myo-
Cardiol 1996; 28:417-428. cardium involve cyclo-oxygenase products.
141. Sun JZ, Tang XL, Knowlton AA et al. Late Cardiovasc Res 1990; 24:1020-1022.
preconditioning against myocardial stunning. 154. Weselcouch EO, Baird AJ, Sleph P et al. In-
An endogenous protective mechanism that hibition of nitric oxide synthesis does not
confers resistance to postischemic dysfunction affect ischemic preconditioning in isolated
24 h after brief ischemia in conscious pigs. J perfused rat hearts. Am J Physiol 1995;
Clin Invest 1995; 95:388-403. 268:H242-H249.
142. Jennings R, Murry C, Reimer K. Precon- 155. Wang P, Gallagher K, Downey J et al. Pre-
ditioning myocardium with ischemia. treatment with endothelin-1 mimics ischemic
Cardiovasc Drugs Ther 1991; 5:933-938. preconditioning against infarction in isolated
143. King L, Opie L. Does preconditioning act by rabbit heart. J Mol Cell Cardiol 1996;
glycogen depletion in the isolated rat heart? 28:577-588.
J Mol Cell Cardiol 1996; 28:2305-2321. 156. Hendrikx M, Toshima Y, Mubagwa K et al.
144. Fralix TA, Steenbergen C, London RE et al. Muscarinic receptor stimulation by carbachol
Metabolic substrates can alter postischemic improves functional recovery in isolated,
recovery in preconditioned ischemic heart. blood perfused rabbit heart. Cardiovasc Res
Am J Physiol 1992; 263:C17-C23. 1993; 27:980-989.
157. Schultz JE, Rose E, Yao Z et al. Evidence
for involvement of opioid receptors in is-
chemic preconditioning in rat hearts. Am J
Physiol 1995; 268:H2157-H2161.
124 Ischemia-Reperfusion Injury in Cardiac Surgery

158. Liu Y, Tsuchida A, Cohen MV et al. Pre- 161. Liu Y, Cohen MV, Downey JM. Chelery-
treatment with angiotensin II activates pro- thrine, a highly selective protein kinase C
tein kinase C and limits myocardial infarc- inhibitor, blocks the anti-infarct effect of is-
tion in isolated rabbit hearts. J Mol Cell chemic preconditioning in rabbit hearts.
Cardiol 1995; 27:883-892. Cardiovasc Drugs Ther 1994; 8:881-882.
159. Mizumura T, Nithipatikom K, Gross GJ. 162. Kitakaze M, Node K, Minamino T et al. Role
Bimakalim, an ATP-sensitive potassium chan- of activation of protein kinase C in the inf-
nel opener, mimics the effects of ischemic arct size-limiting effect of ischemic precondi-
preconditioning to reduce infarct size, adenos- tioning through activation of ecto-5'-nucle-
ine release, and neutrophil function in dogs. otidase. Circulation 1996; 93:781-791.
Circulation 1995; 92:1236-1245.
160. Grover G, Dzwonczyk S, Sleph P et al. The
ATP-sensitive potassium channel blocker
glibenclamide (glyburide) does not abolish
preconditioning in isolated ischemic rat
hearts. J Pharmacol Exp Ther 1993; 265:
559-564.
CHAPTER 11

Away from Ischemic Preconditioning


and Towards Pharmacological
Preconditioning
Louis P. Perrault and Philippe Menasché

Abstract regardless of the various components of the

E
ndogenous myocardial protection intracellular signaling pathway elicited by the
refers to the natural defense preconditioning stimulus, it seems that a major
mechanisms available to the heart to mechanism by which this pathway leads to a
withstand an ischemic injury. So far these cardioprotective effect is a slowing of ATP
mechanisms have been shown to encompass depletion during the protracted period of
two phenomena most likely interrelated: ischemia. If the latter is true, then it can rea-
ischemic preconditioning and stress protein sonably be predicted that this energy-sparing
synthesis. Ischemic PC can be defined as the effect may become redundant to that of car-
adaptive mechanism induced by a brief period dioplegia. These problems emphasize the
of reversible ischemia increasing the heart’s importance of identifying the mechanisms
resistance to a subsequent longer period of underlying endogenous myocardial protection
ischemia. Two different time frames are in an attempt to pharmacologically duplicate
defined for preconditioning, one early or clas- the protective action of ischemically induced
sical preconditioning and one late also called preconditioning. Numerous triggers have been
the second window of preconditioning. The identified as being able to elicit and reproduce
therapeutic exploitation of these natural adap- the effects of ischemic preconditioning. The
tive mechanisms in cardiac surgery is an role of ATP-sensitive potassium channels as
appealing prospect. Preconditioning could be mediators of the cardioprotective effects of
used before aortic cross-clamping to enhance preconditioning has been largely based on the
the current methods of myocardial protection. observations that these effects could be dupli-
Two major conclusions emerge from the bulk cated by potassium channel openers (PCO)
of experimental data on preconditioning. First, whereas they were abolished by potassium
the adaptive phenomenon reduces infarct size channel blockers. PCO given before potassium
after regional ischemia in animal preparations arrest have the ability to enhance functional
across a wide variety of species but its effects recovery during reperfusion and may be inde-
on arrhythmias and on preservation of func- pendent of the temperature of the cardiople-
tion after global ischemia are less consistent. gia administered. Currently the application of
This is relevant to cardiac surgery where these drugs is hampered by the fact that the
postbypass pump failure is more often due to anti-ischemic effects are obtained at much
stunning than to discrete necrosis. Second, higher doses than those required for relaxation

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


126 Ischemia-Reperfusion Injury in Cardiac Surgery

of smooth muscle and thus could cause discussing pharmacological mediators of


important hemodynamic effects due to vasodi- endogenous myocardial protection.
latation before the appearance of the cardio-
protective effects. The modalities of pharma- Clinical Trials:
cologically-induced preconditioning, although An Apparent Discrepancy
intellectually appealing, remain to be deter-
Clinically, the first use of preconditioning
mined including the choice of the optimal
(PC) has been reported by Alkulaifi and
agents and regimens. The only clinical situa-
co-workers2 using conservation of adenosine
tion in which the ischemic preconditioning
triphosphate (ATP) as the major end point.
remains logical would be “off-pump” mini-
Twenty patients were randomised to either
mally invasive cardiac surgery as the occlusion
preconditioning by two 3-minute periods of
of the target vessel during construction of the
cross-clamping separated by 2 minutes of
distal anastomosis most closely mimics the
reperfusion prior to an ischemic insult of 10
experimental scenario that has turned out to
minutes ischemia and ventricular fibrillation
result in reduction of regionally-induced
done at a temperature of 37°C. The ten con-
ischemic damage.
trols underwent 10 minutes of cross-clamp-
Endogenous myocardial protection mecha-
ing with fibrillation without preconditioning.
nisms are available to the heart to withstand
The major finding of this study was that pre-
an ischemic injury. So far clinical and basic
conditioning slowed the rate of ATP deple-
research have shown that these mechanisms
tion to such an extent that at the end of the
include two phenomena most likely interre-
10 minute period, the preconditioned group
lated: ischemic preconditioning and stress pro-
had a significantly higher ATP content than
tein synthesis. Ischemic preconditioning is the
controls. The authors should be credited for
adaptive mechanism induced by a brief period
demonstrating the adaptive response of the
of reversible ischemia increasing the heart’s
human heart to an ischemic stress. However,
resistance to a subsequent longer period of
a major limitation of this study was that the
ischemia. Two different time frames are
normothermic ventricular fibrillation tech-
defined for preconditioning, one early (or
nique studied is not commonly used in many
“classical” preconditioning) which involves
centers (where it is usually employed with a
activation of various membrane receptors and
moderate degree of hypothermia) and likely
one late (termed the second window of pre-
resulted in extensive myocardial injury which
conditioning) which is related to changes in
provided room for improvement from the
gene expression leading to the synthesis of
ischemic stimulus used as preconditioning.
cardioprotective stress proteins.1,2 Exploitation
of these natural adaptive mechanisms in car- Thus to assess whether this type of
diac surgery is an appealing prospect since cardioprotective intervention would be rel-
preconditioning in one form or another could evant to cardioplegic arrest, we4 studied the
be used during aortic cross-clamping or local effects of ischemic preconditioning (achieved
control of target vessels to optimize the effi- with 3 minutes of aortic cross-clamping with
cacy of current methods of myocardial pro- 2 minutes of reperfusion before the onset of
tection. However, major questions have to be cardioplegic arrest under cardiopulmonary
answered before preconditioning can be used bypass support) before continuous retrograde
appropriately in patients undergoing heart normothermic blood cardioplegia in twenty
surgery. Are these endogenous protection patients undergoing coronary artery bypass
mechanisms relevant to surgically-induced operations. Ten case-matched patients serving
myocardial ischemia? How can they be applied as controls, underwent a 5-minute period of
during open-heart procedures and which situ- bypass before the induction of cardioplegic
ations and patients are they best indicated for? arrest. At the end of arrest, the release of
Data concerning ischemic preconditioning in CK-MB from the myocardium (calculated as
the early phase will first be addressed before the difference between coronary sinus and
Away from Ischemic Preconditioning and Towards Pharmacological Preconditioning 127

radial artery values) was markedly greater than of infarction secondary to a 40 minute period
in controls (5.7 ± 1.7 ng/ml vs. 1.9 ± 1.1 ng/ml, of occlusion but did not reduce either infarct
p=0.05). The transmyocardial lactate gradient size nor improve regional function when re-
was shifted towards production in the PC group produced 24 hours later.9 Similar results were
(+0.22 ± 0.13 ∝mol/L) and towards extraction obtained recently in our laboratory on an iso-
in the control group (-0.06 ± 0.21 ∝mol/L). lated buffer-perfused rabbit heart preparation.
The lack of additional protection conferred Sixty minutes of normothermic hyperkalemic
by ischemic preconditioning was further con- arrest caused only minimal necrosis (< 10%
firmed by the absence of difference in of the left ventricle) as assessed by tri-
postarrest myocardial levels of ribonucleic acid phenyltetrazolium staining after 60 minutes
messengers coding for the cardioprotective of reperfusion. Ischemic preconditioning with
heat shock proteins (HSP) 70 between the two 5 minutes of zero-flow ischemia followed by
groups. Although, there were no PC-related 5 minutes of reperfusion before cardioplegic
adverse clinical events, this trend towards in- arrest failed to enhance recovery of myocar-
creased initial ischemic insult cannot be dis- dial function or coronary flow compared with
missed, suggesting that a brief prearrest period nonpreconditioned hearts. Conversely, a simi-
of aortic occlusion largely offsets, under the lar preconditioning stimulus improved func-
conditions studied, its putative cardio- tional recovery after 45 minutes of unprotected
protective action. Since then, similar results global ischemia11 but infarct size was not mea-
showing a detrimental effect of ischemic pre- sured. We duplicated this protocol and showed
conditioning before normothermic aerobic that 45 minutes of unprotected global is-
arrest have been shown by Kaukoranta and chemia caused a massive infarction of the left
associates5 with preconditioned patients show- ventricle thereby lending room for precondi-
ing a greater release of creatine kinase MB tioning to reduce the extent of necrosis and
isoenzymes and troponin T indicating a greater hence functional recovery. These data suggest
amount of tissue injury compared with non- that the pertinence of preconditioning in rou-
preconditioned controls. tine cardiac surgery may be limited since
The apparent discrepancy in the results postbypass failure is more often secondary to
obtained with either noncardioplegic and global reversible dysfunction than to discrete
cardioplegic techniques may be explained by necrosis.12
two basic observations. First, preconditioning An important observation is that most
reduces infarct size but has little effect on stun- experimental studies having documented the
ning and beneficial effects are only detectable salutary effects of preconditioning have done
in situations of unprotected ischemia. Indeed, so in models of unprotected ischemia while a
only a few studies have concomitantly assessed number of studies have shown that the ben-
function and infarct size to shed light on this efits of preconditioning are lost when some
observation.6 Such studies have conclusively protective strategies are added such as hypo-
demonstrated that preconditioning improves thermia or cardioplegic arrest.13 One such
postischemic myocardial function by a reduc- study is that of Cleveland et al14 which showed
tion of the size of the infarct and not by that preconditioning improved mechanical
decreasing the amount of stunning of revers- recovery of human right atrial trabeculae
ibly injured myocardium. Moreover, this con- exposed to hypoxia at normothermia but was
clusion applies both to regional ischemia mod- lost under conditions of hypothermia. This can
els such as obtained through transient be explained by the fact that regardless of the
occlusion of a coronary artery as well as to glo- precise mechanisms by which precondition-
bal ischemia induced by a period of aortic ing exerts its protective effect, slowing of the
cross-clamping.7,8 Indirect evidence of this decay of high energy phosphates and glyco-
also comes from Qiu’s study performed in a gen depletion, which leads to slower lactate
conscious porcine model in which accumulation, during the early phase of
preconditioning significantly reduced the size sustained ischemia, appears to be important
128 Ischemia-Reperfusion Injury in Cardiac Surgery

phenomena.15,16 It is therefore plausible that cardioplegic solutions were suboptimal due to


these energy sparing effects and prevention of proximal occlusions of coronary arteries.24 It
intracellular acidosis may become redundant is likely that in these situations, precondition-
to that of cardioplegia. This hypothesis is con- ing reduces the amount of necrosis, as shown
firmed by the observation, made in isolated by lower postischemic creatine kinase release,
rat hearts subjected to 35 minutes of global resulting from suboptimal protection. Thus in
ischemia, that both ischemic preconditioning clinical practice, high risk situations that could
and cardioplegia independently improved benefit from preconditioning may include
functional recovery compared to control hearts 1) extensive coronary disease with poor
but these effects were not additive.17 More- collaterals that increases the risk of cardiople-
over, combination of preconditioning with gia maldistribution even with the use of com-
magnesium based cardioplegia fails to improve bined antegrade/retrograde perfusion, 2) severe
results over that obtained with cardioplegia left ventricular hypertrophy, 3) anticipated
alone.18 For these reasons, it is conceivable that long ischemic times including those of cardiac
preconditioning may enhance the effects of allografts are exposed to during cold storage,
normothermic ventricular fibrillation used by 4) the senescent myocardium, more prone to
Alkhulaifi3 which is expected to cause a major developing calcium overload injury,25 although
decline in myocardial ATP levels and intracel- the capacity of the aged heart for becoming
lular pH. In confirmation of this, such preser- preconditioned remains unclear.26,27 One final
vation of ATP was lost in a study by the same situation in which preconditioning may find
investigators when preconditioning was used a niche is, 5) beating heart minimally invasive
before induction of ventricular fibrillation at coronary artery bypass operations where the
lower temperatures.19 In the same line of rea- necessary control and occlusion of target ves-
soning, the satisfactory outcomes reported in sels sometimes results in distal ischemia, not
patients undergoing coronary artery bypass unlike the experimental models of regional
surgery under intermittent aortic cross- ischemia in which the infarct-limiting effect
clamping and ventricular fibrillation may be of preconditioning has been best demon-
explained by the fact that cumulative myocar- strated. Potential benefits notwithstanding, the
dial injury by successive no-flow intervals clinical use of a potentially deleterious ischemic
could be limited by the initial period of stimulus is unappealing hence the importance
aortic occlusion acting as a preconditioning of identifying the mediators of this endog-
stimulus.20 enous adaptive phenomenon to best exploit it
therapeutically.
Possible Clinical Indications
of Preconditioning in Cardiac Pharmacological Preconditioning
Surgery According to the commonly accepted
From the above data, it results that pre- scheme, the preconditioning ischemia activates
conditioning may be indicated electively in various membrane receptors, including those
certain situations when the potential exists for for adenosine, catecholamines, acetylcholine,
suboptimal myocardial protection and conse- bradykinin and opioids. The respective role
quently and increased risk of myocardial inf- of these receptors may vary among species as
arction and it could be anticipated that the exemplified by the adenosine receptor which
extent of the infarct will be reduced by pre- mediates preconditioning in rabbit, dog, swine
conditioning and serve to preserve function. and human but not rats and the precise level
This scenario has been investigated in experi- of activation of preconditioning triggers
mental studies in which preconditioning con- required to reach the effective threshold for
ferred additional protection to that of hypo- cardioprotection may also vary.28 Stimulated
thermia and cardioplegia when ischemic times receptors then initiate an intracellular signaling
were prolonged21-23 and when distribution of pathway leading to activation of protein kinase
Away from Ischemic Preconditioning and Towards Pharmacological Preconditioning 129

C (PKC) although its predominant role myocardium is more dismal and based on iso-
remains questionable considering other kinases lated in vitro preparations of human atrial tra-
such as mitogen-activated protein kinases beculae or cultured ventricular myocytes.
(MAP kinases) may also be involved.29 Evi- Recently, clinically relevant data has been
dence supports that activation of PKC leads reported by Lee et al34 which showed that in-
to its translocation from the cytosol to the fusion of adenosine prior to cardiopulmonary
membrane where it phosphorylates substrate bypass improved postoperative ventricular
proteins that ultimately increase the resistance function in patients under CABG. However a
to ischemia. The end-effectors of the precon- period of drug-free intervening reperfusion,
ditioning pathway are not complementaly necessary for qualifying as preconditioning
characterized but the most likely candidate is rather than pretreatment was not used, and
the ATP-dependent potassium channel, sup- could have falsely improved the extent of
ported by evidence in rabbits that PKC can recovery.35 A more convincing study published
activate potassium channels at physiological by Leesar and co-workers36 showed that the
levels of ATP.30 It is generally accepted that intracoronary infusion of adenosine 10 min-
opening of the potassium channels shortens utes before percutaneous transluminal coro-
the duration of the action potential and nary angioplasty rendered the myocardium
reduces calcium influx and the attendant tis- significantly more resistant to subsequent
sue damage. This hypothesis has been chal- ischemia induced by successive balloon infla-
lenged recently by data showing that the tions. In the surgical context, however, the use
potassium channel opener bimakalim induces of adenosine can be fraught with several prob-
cardioprotection without modifying the lems including down-regulation of the recep-
duration of the action potential,31 raising the tors with uncoupling of the intracellular
possibility of alternate protective mechanisms signaling pathway as well as systemic vasodi-
either at the intracellular level (in mitochon- latation and subsequent hypotension. It
drial membranes) and/or extracellularly by remains to be determined whether these po-
preventing adhesion of neutrophils.32 Another tential side effects will be eliminated by the
way for preconditioning to reduce acidosis more selective A1 agonists currently under
during ischemia could be through decreasing development.
the sodium/proton exchange18 leading to a Activation of !1-adrenergic receptors37 for
lesser rise in intracellular sodium and second- induction of preconditioning is another inter-
arily a reduced rise in intracellular calcium esting possibility as agonists of such receptors
through preservation of sarcolemmal sodium/ are available for human use such as phenyl-
potassium ATPase.33 Of great interest is that ephrine. The clinical applicability of this
all end-effectors under consideration share the approach remains unproven which holds true
common property of reducing intracellular for stimulation of bradykinin receptors which
accumulation of calcium and ultimately may mediate the antiarrythmic effects of pre-
enhancing tissue viability. This framework is conditioning through a pathway related to
convenient for classifying interventions tar- nitric oxide.38
geted at pharmacological induction of precon-
ditioning in the myocardium. Interventions Targeted
at the Mediators
Interventions Targeted A number of PKC agonists have been stud-
at the Triggers ied and shown to reproduce the cardio-
The most widely used trigger has been protective effect of ischemic preconditioning
adenosine as stimulation of adenosine A1 but they remain solely mechanistic tools
receptors have been successful in duplicating because of their tumor-promoting effects.
the cardioprotection conferred by ischemic Recent evidence seems to suggest that calcium
preconditioning except in rats. Data in human chloride could effectively precondition the
130 Ischemia-Reperfusion Injury in Cardiac Surgery

heart through a PKC-dependent mechanism sure to potassium channel openers before ar-
and could perhaps become a useful drug if the rest is unclear but may include a lowering of
benefits and safety of an increase in inotropism the threshold for potassium channels opening
before going on bypass is established. favoring a greater activation during the subse-
quent period of global ischemia.48 Unfortu-
Interventions Targeted nately, the clinical usefulness of these drugs in
at the End-Effectors clinical cardiac surgery is currently hampered
Since potassium channel openers duplicate by their sole enteral route of administration.
the cardioprotective effects induced by precon- Furthermore, antiischemic effects related to
ditioning, the ATP-dependent potassium potassium channel opening can only be ob-
channel is currently considered one of the tained at doses also causing smooth muscle cell
major effectors of the signaling pathway.40 This relaxation because of their poor cardiac selec-
has been shown to be true in numerous spe- tivity and may cause important hemodynamic
cies as well as in man as demonstrated by pro- changes before the occurrence of cardio-
tection of human right atrial trabeculae against protective effects.49
hypoxic injury by preconditioning induced by
hypoxia or cromakalim41 and by the absence Confounding Factors
of such a protective effect in trabeculae from in Cardiac Surgery
diabetic patients on potassium-blocking During cardiac operations, numerous pre
hypoglycemic drugs.42 Several reports have also and intraoperative confounding factors may
documented the capacity of potassium chan- skew the preconditioning effect and should be
nel openers to enhance functional recovery at controlled for in both experimental and clini-
reperfusion when given before standard depo- cal studies. These include, in particular, the
larized arrest43-45 but again some of these agents use of opioids agonists, aprotinin and cardio-
were given as pretreatment rather than true pulmonary bypass itself which may all have
preconditioning. We have shown in an iso- preconditioning effects.
lated model rat heart model that the protec- In the rat heart, opioid receptor stimula-
tive effects of ischemic preconditioning tion by morphine results in a reduction in
(achieved by 5 minutes of zero-flow ischemia infarct size similar to that produced by
followed by 5 minutes of reperfusion before ischemic preconditioning and is blocked by
arrest) on post cardioplegia systolic and dias- glibenclamide suggesting its mediation by
tolic function after 45-minute normothermic potassium channel opening.50 We have spe-
potassium arrest could be duplicated by cifically identified the (-opioid receptor as a
nicorandil (10 mM/L) given as a true precon- target for pharmacological interventions
ditioning regimen.46 In addition, both forms duplicating the effects of preconditioning in a
of preconditioning similarly lengthened the rabbit model of prolonged cold storage
time before peak contracture and its magni- (unpublished observation).
tude. Glibenclamide, a potassium channel Preconditioning has been shown to
blocker, completely abolished the cardio- decrease the increase in infarct size caused by
protective effects of nicorandil precondition- aprotinin in a sheep model of regional
ing confirming its action through modulation ischemia.51 Should these observations be con-
of potassium channels, whereas it only par- firmed in the clinical setting, preconditioning
tially blunted those of ischemic precondition- could be useful in patients requiring aprotinin
ing suggesting that other end-effectors may be therapy.
involved, at least in the rat heart. Similar re- Finally, the same group of investigators has
sults were obtained with the more commonly also shown that cardiopulmonary bypass itself
used conditions of hypothermic cardioplegic could act as a preconditioning stimulus
arrest.47 The mechanism by which myocardial through the stimulation of release of mem-
cells retain the memory of the transient expo- brane activating mediators like adenosine and
Away from Ischemic Preconditioning and Towards Pharmacological Preconditioning 131

catecholamines.52 This hypothesis is based on 5. Kaukoranta P, Lepojärvi MPK, Ylitalo KV et


the observation that a period of cardiopulmo- al. Normothermic retrograde blood cardiople-
gia with or without preceding ischemic pre-
nary bypass could duplicate the reduction in conditioning. Ann Thorac Surg 1997; 63:
infarct size achieved by short bursts of pre- 1268-74.
conditioning and was abolished by adenosine 6. Bolling SF, Olszanski DA, Childs KF et al.
and !-1 adrenoreceptors antagonists. If this Stunning, preconditioning, and functional
recovery after global myocardial ischemia.
hypothesis is correct, it would imply that, in Ann Thorac Surg 1994; 58:822-7.
any clinical study of preconditioning, “con- 7. Ovize M, Kloner RA, Przyklenk K. Precon-
trol” patients might be already preconditioned ditioning and myocardial contractile function.
which could lessen the likelihood of dem- In: Przyklenk K, Kloner RA, Yellon DM. eds.
onstrating an added benefit from any Ischemic preconditioning: The concept of
endogenous cardioprotection. Boston: Kluwer
superimposed intraoperative precondition- Academic Publishers, 1994: 41-60.
ing intervention. 8. Iles RW, Wright JK, Inners-McBride K et al.
In conclusion, preconditioning is an Ischemic preconditioning improves preserva-
extremely effective mechanism for limiting tion with crystalloid cardioplegia. Ann Thorac
Surg 1994; 58:1481-5.
ischemia-induced cell necrosis and, conse- 9. Qiu Y, Tang XL, Park SW et al. The early
quently, preserving myocardial function. As no and late phases of ischemic preconditioning.
current methods of cardioplegia is perfect, A comparative analysis of their effects on in-
additional protection could be obtained in farct size, myocardial stunning, and arrhyth-
selected cases through the use of precondition- mias in conscious pigs undergoing a 40-
minute coronary occlusion. Circ Res 1997;
ing. This requires, however, the characteriza- 80:730-42.
tion of the endogenous mediators of this phe- 10. Faris B, Peynet J, Wassef M et al. Failure of
nomenon to develop new agents or exploit preconditioning to improve postcardioplegia
current drugs to duplicate the cardioprotective stunning of minimally infarcted hearts. Ann
Thorac Surg (accepted for publication).
effects of preconditioning. The clinical evalu- 11. Hendricx M, Toshima Y, Mubagwa K et al.
ation of these agents may be difficult consid- Improved functional recovery after ischemic
ering the numerous confounding factors. Nev- preconditioning in the globally ischemic rab-
ertheless, this should not deter us from bit heart is not mediated by adenosine A1
applying continued research efforts for under- receptor activation. Bas Res Cardiol 1993;
88:576-93.
standing and exploiting therapeutically this 12. Kloner RA, Przyklenk K, Kay GL. Clinical
endogenous adaptive defense system which has evidence for stunned myocardium after coro-
proven so far to be one of the most effective nary artery bypass surgery. J Card Surg 1994;
infarct-limiting strategies available. 9[Suppl]:397-402.
13. Valen G, Takeshima S, Vaage J. Precondi-
tioning improves cardiac function after glo-
References bal ischemia, but not after cold cardioplegia.
1. Parratt JR. Protection of the heart by ischae- Ann Thorac Surg 1996; 62:1397-403.
mic preconditioning: Mechanisms and possi- 14. Cleveland JC Jr., Meldrum DR, Rowland RT
bilities for pharmacological exploitation. TiPS et al. Preconditioning and hypothermic car-
1994; 15:19-25. dioplegia protect human heart equally against
2. Marber MS, Heads RJ, Yellon DM. Stress ischemia. Ann Thorac Surg 1997; 63:147-52.
proteins, heat stress and myocardial protec- 15. Kida M, Fujiwara H, Ishida M et al. Ischemic
tion. In: Przyklenk K, Kloner RA, Yellon DM preconditioning preserves creatine phosphate
eds. Ischemic preconditioning: The concept and intracellular pH. Circulation 1991;
of endogenous cardioprotection. Boston: 84:2495-503.
Kluwer Academic Publishers 1994:105-23. 16. Murry CE, Richard VJ, Reimer KA et al. Is-
3. Alkhulaifi AM, Yellon DM, Pugsley WB. chemic preconditioning slows energy metabo-
Preconditioning the human heart during lism and delays ultrastructural damage dur-
aorto-coronary bypass surgery. Eur J Cardio- ing a sustained ischemic episode. Circ Res
Thorac Surg 1994; 8:270-276. 1990; 66:913-31.
4. Perrault LP, Menasché P, Bel A et al.Ischemic 17. Kolocassides KG, Galinanes M, Hearse DJ.
preconditioning in cardiac surgery: A word Ischemic preconditioning, cardioplegia or
of caution. J Thorac Cardiovasc Surg 1996; both? J Mol Cell Cardiol 1994; 26:1411-4.
112:1378-86.
132 Ischemia-Reperfusion Injury in Cardiac Surgery

18. Steenbergen C, Perlman ME, London RE et 31. Yao Z, Gross GJ. Effect of the KATP channel
al. Mechanism of preconditioning; ionic al- opener bimakalim on coronary blood flow,
terations. Circ Res 1993; 72:112-25. monophasic action potential duration, and
19. Di Salvo C, Hemming A, Jenkins D et al. infarct size in dogs. Circulation 1994; 89:
Can the human myocardium be precondi- 1769-75.
tioned with ischaemia under hypothermic 32. Akimitsu T, Gute DC, Korthuis RJ. Ischemic
conditions? Proceedings of the 9th annual preconditioning attenuates postischemic leu-
meeting of the European Association for kocyte adhesion and emigration: Role of ad-
Cardio-thoracic Surgery, Paris, 25-27 Septem- enosine and ATP-sensitive potassium chan-
ber 1995; 324 (abstract). nels. Circulation 1994; 90:(suppl I):I-476 (ab-
20. Abd-Elfattah AS, Ding M, Wechsler AS. In- stract).
termittent aortic cross-clamping prevents cu- 33. Nawada R, Murakami T, Iwase T et al. In-
mulative adenosine triphosphate depletion, hibition of sarcolemmal Na+, K+-ATPase ac-
ventricular fibrillation, and dysfunction tivity reduces the infarct size-limiting effect
(stunning): Is it preconditioning? J Thorac of preconditioning in rabbit hearts. Circula-
Cardiovasc Surg 1995; 110:328-39. tion 1997; 96:599-604.
21. Cave AC, Hearse DJ. Ischaemic precondition- 34. Lee HT, LaFaro RJ, Reed GE. Precondition-
ing and contractile function: Studies with ing of human myocardium with adenosine
normothermic and hypothermic global is- during open-heart surgery. J Card Surg 1995;
chaemia. J Mol Cell Cardiol 1992; 24: 10:665-76.
1113-23. 35. Lasley RD, Noble MA, Konyn PJ et al. Dif-
22. Ogino H, Smolenski RT, Zych M et al. In- ferent effects of an adenosine A1 analogue and
fluence of preconditioning on rat heart sub- ischemic preconditioning in isolated rabbit
jected to prolonged cardioplegic arrest. Ann hearts. Ann Thorac Surg 1995; 60:1698-703.
Thorac Surg 1996; 62:469-74. 36. Leesar MA, Stoddard M, Ahmed M et al.
23. Karck M, Rahmanian P, Haverich A. Is- Preconditioning of human myocardium with
chemic preconditioning enhances donor heart adenosine during coronary angioplasty . Cir-
preservation. Transplantation 1996; 62:17-22. culation 1997; 95:2500-7.
24. Galinanes M, Argano V, Hearse DJ. Can is- 37. Banerjee A, Winter-Locke C, Rogers KB et
chemic preconditioning ensure optimal myo- al. Preconditioning against myocardial dys-
cardial protection when delivery of cardiople- function after ischemia and reperfusion by an
gia is impaired? Circulation 1995; 92 (suppl a 1-adrenergic mechanism. Circ Res 1993;
II):II-389-94. 73:656-70.
25. Tsukube T, McCully JD, Federman M et al. 38. Vegh A, Szekeres L, Parratt JR. Protective
Developmental differences in cytosolic cal- effects of preconditioning on the ischaemic
cium accumulation associated with surgically myocardium involve cyclo-oxygenase prod-
induced global ischemia: Optimization of ucts. Cardiovasc Res 1990; 24:1020-3.
cardioplegic protection and mechanism of 39. Meldrum DR, Cleveland JC Jr., Sheridan BC
action. J Thorac Cardiovasc Surg 1996; et al. Cardiac preconditioning with calcium:
112:175-84. Clinically accessible myocardial protection. J
26. Burns PG, Krukenkamp I, Caldarone CA et Thorac Cardiovasc Surg 1996; 112:778-86.
al. Is the preconditioning response conserved 40. Gross GJ, Auchampach JA. Blockade of ATP-
in senescent myocardium? Ann Thorac Surg sensitive potassium channels prevents myocar-
1996; 61:925-9. dial preconditioning in dogs. Circ Res 1992;
27. Tani M, Suganuma Y, Hasegawa H et al. 70:223-33.
Changes in ischemic tolerance and effects of 41. Speechly-Dick ME, Grover GJ, Yellon DM.
ischemic preconditioning in middle-aged rat Does ischemic preconditioning in the human
hearts. Circulation 1997; 95:2559-66. involve protein kinase C and the ATP-depen-
28. Goto M, Liu Y, Yang XM et al. Role of dent K+ channel? Studies of contractile func-
bradykinin in protection of ischemic precon- tion after simulated ischemia in an atrial in
ditioning in rabbit hearts. Circ Res 1995; vitro model. Circ Res 1995; 77:1030-5.
77:611-21. 42. Cleveland JC, Meldrum DR, Cain BS et al.
29. Brooks G, Hearse DJ. Role of protein kinase Oral sulfonylurea hypoglycemic agents pre-
C in ischemic preconditioning: Player or spec- vent ischemic preconditioning in human
tator? Circ Res 1997; 79:627-30. myocardium. Two paradoxes revisited. Cir-
30. Light P, Sabir AA, Allen BG et al. Protein culation 1997; 96:29-32.
kinase C-induced changes in the stochiometry 43. Grover GJ, Sleph PG. Protective effect of
of ATP binding activate cardiac ATP-sensi- KATP openers in ischemic rat heart treated
tive K+ channels. Circ Res 1996; 79:399-406. with a potassium cardioplegic solution. J
Cardiovasc Pharmacol 1995; 26:698-706.
Away from Ischemic Preconditioning and Towards Pharmacological Preconditioning 133

44. Pignac J, Bourgouin J, Dumont L. Cold car- 48. Shigematsu S, Sato T, Abe T et al. Pharma-
dioplegia and the K + channel modulator cological evidence for the persistent activation
aprikalim (RP 52891): Improved cardio- of ATP-sensitive K+ channels in early phase
protection in isolated ischemic rabbit hearts. of reperfusion and its protective role against
Can J Physiol 1994; 72:126-32. myocardial stunning. Circulation 1995;
45. Sugimoto S, Puddu PE, Monti F et al. Pre- 92:2266-75.
treatment with the adenosine triphosphate- 49. Grover GJ. Protective effects of ATP sensi-
sensitive potassium channel opener nicorandil tive potassium channel openers in models of
and improved myocardial protection during myocardial ischaemia. Cardiovasc Res 1994;
high-potassium cardioplegic hypoxia. J Thorac 28:778-82.
Cardiovasc 1994; 108:455-66. 50. Schultz JE, Hsu AK, Gross GJ. Morphine
46. Menasché P, Kevelaïtis E, Mouas C et al. mimics the cardioprotective effect of ischemic
Preconditioning with potassium channel preconditioning via a glibenclamide-sensitive
openers: A new concept for enhancing cardio- mechanism in the rat heart. Circ Res 1996;
plegic protection? J Thorac Cardiovasc Surg 78:1100-4.
1995; 110:1606-14. 51. Bukhari EA, Krukenkamp I, Burns PG et al.
47. Menasché P, Mouas C, Grousset C. Is po- Does aprotinin increase the myocardial dam-
tassium channel opening an effective form of age in the setting of ischemia and precondi-
preconditioning before cardioplegia? Ann tioning ? Ann Thorac Surg 1995; 60:307-10.
Thorac Surg 1996; 61:1764-8. 52. Burns PG, Krukenkamp IB, Caldarone CA
et al. Does cardiopulmonary bypass alone
elicit myoprotective preconditioning? Circu-
lation 1995; 92(supplII):II-447-51.
Section III:
Intravenous Metabolic Support
CHAPTER 12

Intravenous Metabolic Support


with GIK (Glucose-Insulin-Potassium)
and Amino Acids in Cardiac Surgery
Rolf Svedjeholm

M
yocardial preservation in cardiac failure.4 In light of this, we believe that addi-
surgery has evolved rapidly during tional measures to improve outcome and
the last decades. Some authors of reduce permanent myocardial damage in car-
this book have made major contributions to diac surgery should focus on the preoperative
this development. A variety of methods to pro- and the postoperative phase of CABG surgery.
tect the heart are available and complex pro- Furthermore, efforts should be instituted to
cedures can be safely performed. Inappropri- reduce reperfusion injury and minimize per-
ate protection of the heart during aortic manent myocardial damage in long standing
cross-clamping is now an uncommon cause or severe myocardial ischemia.
of major myocardial damage. A recent survey The basic principles to address myocardial
at our institution showed that PMI (periop- ischemia imply improving the ratio between
erative myocardial infarction) in association oxygen delivery and myocardial oxygen
with valvular surgery was rare.1 When PMI demand. If, however, ischemia is severe enough
occurred in these cases, the myocardial dam- to cause cardiac failure, or if it is followed by
age was usually caused by evident technical cardiac failure after revascularization due to
problems. Thus, PMI in our hands was a sig- stunning or myocardial damage, the treatment
nificant problem in association with surgery traditionally employed (inotropic drugs) is less
for ischemic heart disease (despite shorter than ideal for the ischemic or postischemic
cross-clamp times). Furthermore, in this set- heart. Traditional treatment of cardiac failure
ting PMI was mainly associated with unstable with inotropic drugs improves the hemody-
angina (preoperative ischemia) or poor con- namic state, but at the price of a marked
ditions for revascularization. The role of pre- increase in myocardial expenditure.5-8 In ani-
operative ischemia as a risk factor for PMI has mal experiments adrenergic drugs aggravate
been documented by Slogoff and Keats in a ischemia and precipitate myocardial infarc-
classic paper. 2 Moreover, postoperative tion.9 Inotropes have also been shown to cause
ischemia after coronary artery bypass surgery de-energization of the myocardium, and if
is common and may also play a role for the instituted prematurely it may imply that the
development of PMI.3 Postischemic myocar- condition of the heart is aggravated by further
dial metabolic derangement that is further metabolic derangement.8 The detrimental
aggravated by the systemic neuroendocrine effects of premature use of inotropic drugs for
stress response to operative trauma may, weaning from CPB have been documented in
together with reperfusion injury, cause cardiac animals.10 Therefore, alternative measures that

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


136 Ischemia-Reperfusion Injury in Cardiac Surgery

can enhance myocardial recovery and function ischemia and to facilitate postischemic recov-
without putting further strain on the heart are ery. The role of these substrates is illustrated
desirable. It is with this perspective intrave- by the importance of the myocardial
nous metabolic support will be addressed in glutamate/aspartate pool and myocardial
the following presentation. glycogen content for myocardial recovery after
A brief review of myocardial metabolism ischemia.20,23 Furthermore, administration of
in association with ischemia and cardiac sur- glucose or glutamate enhances myocardial tol-
gery will be presented. After that, the current erance to ischemia, improves the metabolic
state of metabolic support with GIK (glucose- and functional recovery of the postischemic
insulin-potassium) and amino acids will be heart and reduces the size of myocardial inf-
surveyed. Finally, a preliminary report on our arct in experimental coronary ligation.4,6,24,28
clinical experience with metabolic support will In contrast to the beneficial effects of carbo-
be given. hydrates and amino acids, FFA metabolism
may be harmful in association with myocar-
Myocardial Substrate dial ischemia. High plasma levels of FFA
Metabolism impede myocardial uptake of glucose.12 Under
normal conditions FFA metabolism requires
The myocardial ATP content is limited
12% more oxygen per mole ATP produced,
and, therefore, the myocytes must continually
compared with glucose utilization.12 In con-
resynthesize high-energy phosphates to main-
ditions associated with high levels of circulat-
tain contractile function and cellular viabil-
ing catecholamines, such as trauma or
ity.11 To cope with this the heart can use a
treatment with adrenergic drugs, the oxygen-
variety of substrates for energy production.12
wasting effect of FFA metabolism is markedly
The choice in a given situation is normally
aggravated.29 Accumulation of intracellular
determined by the availability of substrates.
FFA metabolites may exert toxic effects and
In the fasting state arterial levels of FFA (free
high levels of circulating FFA depress myo-
fatty acids) are elevated and FFA oxidation
cardial function and increase the incidence of
then dominates. After a carbohydrate meal a
arrhythmias during ischemia.30,33
shift toward glucose oxidation occurs, and
Oxidative metabolism which normally
during vigorous exercise lactate may become
accounts for more than 90% of myocardial
the dominating fuel. In conditions of starva-
energy production11 is impeded after severe
tion or diabetic ketosis ketones may account
ischemia in animal experiments.34,36 Loss of
for a significant amount of myocardial energy
Krebs cycle intermediates during ischemia has
production. Cardiac function is not affected
been proposed as an explanation to this phe-
by the specific substrate utilization under nor-
nomenon.36,38 Another mechanism that may
mal circumstances.12 However, in conditions
contribute is inactivation of pyruvate dehy-
where myocardial perfusion is, or has been
drogenase enzyme during ischemia.6,39 This
inadequate, the preference of substrates may
postischemic mitochondrial dysfunction seems
obtain functional significance.
to occur also in humans after cardiac opera-
Myocardial ischemia enhances the use of
tions. Teoh et al demonstrated zero oxidation
glucose (glycolysis) and the use of amino acid
of FFA during early reperfusion utilizing crys-
metabolites in the Krebs cycle.12,17 Ischemic
talloid cardioplegia, and minimal oxidation
heart disease in humans is characteristically
after blood cardioplegic arrest.40 Svensson et
associated with an increased myocardial uptake
al could not demonstrate significant myocar-
of glutamate.13,14,18 Glutamate facilitates gly-
dial uptake of FFA or carbohydrates in the first
colysis and provides an alternative anaerobic
hours after coronary surgery.41 After cardiac
pathway for regeneration of high-energy phos-
operations with limited ischemic insult these
phates by substrate level phosphorylation in
metabolic abnormalities are minor.42 However,
the Krebs cycle.17,19 These changes serve as
when postischemic metabolic derangement
adaptive measures to endure myocardial
occurs the conditions for recovery are unfa-
Intravenous Metabolic Support in Cardiac Surgery 137

vorable because of the systemic neuroendo- our knowledge by permitting the purification
crine response elicited by cardiac surgery.43 of experimental conditions. They allow sophis-
Krebs cycle intermediates are replenished pri- ticated analyses that are not possible to per-
marily by amino acids and carbohydrate sub- form in humans. However, these studies have
strates.37,44 As the systemic neuroendocrine limitations, not only due to species differences,
stress response is associated with elevated FFA but also because some of them are conducted
levels and a state of insulin resistance, the under unphysiological conditions and some-
myocardial uptake of carbohydrate substrates times with irrelevant dosages. In vitro studies
is impeded.45 Thus, the relative importance cannot take systemic interactions or neuro-
of amino acids for the recovery of myocardial endocrine responses into account. Further-
oxidative metabolism is enhanced in this set- more, important interactions between differ-
ting.46 Accordingly, myocardial uptake of ent substrates and metabolites may be
glutamate has been shown to precede the overlooked. However, the number of studies
recovery of oxidative metabolism after concerning myocardial metabolism and meta-
CABG.43 A relative shortage of glutamate in bolic intervention in humans are still few. Lack
this setting is suggested by the high fractional of commercial interest and the tradition
extraction rate of glutamate from plasma regarding therapeutic policies have not helped
(Fig. 12.1).43,46 to promote metabolic studies nor the intro-
duction of metabolic treatment. With guid-
Metabolic Intervention ance of the knowledge gained from animal
The ideal condition for metabolic inter- studies we have to identify metabolic states in
vention is a state of functional disorder sec- humans that may be accessible to metabolic
ondary to a correctable metabolic abnormal- interventions. From such studies metabolic
ity. This may be exemplified by coma interventions that are appropriate regarding
secondary to severe hypoglycemia. In this con- indication, choice of substrate and dosage can
dition the administration of a small amount be evolved. So far, the limited data available
of glucose will have dramatic effects. In con- show that metabolic trials in humans that have
ditions where there is no lack of glucose, the tried to define the rationale for the treatment,
addition of glucose will not affect brain func- and proved that the metabolic objectives were
tion. If we were to treat all comatose patients achieved, have been successful also regarding
with glucose, the results would soon become variables of clinical interest.
discouraging. This may serve as a simple and
illustrative example of the importance of GIK (Glucose-Insulin-
delineating what we are treating. Since the Potassium)
heart can use a wide range of substrates, it Basic research suggests that glucose is ben-
implies that under many conditions the addi- eficial to the ischemic myocardium and may
tion of extra substrates may have little influ- influence the preservation of mechanical func-
ence on the metabolic and hemodynamic func- tion, structure, histology and ionic balance.48
tion. Therefore, it would be surprising to find The glucose for glycolysis originates from both
one substrate that in every experimental con- breakdown of myocardial glycogen stores and
dition improved the metabolic and hemody- the uptake of glucose from blood.49 Myocar-
namic function of the heart.47 Our knowledge dial glucose uptake is stimulated during
about the precise indications and dosages of ischemia by translocation of glucose transport-
substrates still is limited. It is only in the last ers from the intracellular pool to the sarcolem-
decade that we have began to understand the mal membrane. 49,51 However, in severe
transport mechanisms of glucose and amino ischemia glucose uptake may decrease due to
acids across the cell membrane of the inadequate glucose delivery or accumulation
cardiomyocytes. In vivo and in vitro experi- of protons and lactate.12,49
ments on animal hearts have contributed to
138 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 12.1. Relationship between arterial plasma concentration of glutamate and the arterial-coronary sinus plasma
concentration difference of glutamate in 18 patients 4 to5 hours after coronary surgery. Each square denotes one
patient. Reprinted with permission from: Svedjeholm et al. J Thorac Cardiovasc Surg 1991; 101:688-94. © Mosby-
Year Book Inc.

Myocardial uptake of glucose is not depen- However, the reverse—a state of insulin resis-
dent on insulin. However, insulin stimulates tance—is observed after trauma and other
glucose uptake by translocation of glucose conditions that activate the neuro-endocrine
transporters to the sarcolemmal membrane system.45,55 Therefore, the need for exogenous
and possibly by improving their capacity.52 insulin to achieve metabolic effects with GIK
Plasma FFA level is reduced and myocardial treatment may be expected to be most pro-
glycogen synthesis is promoted.6,23 The effects nounced in states of relative insulin deficiency,
of ischemia and insulin on glucose uptake seem such as diabetes mellitus, surgical stress and
to be additive.50 Furthermore, insulin activates other states of insulin resistance.
hexokinase and causes transfer of hexokinase As substrate levels normally play a role for
to the mitochondrial membrane, thus increas- the substrate preference by the heart, blood
ing phosphorylation of free glucose in the glucose level obviously has to be considered.56
cytoplasm.53 It has also been proposed that After cardiac surgery, when systemic glucose
insulin may stimulate the pyruvate dehydro- uptake is severely restricted in spite of using
genase enzyme and thereby enhance Krebs high insulin doses, systemic glucose uptake can
cycle metabolism after ischemia.6,39 be further enhanced by increasing blood
The need for insulin in non-diabetics is glucose.57
debatable. Endogenous insulin production Interactions between substrates have to be
responds to changes in nutritional state and considered. High FFA levels impede myocar-
activity of other hormones. Furthermore, the dial glucose uptake and this is at least partly
myocardial sensitivity to insulin may increase caused by inward translocation of glucose
as an adaptive measure, as demonstrated in transporters.50 Myocardial uptake of glutamate
patients with severe valvular aortic stenosis.54 correlates with myocardial uptake of glucose
Intravenous Metabolic Support in Cardiac Surgery 139

and other carbohydrate substrates in coronary tion of small doses of insulin and glucose from
patients.14 This may, however, merely reflect the induction of anesthesia to the start of CPB
the substrate preference of the ischemic heart. in elective CABG failed to influence the post-
Exposure of the isolated heart to pyruvate and operative hemodynamic state or the release of
lactate leads to a reduction in glucose uptake.58 cardiac enzymes compared with controls (16
In the preoperative setting most of the patients in each group of whom 75% received
interest concerning GIK has focused on myo- counteracting treatment with dopamine).63
cardial glycogen content. Experimental data Inasmuch as preoperative fasting per se pro-
suggest that elevation of myocardial glycogen motes maintenance of myocardial glycogen,
content improves tolerance to ischemia.12,59 stable coronary heart disease may not be the
Preoperative GIK given during preoperative ideal condition for studies on prophylactic
fasting has been shown to enhance myocar- GIK treatment. 60 In patients undergoing
dial glycogen level. This was associated with urgent CABG due to unstable angina an
improved myocardial preservation and post- improved hemodynamic recovery and clinical
operative course.21-23 However, the value of outcome have been demonstrated.66 In the
preoperative GIK is attenuated by the fact that postoperative setting positive hemodynamic
fasting per se promotes maintenance of myo- effects have been obtained with supra-physi-
cardial glycogen. This is due to the inhibition ological doses of insulin.67,68 The hemody-
of glycolysis caused by the elevated plasma FFA namic effects of very large doses of insulin
in the fasting state.60 The impact of preopera- though, are caused by insulin per se. Insulin
tive GIK may be expected to be most pro- has both vasodilative and inotropic properties
nounced in energy depleted hearts. according to animal experiments. 69,70 In
Whereas small doses of insulin and glucose humans the vasodilative effect has been dem-
seem to have been adequate during preopera- onstrated, while the inotropic effect has been
tive fasting,21-23,60 little is known about the more difficult to evaluate.71,72
requirements of insulin and glucose in the early Gradinac et al demonstrated favorable
stages of surgery.61-63 Recent data (unpub- effects of postoperative GIK treatment in
lished) suggest that insulin resistance and patients requiring IABP (intra-aortic balloon
impeded glucose uptake is an issue of concern pump).73 The early hemodynamic recovery
primarily during the early postoperative was improved, the time on IABP was short-
period. In the anesthetized patient we found ened and the requirement for inotropic drugs
substantial myocardial uptake of glucose dur- reduced. These results were achieved in spite
ing early reperfusion. In the early postopera- of moderate doses of insulin (0.08 IU/kg/hour
tive setting the rationale for GIK treatment is for up to 48 hours) not known to exert any
more evident. A pronounced insulin resistance hemodynamic effects. Therefore, it is conceiv-
and a markedly impeded uptake of glucose able that the results obtained were due to meta-
have been demonstrated. Whereas moderate bolic effects. As this study lacked a metabolic
doses of insulin may suffice to reduce plasma correlate, we do not know to what extent the
FFA, insulin doses up to 1 IU/kg/hour may metabolic objectives were achieved. It is pos-
be required to achieve full metabolic effects sible that larger doses of insulin would have
early after cardiac surgery.41,45,55,64 Several stud- amplified the impact of GIK treatment.
ies on GIK have failed to take the insulin The metabolic effects of high-dose GIK on
resistance caused by the postoperative neu- the heart, have been demonstrated in patients
roendocrine stress response into account. It after CABG surgery. Svensson et al observed a
appears that studies on prebypass GIK have significantly improved myocardial uptake of
been successful when a sustained postopera- carbohydrate substrates one hour after sur-
tive metabolic effect could have been expected, gery.41,45 Furthermore, even in patients treated
either due to high doses of insulin given with dopamine the addition of high-dose
prebypass or by treatment extended into the insulin induced a shift of energy substrate pref-
postoperative course.61,65,66 The administra- erence toward carbohydrate oxidation at the
140 Ischemia-Reperfusion Injury in Cardiac Surgery

expense of FFA. The hemodynamic efficacy of glucose metabolites, such as lactate and
of dopamine was improved by high-dose GIK pyruvate, on postischemic recovery have also
without further increasing myocardial oxygen been reported.8,76,77 In an extensive review
demand.6 Stanley recently concluded that metabolic in-
It is important to consider that the rate of terventions aimed at enhancing glucose utili-
glucose uptake, glycogen synthesis and break- zation and pyruvate oxidation at the expense
down, and the rate of glycolysis and glucose of FFA oxidation is a valid therapeutic
oxidation are controlled at multiple sites. In approach to the treatment of myocardial
postischemic hearts, high rates of glycolysis and ischemia.53 The introduction of pharmaco-
low rates of glucose oxidation (due to logic agents which enhance full glucose oxi-
inhibition of pyruvate dehydrogenase by FFA dation at the expense of FFA may create the
metabolism) may lead to an uncoupling of commercial interest necessary to achieve a
glycolysis from glucose oxidation. The conse- widespread use of metabolic therapies.53
quent production of protons can contribute
to ischemic injury and myocardial dysfunc- Amino Acids
tion.49 Adrenergic inotropic drugs may by their Amino acids are not quantitatively impor-
metabolic actions be expected to aggravate this tant substrates for myocardial energy produc-
metabolic condition. Accordingly, elevation of tion. However, increasing evidence suggest that
plasma FFA and a myocardial release of pyru- amino acids are qualitatively important for the
vate correlating with myocardial oxygen intermediary metabolism of the cardio-
demand was found during dopamine stimu- myocytes.15,19,37,44 Their importance is fur-
lation in nondiabetic patients after CABG. ther enhanced during ischemia and after
The addition of high-dose GIK to dopamine ischemia.15-17,78-80 Amino acids associated with
reverted this relationship to an uptake of pyru- the malate-aspartate shuttle (glutamate, aspar-
vate which correlated positively with myocar- tate, arginine, ornithine) are of particular
dial oxygen demand.6 Therefore, it appears importance in this respect.17,19 The effects of
that pyruvate oxidation was stimulated. these amino acids are partly related to their
Whether this beneficial effect was caused by role in the malate-aspartate shuttle, transport-
the reduction of plasma FFA or by direct ing reducing equivalents across the mitochon-
intracellular action of insulin on the pyruvate drial membrane, regulating the NAD/NADH
dehydrogenase enzyme, or both, remains to balance in the cytosol of the cells, and thereby
be clarified.6,39 Also in the diabetic heart a enhancing anaerobic glycolysis during
defect in pyruvate oxidation rather than a ischemia. Furthermore, glutamate and aspar-
defect in glucose uptake has been demon- tate contribute to an alternative anaerobic
strated during dobutamine stimulation.74 In pathway for regeneration of high-energy phos-
acute myocardial infarction it has been dem- phates, by substrate level phosphorylation in
onstrated that insulin-glucose infusion fol- the Krebs cycle. Glutamate also improves the
lowed by a multidose insulin regime improves clearance of lactate and NH3 excess, by taking
one-year survival in diabetics patients.75 It is part in the reactions involving transamination
plausible that GIK treatment will prove to be of pyruvate to alanine and of glutamate to
of particular benefit in diabetic patients un- glutamine. During reperfusion glutamate and
dergoing cardiac surgery. aspartate serve as substrates for the replenish-
The introduction of GIK in clinical prac- ment of Krebs cycle intermediates lost during
tice has been offset by a reputation of con- ischemia.15-17,20,36,78-81
flicting data in the medical literature. How- BCAA (branched chain amino acids—leu-
ever, the number of inconclusive trials in the cine, isoleucine, valine) are with glutamate the
literature are surprisingly few compared with amino acids that are preferentially taken up
the number of papers demonstrating beneficial by the heart after CABG surgery.46,82 Based
effects of GIK for the ischemic and postis- on a study using mixed amino acid solutions
chemic heart. In recent years beneficial effects
Intravenous Metabolic Support in Cardiac Surgery 141

enriched with BCAA it was suggested that undergoing CABG surgery when added to
BCAA improve the metabolic and functional blood cardioplegia.97 Based on the knowledge
recovery of the heart after ischemia.83 How- about amino acid transporters that has
ever, the relative impact of BCAA compared emerged there is reason to question whether
with other amino acids contained in the solu- cardioplegic solutions are ideal for delivery of
tion may be difficult to evaluate. Other inves- amino acids to the heart. Glutamate and
tigators suggest that increased intracellular aspartate are transported into muscle cells by
concentrations of BCAA stimulate formation Na+-dependent and Na+-independent sys-
of acetyl-coenzyme and succinyl-CoA and, tems.98 Suleiman has suggested that delivery
thus, recovery of oxidative metabolism.84 On of glutamate and aspartate by cardioplegia
the other hand, it is uncertain whether exog- could be self defeating due to the anticipated
enous administration alone would influence effect on the amino acid transporters.99 Myo-
myocardial BCAA uptake after CABG, as no cardial loss of glutamate has now been dem-
relationship between plasma levels of BCAA onstrated both after crystalloid cardioplegic
and myocardial uptake has been found.46,82 arrest and during cold blood cardiople-
Thus, the role of BCAA in this setting remains gia.97,99,100 This may explain some conflicting
to be clarified. Other amino acids of interest data regarding amino acid enrichment of
for myocardial protection and metabolism in cardioplegic solutions.101,102 However, it can-
association with ischemia include taurine, not yet be excluded that loss of glutamate is
methionine, cysteine and histidine. A compre- attenuated by glutamate enrichment of the
hensive review on this subject has been given cardioplegic solution. Furthermore, plasma
by Pisarenko.84 levels of glutamate and aspartate are markedly
Of the amino acids associated with the increased during reperfusion and these amino
malate-aspartate shuttle, glutamate and aspar- acids will be available to the postischemic
tate have by far gained the greatest attention heart.103
as regards the heart and metabolic interven- Intravenous infusions offer an alternative
tion. Arginine has received attention recently to amino acid enhancement of cardioplegic
mainly due to its metabolism to nitric solutions. As glutamate is reported to improve
oxide.85,86 myocardial tolerance to ischemia in coronary
In cardiac surgical practice most of the patients, intravenous glutamate administration
interest concerning amino acids has focused offers the prospect of improving myocardial
on their use as additives in cardioplegic and protection during the pre- and postoperative
reperfusion solutions. The glutamate and periods of CABG surgery.25,104 Furthermore,
aspartate enrichment of these solutions relies glutamate and aspartate may eventually play a
on basic metabolic research and studies on role in the treatment of myocardial infarcts as
myocardial preservation in animal models. the infusion of glutamate and aspartate reduces
These studies show that glutamate and aspar- the size of myocardial infarcts in experimental
tate improve myocardial tolerance to ischemia, coronary artery occlusion.27,105 Moreover,
and enhance metabolic and functional recov- infusion of these amino acids together with
ery of the heart after ischemia.15,17,36,87-92 The GIK had salutary effects on the contractility
relative clinical impact of substrates in of viable remote myocardium in experimental
cardioplegic/reperfusion solutions remains to coronary occlusion.106 This is a significant
be clarified in humans. Improved postopera- finding, since failure of remote myocardium
tive hemodynamic recovery when applied to to sustain the compensatory contractility
high-risk coronary patients and an improved required to maintain cardiac output may play
myocardial ATP maintenance in congenital an important role in the development of car-
surgical patients have been suggested.93-96 It diogenic shock.
has also been demonstrated that alpha- Due to the importance of glutamate for
ketoglutarate, a metabolite of glutamate, the recovery of oxidative metabolism after
improves myocardial preservation in humans ischemia36,80 and the shortage of glutamate
142 Ischemia-Reperfusion Injury in Cardiac Surgery

demonstrated early after coronary surgery, a been shown to cause cerebral damage.112 The
rationale for metabolic intervention is pro- possible influence of extracorporeal circulation
vided in this setting. 46,82 Postoperative on the blood brain barrier has to be consid-
glutamate infusion has been shown to improve ered in cardiac surgery. However, extensive
the metabolic and functional state of the heart experience with amino acid enhanced blood
after routine CABG and in postoperative car- cardioplegia is not reported to be associated
diac failure (Figs. 12.2 and 12.3). 26,107 with an increased incidence of cerebral com-
Glutamate has no inotropic effects per se. plications.113 On the other hand, glutamate
Therefore, it is conceivable that the hemody- causes a transient but unpleasant side effect
namic effects during postoperative treatment known as the “Chinese restaurant” syndrome.
were due to the improved metabolic state of It is considered to be caused by stimulation of
the heart. In keeping with this, the hemody- the peripheral nervous system, and may be re-
namic impact of postoperative glutamate lated both to individual susceptibility and dos-
infusion seems to be related to the magnitude age of glutamate.114,115
of metabolic derangement before treatment. A high turnover of glutamate in the human
In animal experiments glutamate and body is suggested by the rapid decrease of
aspartate have additive effects on myocardial arterial levels after discontinuation of
recovery after ischemia.108 Based on data from glutamate infusion.26 If arterial levels play an
the isolated rat heart it has been claimed that important role for adverse reactions, it implies
aspartate may be more important than that the time during which glutamate is
glutamate for the recovery of myocardial me- administered may be more important than the
tabolism after cardioplegic arrest.109 However, amount given. Accordingly, Thomassen
in humans early after CABG we were unable observed short-lasting symptoms compatible
to detect any beneficial effect of aspartate on with the “Chinese restaurant syndrome” in a
myocardial metabolism or function (unpub- patient with coronary artery disease receiving
lished). Intravenous aspartate infusion caused an intravenous injection of only 2.5 mg/kg
an increase of myocardial aspartate uptake but BW. The arterial plasma levels reported were
considerable interactions with glutamate com- only moderately increased, but the peak levels
patible with competitive inhibition of may not have been caught due to rapid clear-
glutamate uptake were observed. These obser- ance of glutamate.25 Adverse effects were not
vations agree with findings in the rat heart seen in patients receiving 1.5 mg/kg BW
where aspartate and glutamate compete for the intravenously or 40 mg/kg BW orally.104
same Na+ dependent amino acid carrier.110 In Excess amounts of glutamate seem to have
humans the arterial plasma aspartate level is been employed in cardioplegic and reperfusion
very low and mainly glutamate is taken up by solutions.103 In anesthetized patients the issue
the ischemic and postischemic heart, suggest- of transient adverse effects is of little signifi-
ing a physiologically more important role for cance and explains why the issue of dosages
glutamate.13,14,46,82 Current knowledge, thus, has received limited attention. However,
indicates that glutamate is of particular potential applications for intravenous gluta-
importance for the ischemic and postischemic mate infusions include awake patients during
human heart, whereas similar data for aspar- the pre- and postoperative course, and patients
tate is lacking. with acute myocardial ischemia in the coro-
nary care unit. Therefore, establishing a dos-
Dosage and Toxicity of Glutamate age of glutamate that would supply the needs
In rodents exogenous administration of of the heart with a minimum risk of side effects
glutamate is associated with dose dependent is desirable.
neurotoxicity.111 In primates glutamate does After CABG surgery myocardial glutamate
not pass the blood brain barrier and the uptake is closely related to arterial plasma lev-
administration of glutamate has, therefore, not els.43,46 However, due to the high capacity of
the heart to extract glutamate, a moderate
Intravenous Metabolic Support in Cardiac Surgery 143

Fig. 12.2. Change in myocardial flux of substrates in controls and patients treated with intravenous glutamate
infusion (early after elective CABG) from the basal state to 60 minutes (mean ± SEM). Hatched bars = Control
group. Grey bars = Glutamate group. Glut = glutamate, Lact = lactate, Ala = alanine, Gluc = glucose and FFA =
free fatty acids. * Indicate statistically significant differences compared with the basal state, * p<0.05, **p<0.01.
Reprinted with permission from: Svedjeholm et al, J Thorac Cardiovasc Surg 1996; 112:1468-77. © Mosby-Year
Book Inc.

Fig. 12.3. Change in hemodynamic state in controls and patients treated with intravenous glutamate infusion
(early after elective CABG) from the basal state to 60 minutes expressed in percentages (mean ± SEM). Hatched
bars = Control group. Grey bars = Glutamate group. CI = cardiac index, HR = heart rate, MAP = mean arterial
pressure, SVRI = systemic vascular resistance index, LVSWI = left ventricular stroke work index. * Indicate
statistically significant differences compared with the basal state, * p<0.05, **p<0.01. Reprinted with permission
from: Svedjeholm et al, J Thorac Cardiovasc Surg 1996; 112:1468-77. © Mosby-Year Book Inc.
144 Ischemia-Reperfusion Injury in Cardiac Surgery

increase of arterial glutamate sufficed to opti- limited ischemic insult, may be expected to
mize myocardial glutamate uptake after elec- respond better to inotropic stimulation than
tive CABG. A significant increase in the myo- seriously injured hearts. In cardiac failure
cardial uptake of glutamate was observed caused by severe myocardial ischemia or evolv-
during glutamate infusion but increasing ing myocardial infarction the use of inotropic
arterial (whole blood) levels by more than drugs can be detrimental. Basic science has
100-200 mmol/L was not associated with a demonstrated that adrenergic stimulation
further substantial increment in myocardial aggravates ischemia and precipitates myocar-
glutamate uptake (Fig. 12.4). Glutamate dial infarction.9 Inotropes have also been
infusion caused a dose dependent linear shown to cause de-energization of the myo-
increase of arterial glutamate levels and an cardium, and if instituted prematurely it may
infusion rate of 30-40 mg/kg BW/hour may imply that the condition of the heart is aggra-
be sufficient to supply the needs of the heart vated by further metabolic derangement. 8
(unpublished data). The adequacy of this dos- However, as the clinical usefulness of
age remains to be confirmed in high-risk alternative treatments other than mechanical
patients. circulatory support has not been demon-
Recently published data show the pH-de- strated, conventional pharmacological treat-
pendence of L-glutamate transport in sar- ment has prevailed in clinical practice. It has
colemmal vesicles from rat heart. Intravesicular been assumed that inotropic drugs are required
acidosis enhanced Na+-dependent glutamate to treat myocardial stunning and to avoid det-
transport, whereas acidification of the in- rimental effects of hypoperfusion. These issues
cubation medium enhanced Na +-indepen- will be challenged below by a metabolic strat-
dent uptake of glutamate.110 Therefore, aci- egy that implies reliance on metabolic mea-
dosis in ischemic heart regions could sures for the enhancement of myocardial
explain the increased uptake of glutamate recovery.
observed in ischemic and postischemic
hearts.13,14,46,82,107,112,116 It remains to be clari- Metabolic Strategy
fied to what extent myocardial glutamate Parallel to metabolic studies, our research
uptake is governed by myocardial require- group has applied metabolic treatment in clini-
ments (acidosis) and to what extent it may be cal practice, to gain experience and to find out
limited by saturable carriers. to what extent metabolic support can replace
traditional treatment of cardiac failure. As data
Clinical Experience from metabolic studies have emerged and our
with Metabolic Support experience has increased, a metabolic strategy
has gradually evolved. This strategy is com-
in Cardiac Surgery piled of metabolic, pharmacological and physi-
The majority of patients undergoing car-
ological principles that have been found to
diac surgery will do well independent of the
enhance myocardial tolerance to ischemia and
treatment given postoperatively. Therefore, it
to promote recovery after ischemia. Metabolic
is tempting to consider the hazards associated
support found to be adequate regarding dos-
with adrenergic stimulation of the heart to be
age and choice of substrates has been chosen
of theoretical interest only. Paradoxically over-
and modified to achieve metabolic effects with
use of inotropes may give an impression of
stable hemodynamic conditions. Therefore,
efficiency that may be lacking when these
the metabolic strategy includes general mea-
drugs are really “needed”. Many patients are
sures in all patients to reduce the metabolic
treated with inotropes because of low blood
stress on the heart, and metabolic support in
pressure caused by low systemic vascular re-
selected cases. Contrary to common belief we
sistance or hypovolemia. Furthermore, hearts
have found that these principles can replace
with benign causes of cardiac failure, such as
traditional treatment as the main therapy in
residual depressant effect of cardioplegia or
the postoperative setting after cardiac surgery.
Intravenous Metabolic Support in Cardiac Surgery 145

Fig. 12.4. Myocardial glutamate uptake (∝mol/min) [bars] and arterial glutamate level during intravenous glutamate
infusion early after CABG (∝mol/L) [straight line + squares] according to whole blood analyses (Mean ± SEM).
* Indicate statistically significant differences compared with the basal state, * p < 0.05, ** p < 0.01. Reprinted with
permission from: Svedjeholm et al, J Thorac Cardiovasc Surg 1996; 112:1468-77. ©Mosby-Year Book Inc.

General Measures where myocardial energy reserve may be lim-


The general measures include conventional ited and in impending or evolving myocardial
pharmacological treatment of myocardial infarction. Accordingly, Lazar showed detri-
ischemia and unconventional measures to mental effects of premature use of inotropic
reduce myocardial energy expenditure and sys- drugs for weaning from CPB.10 Inotropic
temic oxygen demand.117 In patients with car- drugs for weaning from CPB are therefore
diac failure care is taken to reduce strain on avoided or the use of them delayed. As car-
the heart by reducing systemic oxygen diac failure is usually apparent on weaning
demand, which is, in fact the main determi- from CPB, prolonged unloading of the heart
nant of cardiac output. Mixed venous oxygen on CPB and metabolic support are used to
saturation (SvO2) reflects the balance between enhance myocardial recovery. Volume work by
cardiac output and systemic oxygen consump- the heart (in favor of pressure work) is pro-
tion and indicates whether cardiac output can moted by after load reduction with nitroprus-
adequately provide the peripheral tissues with side. In patients with severe postoperative car-
oxygen. This physiological approach toward diac failure a systolic blood pressure of
the treatment of postoperative cardiac failure 80-100 mm Hg is accepted if urinary output
helps to avoid overtreatment, that is, stimu- is adequate and the patient has no critical
lating cardiac output more than necessary. In arterial stenosis. If SvO2 or urinary output
patients with signs of inadequate hemodynam- suggests that cardiac output is inadequate,
ics, systemic oxygen demand is lowered by inotropes are used in low dosages. A mechani-
sedatives and muscle relaxants. Inotropic drugs cal assist device is preferred in favor of increas-
increase myocardial energy expenditure ing the dose of inotropic drugs such as
directly by increasing myocardial oxygen dobutamine above 4-5 ∝g/kg/min.
demand and indirectly by increasing systemic
oxygen demand.6,7 The negative effects of ino- Metabolic Support
tropic drugs may be expected to be particu- Metabolic support has been used in selected
larly pronounced during early reperfusion patients and included the use of glutamate
146 Ischemia-Reperfusion Injury in Cardiac Surgery

infusion and high-dose GIK (glucose-insulin- High-Dose GIK (Glucose-Insulin-


potassium). Although theoretically attractive, Potassium)
the role of prophylactic treatment with GIK GIK (glucose-insulin-potassium) treatment
or glutamate in cardiac surgery remains to be has mainly been used to treat or prevent car-
clarified. At present preoperative GIK is con- diac failure on weaning from CPB and in the
sidered in energy depleted hearts. As glutamate early postoperative setting. The rationale in
improves myocardial tolerance to ischemia in this setting is to counteract the negative effects
coronary patients during exercise tests and of systemic trauma metabolism on the heart;
pacing we have used glutamate preoperatively reduce plasma FFA, give the heart carbohy-
in severely unstable patients.25,104 In contrast, drate substrates and enhance full glucose oxi-
the rationale for postoperative metabolic dation.43 Due to the insulin resistance after
treatment in cardiac surgery has been more cardiac surgery, high doses of insulin may be
clearly defined. First the metabolic abnor- required to achieve these objectives.41,43,45 We
malities and the state of insulin resistance have employed a high-dose GIK regime that
have been demonstrated.41,45,82 The desired will ensure maximal metabolic effects in most
metabolic effects, and positive hemody- patients, without having the pronounced
namic effects, have been achieved with vasodilative properties of the “hemodynamic
glutamate and high-dose GIK.6,26,41,45 For doses” of insulin described in previous stud-
treatment of cardiac failure after CABG the ies.72,118 The reason is that after load reduc-
combined use of glutamate and GIK seems tion with nitroprusside is easier to control in
advisable by addressing the metabolic conse- critically ill patients. Moderate hyperglycemia
quences of both myocardial ischemia and the is employed as systemic glucose uptake after
systemic neuroendocrine stress response.4 cardiac surgery can be further enhanced (in
However, it remains to be shown that the com- spite of high plasma insulin) by increasing
bined use of glutamate and GIK provides ad- blood glucose up to 10 ∝mol/L.57
ditional metabolic benefit. In clinical practice a 30% glucose solution,
supplemented by 10 ∝mol Mg and 40 mmol
Glutamate Infusion of phosphate/1000 ml, is infused at a rate of
Glutamate infusion has been used preop- 60-100 ml/hour. We have found it advisable
eratively and during the early stages of sur- to start with approximately 1ml/kg/hour. The
gery to increase myocardial tolerance to infusion rate is determined by regular checks
ischemia in severely unstable patients. As of s-glucose to achieve a stable blood glucose
glutamate enhances postischemic recovery of level between 7-12 ∝mol/L. We prefer to keep
myocardial metabolism it has also been used blood glucose just below 10 ∝mol/L.
to treat or prevent cardiac failure on weaning Insulin of a fast-acting type (Actrapid
from CPB. According to data that we are about Novo) is infused at a rate of 1 IU/kg/hour for
to publish an infusion rate of 30-40 mg/kg/ 6 hours. A bolus dose of 25 IU is injected i.v.
hour should be adequate in most patients to after 5 minutes to achieve an early steady state
optimize myocardial uptake of glutamate. In metabolic effect. After the insulin infusion has
clinical practice an infusion rate of 1.5 ml/kg/ been completed, the glucose infusion is main-
hour of a 0.125M solution has been employed tained for another 8-18 hours, the infusion
and has so far not been associated with any ill rate gradually decreased according to blood
effects. The total volume infused has usually glucose level. This treatment is a hyper-
been 250 ml, however, in exceptional cases the insulinemic glucose clamp, therefore, blood
infusion has been continued to a total volume glucose levels in the short perspective are
of 500 ml. As we have sometimes experienced influenced only by the rate of glucose infu-
that the patients “sag” a little after stopping sion. Consequently, the high-dose GIK treat-
glutamate, we now reduce the infusion rate to ment is easy to manage and stable glucose lev-
half the original when 1/2 hour remains els are routine. However, this treatment
according to the original infusion rate. requires careful attention and the glucose
Intravenous Metabolic Support in Cardiac Surgery 147

infusion should not be stopped until the ef- ible cardiac failure. It may be argued that it
fect of insulin has subsided. was time and prolonged unloading of the heart
Potassium is infused separately but s- by CPB rather than metabolic treatment that
potassium is allowed to decrease to 3.5 ∝mol/ improved the condition of the hearts. How-
L to avoid rebound hyperkalemia at the dis- ever, there is no contradiction between the use
continuation of insulin infusion. of prolonged CPB and metabolic support.
As the treatment proceeds, some degree of Other investigators have achieved encourag-
vasodilation will develop in most patients. ing results in critically ill patients after cardiac
Nitroprusside is discontinued and in approxi- surgery employing the principles of substrate
mately one third of the cases small to moder- enhancement and unloading of the heart.93
ate doses of a counteracting drug (angiotensin Both principles enhance the recovery of myo-
II or noradrenaline) has to be instituted. cardial metabolism. In experimental coronary
ligation the beneficial effects of GIK and IABP
Metabolic Support in Severe on myocardial metabolism are additive.120 We
Cardiac Failure have found substantial uptake of both glucose
Initially metabolic support was used mainly and glutamate in the anesthetized patient dur-
in patients with severe cardiac failure to gain ing CPB (unpublished). Metabolic support,
experience and to find out to what extent meta- however, may enhance the speed of recovery
bolic support can be used to replace inotropes by improving myocardial uptake of glucose
and mechanical assist devices.119 In 16 (3.1%) and glutamate and by reducing plasma FFA.
out of 515 patients cardiac failure on weaning Furthermore a sustained effect is acquired by
from CPB (cardiopulmonary bypass) was con- continuing metabolic support into the early
sidered severe enough to have justified the use postoperative course when the consequences
of IABP treatment. The average systolic arte- of the neuro-endocrine stress response may
rial blood pressure (SAP) was 54 ± 19 mmHg become more obvious. The hemodynamic
when the decision to start metabolic support recovery within six hours compared with the
was taken. The condition of most patients was 2-3 days generally required with conventional
too poor to allow further hemodynamic mea- mechanical assist devices is in keeping with
surements or blood sampling. The first SVO2 this assumption.73,121
and cardiac index (CI) obtained were on average These results should not be interpreted as
45 ± 7% and 1.6 ± 0.4 L/min/m2 respectively. though avoidance of IABP or other mechani-
These patients were treated by unloading of cal assist devices is a major objective of the
the heart with prolonged CPB and metabolic metabolic strategy. Unloading of the heart is
support with glutamate and high-dose GIK. an attractive mode of treatment from a meta-
Thirteen out of 16 patients could be bolic point of view.122,123 In fact due to this
weaned from CPB after an estimated average early encouraging experience we have broad-
prolongation of CPB with 75 minutes. A rapid ened the indications for both metabolic sup-
improvement of hemodynamic performance port and mechanical assist devices to reduce
was seen in the first hour after weaning from the need for inotropic drugs further.
CPB, and almost full recovery within 6 hours
(Fig. 12.5). The average dobutamine dose in Metabolic Strategy in Surgery
the first 6 hours was 2.2 ± 1.8 ∝g/kg/min. for Ischemic Heart Disease
Three patients failed to wean from CPB and Data from 785 consecutive patients oper-
required a mechanical assist device. None of ated by our group for ischemic heart disease
these patients, however, showed any signs of (745 CABG and 40 CABG + valve procedures)
reversible cardiac failure and they subsequently according to the metabolic strategy are cur-
died. One further patient died of sepsis one rently undergoing analysis (unpublished). The
week postoperatively. Thus, with a question- average age was 65 years and the proportion
mark for one patient, metabolic support was of females was 20%. 229 patients had unstable
associated with a 100% success rate in revers-
148 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 12.5. Hemodynamic recovery in patients with severe cardiac failure on weaning from CPB treated with
intravenous glutamate and high-dose GIK (n=13). The change in systolic arterial blood pressure (SAP, n=13; right
axis) and cardiac index (CI, n=7; left axis) after institution of metabolic support and the change in mixed venous
oxygen saturation (SVO2, n=13; right axis) after weaning from CPB. Data are presented as mean ± SEM. The
horizontal bars depict the time used for metabolic treatment. G = glutamate, GIK = high-dose Glucose-Insulin-
Potassium. Reprinted with permission from: Svedjeholm et al, Ann Thorac Surg 1995; 59:S23-30. © Elsevier
Science Inc.

angina (29.2%) and 108 patients (13.8%) had (0.1%). No patient developed evident signs
severely compromised left ventricular function of intestinal ischemia. Average stay in the ICU
preoperatively (ejection fraction ∀ 0.35). Over- was 1.6 days and the median time on the ven-
all 96% of the patients were weaned from CPB tilator 8 hours. The 30-day mortality was
without inotropic drugs. In Table 12.1 the use 0.9%.
of dobutamine and metabolic support from Inotropic drugs improve cardiac output
1991-1995 is shown. Despite an increasing and thus can contribute to termination of
proportion of high risk patients, the need for CPB. However, there are to our knowledge
inotropes for weaning from CPB was gradu- no evidence that these drugs improve outcome
ally abolished as the use of metabolic support in cardiac surgery. After cardiac surgery
increased. This was not achieved at the price inotropic drugs cause a marked increase in
of a poorer hemodynamic state according to myocardial oxygen demand and aggravate the
SvO2 measurements on arrival to ICU. How- metabolic condition of the heart.6 From car-
ever, in the ICU a small dose of dobutamine diology practice we know that reduction of
to stimulate urinary output was later insti- adrenergic stress with beta-blockers reduces
tuted in 18% of the patients overall. The mortality in myocardial infarction.124 There
average dose of dobutamine when used was is no reason to believe that a myocardial inf-
2.2 ± 1.4 ∝g/kg/min. The maximum dobu- arct in association with cardiac surgery would
tamine dose in an individual patient was differ in this respect. A high incidence of myo-
5.7 ∝g/kg/min and during the last two-year cardial ischemia and myocardial infarction has
period no patient received a dobutamine dose been reported when inotropes are used to ter-
exceeding 5 ∝g/kg/min. PDA inhibitors were minate CPB.125 Another negative aspect is that
used in 0.5% and mechanical assist devices in adrenergic drugs also increase the metabolic
1.4% of the cases. One patient required dialy- rate in the peripheral tissues.6,7 As the main
sis because of postoperative renal failure purpose of the heart is to supply the periph-
Table 12.1. Changes in some risk groups and trends in treatment from 1991 to 1995 in our clinical experience with the metabolic strategy
(n=785 patients).

Year Age ∃ 70 EF ∀ 0.35 CABG Inotropes SvO2% Dobutamine Dobutamine Dobutamine Glutamate GIK
+ valve for weaning on arrival ∝g/kg/min ∝g/kg/min 5 ∝g/kg/min
from CPB to ICU 0-6 hrs 6-24 hrs

1991 25.9% 6.9% 1.7% 6% 66.5 ± 7.6 2.4 ± 1.5 1.8 ± 1.4 0.9% 1.7% 3.4%
Intravenous Metabolic Support in Cardiac Surgery

1992 29.9% 10.4% 1.7% 5.7% 67.2 ± 7.0 2.2 ± 1.5 2.2 ± 1.2 0.6% 12.6% 5.7%

1993 34.0% 15.2% 5.0% 6.6% 66.7 ± 7.3 2.4 ± 1.0 2.4 ± 1.6 1.0% 16.0% 8.0%

1994 37.8% 17.0% 6.9% 0.5% 66.3 ± 8.4 1.6 ± 1.1 2.3 ± 0.8 0 21.4% 16.8%

1995 35.5% 19.0% 10.3% 0 67.6 ± 7.9 1.3 ± 1.5 2.3 ± 1.4 0 30.8% 25.2%
From left to right the table shows the proportion of patients aged 70 years or older, the proportion of patients with preoperatively compromised left ventricular function,
the proportion of combined CABG + valve procedures, the proportion of patients weaned from CPB with inotropes, average SvO2 on arrival to ICU (intensive care
unit), the average dobutamine dose when used during the first 0-6 hours and 6-24 hours after surgery respectively, the proportion of patients receiving dobutamine
doses exceeding 5 mg/kg/min and finally the proportion of patients treated with glutamate and GIK respectively. Results are presented in percentages or mean ± SD.
149
150 Ischemia-Reperfusion Injury in Cardiac Surgery

eral tissues with oxygen and substrates, this cardial phosphorylation potential.77 It has been
therapy is to some extent self defeating. It has demonstrated that pyruvate enhances contrac-
been claimed that phosphodiesterase inhibi- tility in stunned myocardium by enhancing
tors are less noxious to the diseased heart, but myocardial phosphorylation potential.77 Simi-
this remains to be proved. In chronic conges- lar effects may be expected by other substrates
tive heart failure these drugs have been associ- that can enhance pyruvate oxidation such as
ated with a significant increase in overall mor- GIK and glutamate.6,26
tality.126 Although most patients tolerate Renal function is a sensitive parameter of
inotropes, there is now reason to believe that the adequacy of hemodynamic treatment.
they may have a detrimental influence on out- Also, splanchnic circulation may be jeopar-
come in a subset of patients with cardiac fail- dized in low output syndrome. In our five-
ure caused by a serious ischemic insult or evolv- year experience only one patient (0.1%)
ing myocardial infarction. required dialysis because of postoperative renal
One further disadvantage of inotropic failure and we found no obvious case of intes-
treatment is that the natural course of myo- tinal ischemia. In contrast, an incidence of
cardial recovery is concealed by the effects of renal impairment requiring dialysis of
these drugs. As down regulation of beta 1.2-2.5% has been reported from comparable
receptors in the myocardium occurs the drug centers after adult cardiac surgery.129,130 How-
requirements are increased. Deteriorations in ever, less pronounced changes of renal func-
the hemodynamic condition are therefore fre- tion may have to be evaluated to assess the
quently addressed by increasing the dosage of adequacy of hemodynamic treatment. The
inotropic drugs. This implies that identifica- incidence of postoperative renal failure,
tion of other causes for deterioration may be defined as a postoperative increase of s-creati-
delayed or even overlooked. By employing the nine by 50% or more compared with preop-
metabolic strategy we have improved our erative values, was 2.8% in our experience. In
understanding of the natural postoperative contrast, a 16% incidence of postoperative
course. renal failure (same definition) after routine
Since, this is a report of clinical experience CABG was recently reported.129 Multiple
and not a formal scientific trial, we have no logistic regression analysis singled out the use
data on the metabolic effects of metabolic sup- of adrenergic drugs as the most important
port in these patients. However, in previous determinant for development of postoperative
studies the desired metabolic effects, and posi- renal failure. It may be argued that it was the
tive hemodynamic effects, have been achieved need for, rather than the use of these drugs
with glutamate and high-dose GIK.41 After that was the causative factor. However, our
elective CABG, the parallel sequence of experience shows that cardiac failure can be
metabolic and functional recovery of the heart treated with a low incidence of renal prob-
suggests a casual link between metabolic and lems.119 If adrenergic drugs are used in high
contractile dysfunction.43,26,127 In animal doses, vasoconstrictive properties predominate
experiments a close relationship between and renal perfusion may be adversely affected.
mitochondrial respiration and myocardial In contrast, GIK and amino acid infusion may
function during reperfusion has been demon- enhance renal perfusion.131 Also, the negative
strated.128 It is, therefore, conceivable that the effects of inotropic drugs on myocardial
recovery of myocardial function observed dur- recovery may contribute to more severe and
ing glutamate and high-dose GIK was caused prolonged states of low output syndrome.
by recovery of myocardial metabolism. The Therefore, the choice of treatment may have
maintenance of intracellular calcium homeo- a decisive influence on maintenance of renal
stasis and force development by the contrac- function. A similar link between the use of
tile apparatus are dependent upon the free inotropic drugs and the development of
energy derived from ATP hydrolysis. This intestinal ischemia may also be suspected.
energy of hydrolysis is determined by the myo- Thus, the use of inotropic adrenergic drugs
Intravenous Metabolic Support in Cardiac Surgery 151

may be two-edged, not only from a myocar- myocardial ischemia and infarction.9 However,
dial point of view. as the clinical usefulness of alternative treat-
Several indices of improved outcome ments has not been shown, conventional phar-
(improved hemodynamic recovery, less need macological treatment has prevailed in clini-
for inotropes and IABP, less need for ventila- cal practice. Our clinical experience over a
tor treatment, fewer serious complications, five-year period proves that traditional treat-
lower incidence of postoperative arrhythmias, ment with inotropes can be replaced by meta-
shorter ICU and hospital stay) have been bolic measures, involving metabolic support
reported with GIK in high risk patients.23,66,73 with glutamate and high-dose GIK in selected
Although it remains to be proved that meta- patients. Intravenous infusions of glutamate
bolic interventions improve survival in cardiac and high-dose GIK have provided a consis-
surgery, metabolic treatment evidently offers tently effective alternative in the treatment of
several advantages over traditional inotropic postoperative cardiac failure. The overall re-
treatment, 1) Myocardial metabolic derange- sults have been satisfactory and the low inci-
ment—an important cause of postoperative dence of renal problems has been particularly
cardiac failure is treated. Whereas inotropic gratifying. Further studies, though, are needed
drugs impede metabolic recovery 2) Glucose to delineate the optimal metabolic treatment
and amino acids add energy to the heart for postischemic cardiac failure. More impor-
whereas inotropes drain energy from the heart tant, the role of prophylactic metabolic treat-
3) Glucose and amino acids increase myocar- ment in cardiac surgery and myocardial
dial tolerance to ischemia whereas inotropes ischemia remains to be clarified.
aggravate ischemia and may precipitate myo-
cardial infarction 4) GIK and amino acids are References
reported to improve renal perfusion whereas 1. Svedjeholm R, Dahlin LG, Olin C. Periop-
adrenergic drugs in high doses impair renal erative myocardial infarction (PMI) in cardiac
perfusion. surgery—risk factors and clinical outcome.
Brit J Anaesth 1996; 76:(Abstract)

Summary 2. Slogoff S, Keats AS. Does perioperative myo-


cardial ischemia lead to postoperative myo-
The availability of different metabolic sub- cardial infarction? Anesthesiology 1985;
strates can evidently influence postischemic 62:107-114.
functional recovery and the damage incurred 3. Smith RC, Leung JM, Mangano DT. Post-
during episodes of myocardial ischemia. It is operative myocardial ischemia in patients
undergoing coronary artery bypass graft sur-
no longer a question of whether metabolic gery. Anesthesiology 1991; 74:464-473.
support is of benefit to the heart or not, rather 4. Svedjeholm R, Håkanson E, Vanhanen I.
a question of defining the conditions under Rationale for metabolic support with amino
which metabolic interventions are beneficial. acids and glucose-insulin-potassium (GIK) in
Basic research, studies on human myocardial cardiac surgery. Ann Thorac Surg 1995;
59:S15-S22
metabolism after cardiac surgery and available 5. Fowler MB, Alderman EL, Oesterle SN et al.
experience of metabolic interventions provide Dobutamine and dopamine after cardiac sur-
a solid rationale for metabolic support in post- gery: greater augmentation of myocardial
operative cardiac failure. The rationale for blood flow with dobutamine. Circulation
1984; 70 (suppl I):I 103-111.
metabolic treatment of postoperative cardiac 6. Svedjeholm R, Hallhagen S, Ekroth R et al.
failure is further supported by the drawbacks Dopamine and High-dose Insulin infusion
of traditional treatment. Adrenergic inotropic (Glucose-Insulin-Potassium) after a cardiac
drugs improve the hemodynamic condition, operation. Effects on myocardial metabolism.
but at the price of a marked increase in myo- Ann Thorac Surg 1991; 51:262-270.
cardial energy expenditure.5,6,10 This puts fur-
ther strain on the already compromised energy
metabolism of the heart, and may precipitate
152 Ischemia-Reperfusion Injury in Cardiac Surgery

7. Hiesmayr M, Haider WJ, Grubhofer G et al. 21. Berggren H, Ekroth R, Herlitz J et al. Im-
Effects of dobutamine versus insulin on car- proved myocardial protection during cold
diac performance, myocardial oxygen de- cardioplegia by means of increased myocar-
mand, and total body metabolism after coro- dial glycogen stores. Thorac Cardiovasc Surg
nary artery bypass grafting. J Cardiothorac & 1982; 30:389-392.
Vasc Anesth 1995; 9:653-658. 22. Lolley DM, Ray JFI, Myers WO et al. Im-
8. Zhou Z, Lasley RD, Hegge JO et al. Myo- portance of preoperative myocardial glycogen
cardial stunning: a therapeutic conundrum. J levels in human cardiac preservation. Prelimi-
Thorac Cardiovas Surg 1995; 110:1391-1400. nary report. J Thorac Cardiovasc Surg 1979;
9. Maroko PR, Kjekshus JK, Sobel BE et al. 78:678-687.
Factors influencing infarct size following ex- 23. Oldfield GS, Commerford PJ, Opie LH. Ef-
perimental coronary artery occlusions. Circu- fects of preoperative glucose-insulin-potassium
lation 1971; 43:67-82. on myocardial glycogen levels and on com-
10. Lazar HL, Buckberg GD, Foglia RP et al. plications of mitral valve replacement. J
Detrimental effects of premature use of ino- Thorac Cardiovasc Surg 1986; 91:874-878.
tropic drugs to discontinue cardiopulmonary 24. Thomassen A, Nielsen TT, Bagger JP et al.
bypass. J Thorac Cardiovasc Surg 1981; Antianginal and cardiac metabolic effects of
82:18-25. low-dose glucose infusion during pacing in
11. Lewandowski ED, Ingwall JS. The physiologi- patients with and without coronary artery
cal chemistry of energy production in the disease. Am Heart J 1989; 118:25-32.
heart. In: Schlant RC, Alexander RW, eds. 25. Thomassen A, Nielsen TT, Bagger JP et al.
Hurst’s The Heart. 8 ed. New York: Antiischemic and metabolic effects of gluta-
McGraw-Hill, 1994:153-164. mate during pacing in patients with stable
12. Opie L: Carbohydrates and Lipids. In: The angina pectoris secondary to either coronary
Heart. 1 ed. London: Grune & Stratton, artery disease or syndrome X. Am J Cardiol
1984:111-135. 1991; 68:291-295.
13. Mudge GH, Mills RM, Taegtmeyer H et al. 26. Svedjeholm R, Vanhanen I, Håkanson E et
Alterations of myocardial amino acid metabo- al. Metabolic and hemodynamic effects of
lism in chronic ischemic heart disease. J Clin intravenous glutamate infusion early after
Invest 1976; 58:1185-1192. coronary surgery. J Thorac Cardiovasc Surg
14. Thomassen A, Nielsen TT, Bagger JP et al. 1996; 112:1468-1477.
Myocardial glutamate and alanine exchanges 27. Engelman RM, Rousou JA, Flack JE et al.
related to carbohydrate metabolism in pa- Reduction of infarct size by systemic amino
tients with normal and stenotic coronary ar- acid supplementation during reperfusion. J
teries. Clin Physiol 1984; 4:425-434. Thorac Cardiovasc Surg 1991; 101:855-859.
15. Taegtmeyer H. Metabolic responses to hy- 28. Maroko PR, Libby P, Sobel BE et al. Effect
poxia; increased production of succinate by of Glucose-Insulin-Potassium infusion on
rabbit papillary muscles. Circ Res 1978; myocardial infarction following experimental
43:808-815. coronary artery occlusion. Circulation 1972;
16. Sanborn T, Gavin W, Berkowitz S et al. 45:11601175
Augmented conversion of aspartate and 29. Simonsen S, Kjekshus JK. The effect of free
glutamate to succinate during anoxia in rab- fatty acids on myocardial oxygen consump-
bit heart. Am J Physiol 1979; 237:H535- tion during atrial pacing and catecholamine
H541. infusion in man. Circulation 1978; 58:
17. Rau EE, Shine KI, Gervais A et al. Enhanced 484-491.
mechanical recovery of anoxic and ischemic 30. Vik-Mo H, Mjos OD. Influence of free fatty
myocardium by amino acid perfusion. Am J acids on myocardial oxygen consumption and
Physiol 1979; 236:H873-H879 ischemic injury. Am J Cardiol 1981; 48:
18. Pisarenko OI, Baranov AV, Pomerantsev EV 361-365.
et al. Myocardial metabolism of glutamate 31. Liedtke AJ, Nellis S, Neely JR. Effects of
and left ventricular function in patients with excess free fatty acids on mechanical and
coronary arterial disease. Int J Cardiol 1989; metabolic function in normal and ischemic
23:43-52. myocardium in swine. Circ Res 1978; 43:
19. Safer B. The metabolic significance of the 652-661.
malate-aspartate cycle in heart. Circ Res 1975; 32. Henderson AH, Craig RJ, Gorlin R et al. Free
37:527-533. fatty acids and myocardial function in per-
20. Pisarenko OI, Oleynikov OD, Shulzhenko VS fused rat hearts. Cardiovasc Res 1970; 4:
et al. Association of myocardial glutamate and 466-472.
aspartate pool and functional recovery of post- 33. Borst P, Loos JA, Christ EJ et al. Uncou-
ischemic heart. Biochem Med Metab Biol pling activity of long-chain fatty acids.
1989; 42:105-117. Biochim Biophys Acta 1962; 62:509-518.
Intravenous Metabolic Support in Cardiac Surgery 153

34. Lucas SK, Kanter KR, Schaff HV et al. Re- 47. Apstein CS, Gravino FN, Haudenschild CC.
duced oxygen extraction during reperfusion: Determinants of a protective effect of glucose
A consequence of global ischemic arrest. J and insulin on the ischemic myocardium.
Surg Res 1980; 28:434-441. Effects on contractile function, diastolic com-
35. Kanter KR, Glower DD, Schaff HV et al. pliance, metabolism, and ultrastructure dur-
Mechanism of defective oxygen extraction ing ischemia and reperfusion. Circ Res 1983;
following global ischemia. J Surg Res 1981; 52:515-526.
30:482-488. 48. Rackley CE, Russel RO, Rogers JW et al.
36. Lazar HL, Buckberg GD, Manganaro AJ et Glucose-Insulin-Potassium in acute myocar-
al. Reversal of ischemic damage with amino dial infarction. Ann Rev Med 1982; 33:
acid substrate enhancement during reper- 375-383.
fusion. Surgery 1980; 80:702-709. 49. Lopaschuk GD, Stanley WC. Glucose me-
37. Peuhkurinen KJ. Regulation of the tricarboxy- tabolism in the ischemic heart [editorial com-
lic acid pool size in heart muscle. J Mol Cell ment]. [Review] [22 refs]. Circulation 1997;
Cardiol 1984; 16:487-495. 95:313-315.
38. Pisarenko O, Studneva I, Khlopkov V et al. 50. Wheeler TJ, Fell RD, Hauck MA. Translo-
An assessment of anaerobic metabolism dur- cation of two glucose transporters in heart:
ing ischemia and reperfusion in isolated effects of rotenone, uncouplers, workload,
guinea pig heart. Biochim Biophys Acta 1988; palmitate, insulin and anoxia. Biochim
934:55-63. Biophys Acta 1994; 1196:191-200.
39. Kobayashi K, Neely JR. Effects of ischemia 51. McNulty PH, Sinusas AJ, Shi CQ et al. Glu-
and reperfusion on pyruvate dehydrogenase cose metabolism distal to a critical coronary
activity in isolated rat hearts. J Mol Cell stenosis in a canine model of low-flow myo-
Cardiol 1983; 15:359-367. cardial ischemia. J Clin Invest 1996; 98:
40. Teoh KH, Mickle DAG, Weisel RD et al. 62-69.
Decreased postoperative myocardial free fatty 52. Zaninetti D, Greco-Perotto R, Assima-
acid oxidation. J Surg Res 1988; 44:36-44. copoulos-Jeannet F et al. Effects of insu-
41. Svensson S, Svedjeholm R, Ekroth R et al. lin on glucose transport and glucose trans-
Trauma metabolism and the heart. Uptake of porters in rat heart. Biochem J 1988; 250:
substrates and effects of insulin early after 277-283.
cardiac operations. J Thorac Cardiovasc Surg 53. Stanley WC, Lopaschuk GD, Hall JL et al.
1990; 99:1063-1073. Regulation of myocardial carbohydrate me-
42. Svedjeholm R, Vanhanen I, Hakanson E et tabolism under normal and ischaemic condi-
al. Metabolic and hemodynamic effects of tions. Potential for pharmacological interven-
intravenous glutamate infusion early after tions. [Review] [184 refs]. Cardiovasc Res
coronary operations. J Thorac Cardiovas Surg 1997; 33:243-257.
1996; 112:1468-1477. 54. Ekroth R, Berggren H, Björntorp J et al.
43. Svedjeholm, R. Myocardial uptake of sub- Effect of valvular aortic stenosis on insulin
strates early after cardiac surgery. A clinical sensitivity. Scand J Thorac Cardiovasc Surg
study of metabolic derangement and recov- 1982; 16:141-144.
ery with special reference to effects of dopam- 55. Nilsson F, Ekroth R, Milocco I et al.
ine and insulin. 7-54. 1990. Acta Universita- Splanchnic glucose balance and insulin resis-
tis Upsaliensis. Thesis/Dissertation tance in the early postoperative phase of car-
44. Safer B, Williamson JR. Mitochondrial-cyto- diac surgery. JPEN J Parenter Enteral Nutr
solic interactions in perfused rat heart. Role 1988; 12:574-578.
of coupled transamination in repletion of cit- 56. Zaninetti D, Greco-Perotto R, Jeanrenaud B.
ric acid cycle intermediates. J Biol Chem Heart glucose transport and transporters in
1973; 248:2570-2579. rat heart: regulation by insulin, workload and
45. Svensson S, Ekroth R, Milocco I et al. Glu- glucose. Diabetologia 1988; 31:108-113.
cose and lactate balances in heart and leg af- 57. Nilsson F, Bake B, Berglin E et al. Glucose
ter coronary surgery: influence of insulin in- and insulin infusion directly after cardiac sur-
fusion. Scand J Thorac Cardiovasc Surg 1989; gery: effects on systemic glucose uptake, cat-
23:145-150. echolamine excretion, O2 consumption, and
46. Svedjeholm R, Ekroth R, Joachimsson PO et CO2 production. JPEN J Parenter Enteral
al. Myocardial uptake of amino acids and Nutr 1985; 9:159-164.
other substrates in relation to myocardial oxy- 58. Fischer Y, Bottcher U, Eblenkamp M et al.
gen consumption four hours after cardiac Glucose transport and glucose transporter
surgery. J Thorac Cardiovasc Surg 1991; GLUT4 are regulated by product(s) of inter-
101:688-694. mediary metabolism in cardiomyocytes. Bio-
chemical Journal 1997; 321:629-638.
154 Ischemia-Reperfusion Injury in Cardiac Surgery

59. Scheuer J, Stezoski SW. Protective role of 72. Svensson S, Ekroth R, Nilsson F et al. Insu-
increased myocardial glycogen stores in car- lin as a vasodilating agent in the first hour
diac anoxia in the rat. Circ Res 1970; after cardiopulmonary bypass. Scand J Thorac
27:835-849. Cardiovasc Surg 1989; 23:139-143.
60. Wiese S, Askanazi J. Glucose/Insulin/Potas- 73. Gradinac S, Coleman GM, Taegtmeyer H et
sium therapy: A reevaluation of myocardial al. Improved cardiac function with Glucose-
benefits during cardiopulmonary bypass. J Insulin-Potassium after aortocoronary bypass
Cardiothorac Vasc Anesth 1992; 6:517-520. grafting. Ann Thorac Surg 1989; 48:484-489.
61. Girard C, Quentin P, Bouvier H et al. Glu- 74. Hall JL, Stanley WC, Lopaschuk GD et al.
cose and insulin supply before cardiopulmo- Impaired pyruvate oxidation but normal glu-
nary bypass in cardiac surgery: A double-blind cose uptake in diabetic pig heart during
study. Ann Thorac Surg 1992; 54:259-263. dobutamine-induced work. Am J Physiol
62. Haider W, Eckersberger F, Losert U et al. 1996; 271:H2320-H2329.
Improved myocardial protection in open heart 75. Malmberg K, Ryden L, Efendic S et al. Ran-
surgery by massive preoperative insulin sup- domized trial of insulin-glucose infusion fol-
ply. Anaesthesist 1982; 31:124-128. lowed by subcutaneous insulin treatment in
63. Wistbacka JM, Paivi KK, Nuutinen LS. diabetic patients with acute myocardial inf-
Prebypass glucose-insulin-potassium infusion arction (DIGAMI study): effects on mortal-
in elective non-diabetic coronary artery sur- ity at 1 year. J Am Coll Card 1995; 26:57-65.
gery patients. J Cardiothorac Vasc Anesth 76. Teoh KH, Mickle DAG, Weisel RD et al.
1992; 6:517-520. Improving myocardial metabolic and func-
64. Nilsson F, Ekroth R, Milocco I et al. The tional recovery after cardiac arrest. J Thorac
effect of insulin on the leg and splanchnic Cardiovasc Surg 1988; 95:788-798.
balance of glucose and gluconeogenic sub- 77. Lasley RD, Bunger R, Zhou Z et al. Meta-
strates after cardiac surgery. Thorac bolically based treatment of stunned myocar-
Cardiovasc Surg 1988; 36:343-347. dium. J Card Surg 1994; 9:469-473.
65. Brodin LA, Dahlgren G, Ekestrom S et al. 78. Gailis L, Benmouyal E. Endogenous alanine,
Influence of glucose-insulin-potassium on left glutamate, aspartate and glutamine in the
ventricular function during coronary artery perfused guinea pig heart: effect of substrates
bypass grafting. Scand J Thorac Cardiovasc and cardioactive agents. Can J Biochem 1973;
Surg 1993; 27:27-34. 51:11-20.
66. Lazar HL, Philippides G, Fitzgerald C et al. 79. Bittl J, Shine K. Protection of ischemic rab-
Glucose-insulin-potassium solutions enhance bit myocardium by glutamic acid. Am J
recovery after urgent coronary artery bypass Physiol 1983; 245:H406-H412
grafting. J Thorac Cardiovas Surg 1997; 80. Burns AH, Reddy WJ. Amino acid stimula-
113:354-60; discussion 360-2. tion of oxygen and substrate utilization by
67. Svensson SE, Berglin-W-OE, Ekroth R, cardiac myocytes. Am J Physiol 1978; 235:
Milocco I et al. Haemodynamic effects of a E461-E466.
single large dose of insulin in open heart sur- 81. Pisarenko OI. Regulation of cardiac metabo-
gery. Cardiovasc Res 1984; 18:697-701. lism by amino acids in ischemia and hypoxia.
68. Haider W, Benzer H, Coraim F et al. Post- Sov Med Rev A Cardiol 1989; 2:133-167.
operative therapy by means of acute 82. Svedjeholm R, Svensson SE, Ekroth R et al.
parenteral alimentation (APA) with high doses Trauma metabolism and the heart; studies of
of insulin and glucose after open heart sur- heart and leg amino acid flux after cardiac
gery (author’s transl). Anaesthesist 1981; surgery. Thorac Cardiovasc Surg 1990;
30:53-63. 38:1-5.
69. Lucchesi BR, Median M, Kniffen FJ. The 83. Schwalb H, Izhar U, Yaroslavsky E et al. The
positive inotropic action of insulin in the effect of amino acids on the ischemic heart.
canine heart. Eur J Pharmacol 1972; 18: Improvement of oxygenated crystalloid car-
107-115. dioplegic solution by an enriched branched
70. Reikerås O, Gunnes P, Sörlie D et al. amino acid formulation. J Thorac Cardiovasc
Haemodynamic effects of high doses of insu- Surg 1989; 98:551-556.
lin during acute left ventricular failure in 84. Pisarenko OI. Mechanisms of myocardial pro-
dogs. Eur Heart J 1985; 6:451-457. tection by amino acids: facts and hypotheses.
71. Svedjeholm R, Ekroth R, Joachimsson PO et [Review] [88 refs]. Clin & Exper Pharmacol
al. High-dose insulin improves the efficacy of & Physiol 1996; 23: 627-633.
dopamine early after cardiac surgery. A study
of myocardial performance and oxygen con-
sumption. Scand J Thorac Cardiovasc Surg
1991; 25:215-221.
Intravenous Metabolic Support in Cardiac Surgery 155

85. Morita K, Sherman MP, Buckberg GD et al. 96. Pisarenko OI, Portnoy VF, Studneva IM et
Studies of hypoxemic/reoxygenation injury: al. Glutamate-blood cardioplegia improves
Without aortic clamping. V. Role of the ATP preservation in human myocardium.
L-arginine-nitric oxide pathway: the nitric ox- Biomed Biochim Acta 1987; 46:499-504.
ide paradox. J Thorac Cardiovas Surg 1995; 97. Kjellman UW, Björk K, Ekroth R et al. Ad-
110:1200-1211. dition of alpha-ketoglutarate to blood car-
86. Sato H, Zhao ZQ, McGee DS et al. Supple- dioplegia improves cardioprotection. Ann
mental L-arginine during cardioplegic arrest Thorac Surg 1997; 63:1625-1634.
and reperfusion avoids regional postischemic 98. Low SY, Rennie MJ, Taylor PM. Sodium-
injury. J Thorac Cardiovas Surg 1995; dependent glutamate transport in cultured rat
110:302-314. myotubes increases after glutamine depriva-
87. Sanborn T, Gavin W, Berkowitz S et al. tion. FASEB J 1994; 8:127-131.
Augmented conversion of aspartate and 99. Suleiman MS, Chapman RA. Changes in the
glutamate to succinate during anoxia in rab- principal free intracellular amino acids in the
bit heart. Am J Physiol 1979; 237:H535- Langendorff perfused guinea pig heart dur-
H541 ing arrest with calcium-free or high potassium
88. Haas GS, DeBoer LWV, O’Keefe DD et al. media. Cardiovasc Res 1993; 27:1810-1814.
Reduction of postischemic myocardial dys- 100. Kimose HH, Ravkilde J, Helligsö P et al.
function by substrate repletion during reper- Myocardial loss of glutamate after cold chemi-
fusion. Circulation 1984; 70(suppl I):65-74. cal cardioplegia and storage in isolated blood-
89. Lazar HL, Buckberg GD, Manganaro AJ et perfused pig hearts. Thorac Cardiovasc Surg
al. Myocardial energy replenishment and re- 1993; 41:93-100.
versal of ischemic damage by substrate en- 101. Bical OM, Gerhardt MF, Paumier D et al.
hancement of secondary blood cardioplegia Effects of two different crystalloid cardioplegic
with amino acids during reperfusion. J Thorac solutions assessed by myocardial pH, tissue
Cardiovasc Surg 1980; 80:350-359. lactate content and energy metabolism. Eur
90. Weldner PW, Myers JL, Miller CA et al. J Cardiothorac Surg 1996; 10:417-421.
Improved recovery of immature myocardium 102. Reed MK, Barak C, Malloy CR et al. Effects
with L-glutamate blood cardioplegia. Ann of glutamate and aspartate on myocardial sub-
Thorac Surg 1993; 55:102-105. strate oxidation during potassium arrest [see
91. Engelman RM, Dobbs WA, Rousou JH et comments]. J Thorac Cardiovas Surg 1996;
al. Myocardial high-energy phosphate replen- 112:1651-1660.
ishment during ischemic arrest: aerobic ver- 103. Wistbacka J-OM, Lepojärvi MVK, Karlqvist
sus anaerobic metabolism. Ann Thorac Surg KEV et al. Amino acid-enriched glucose-in-
1982; 33:453-458. sulin-potassium infusion improves hemody-
92. Kimose HH, Helligso P, Randsbaek F et al. namic function after coronary surgery. Infu-
Improved recovery after crystalloid cardiople- sions Ther Transfusions Med 1995; 22:82-90.
gia using low-dose glutamate enrichment dur- 104. Thomassen A, Bötker HE, Nielsen TT et al.
ing reperfusion after aortic unclamping: a Effects of glutamate on exercise tolerance and
study in isolated blood-perfused pig hearts. circulating substrate levels in stable angina
Thorac Cardiovasc Surg 1996; 44:118-125. pectoris. Am J Cardiol 1990; 65:173-178.
93. Beyersdorf F, Kirsh M, Buckberg GD et al. 105. Singh J, Garg KN, Garg D et al. Effect of
Warm glutamate/aspartate-enriched blood aspartate and glutamate on experimental myo-
cardioplegic solution for perioperative sudden cardial infarction in rats. Indian J Exp Biol
death. J Thorac Cardiovasc Surg 1992; 1989; 27:621-624.
104:1141-1147. 106. Beyersdorf F, Acar C, Buckberg GD et al.
94. Allen BS, Buckberg GD, Schwaiger M et al. Studies on prolonged acute regional ischemia.
Early recovery of regional wall motion in V. Metabolic support of remote myocardium
patients following surgical revascularization during left ventricular power failure. J Thorac
after eight hours of acute coronary occlusion: Cardiovasc Surg 1989; 98:567-579.
Published erratum appears in J Thorac 107. Pisarenko OI, Lepilin MG, Ivanov VE. Car-
Cardiovasc Surg 1987 Jan;93(1):140. J diac metabolism and performance during L-
Thorac Cardiovasc Surg 1986; 92:636-648. glutamic acid infusion in postoperative car-
95. Rosenkranz ER, Buckberg GD, Laks H et al. diac failure. Clin Science 1986; 70:7-12.
Warm induction of cardioplegia with gluta- 108. Rosenkranz ER, Okamoto F, Buckberg GD
mate-enriched blood in coronary artery pa- et al. Safety of prolonged aortic clamping
tients with cardiogenic shock who are depen- with blood cardioplegia. III. Aspartate enrich-
dent on inotropic drugs and intra-aortic bal- ment of glutamate-blood cardioplegia in en-
loon support. J Thorac Cardiovasc Surg 1983; ergy-depleted hearts after ischemic and
86:507-518. reperfusion injury. J Thorac Cardiovasc Surg
1986; 91:428-435.
156 Ischemia-Reperfusion Injury in Cardiac Surgery

109. Pisarenko OI, Rosenfeldt FL, Langley L et 121. Creswell LL, Rosenbloom M, Cox JL et al.
al. Differing protection with aspartate and Intraaortic balloon counterpulsation: patterns
glutamate cardioplegia in the isolated rat of usage and outcome in cardiac surgery pa-
heart. Ann Thorac Surg 1995; 59:1541-1548. tients. Ann Thorac Surg 1992; 54:11-20.
110. Dinkelborg LM, Kinne RK, Grieshaber MK. 122. Allen BS, Okamoto F, Buckberg GD et al.
Characterization and pH dependence of L- Reperfusion conditions: critical importance of
glutamate transport in sarcolemmal vesicles total ventricular decompression during re-
from rat hearts. Am J Physiol 1995; 268: gional reperfusion. J Thorac Cardiovasc Surg
H194-201. 1986; 92:605-612.
111. Daabees TT, Finkelstein MW, Stegink LD 123. Waldenberger FR, Andres J, Flameng W.
et al. Correlation of glutamate plus aspartate Effects of unloaded reperfusion on mitochon-
dose, plasma amino acid concentration and drial function in the postischemic myocar-
neuronal necrosis in infant mice. Food Chem dium. Artif Organs 1994; 18:206-213.
Toxicol 1985; 23:887-893. 124. Herliz J, Elmfeldt D, Hjalmarson Å et al.
112. Thomassen AR. Myocardial uptake and ef- Effect of metoprolol on indirect signs of the
fects of glutamate during non-ischaemic and size and severity of acute myocardial infarc-
ischaemic conditions. A clinical study with tion. Am J Cardiol 1983; 51:1282-1288.
special reference to possible interrelationships 125. Dupuis JY, Bondy R, Cattran C et al.
between glutamate and myocardial utilization Amrinone and dobutamine as primary treat-
of carbohydrate substrates. Dan Med Bull ment of low cardiac output syndrome follow-
1992; 39:471-488. ing coronary artery surgery: a comparison of
113. Loop FD, Higgins TL, Panda R et al. Myo- their effects on hemodynamics and outcome.
cardial protection during cardiac operations: Journal of Cardiothoracic & Vascular Anes-
Decreased morbidity and lower cost with thesia 1992; 6:542-553.
blood cardioplegia and coronary sinus perfu- 126. Andrews R, Cowley AJ. Phosphodiesterase
sion. J Thorac Cardiovasc Surg 1992; 104: inhibitors. Do the risks outweight the ben-
608-618. efits?. [Review] [38 refs]. Drug Safety 1993;
114. Schaumburg HH, Byck R, Gerstl R et al. 9:404-409.
Monosodium L-glutamate: Its pharmacology 127. Wesslén, Ö. &1-blockade early after coronary
and role in the “Chinese restaurant syn- surgery. A hemodynamic, metabolic and
drome”. Science 1969; 163:826-828. electrophysiologic study with special reference
115. Kenney RA, Tidball CS. Human susceptibil- to effects of metoprolol. 7-45. 1990. Acta
ity to oral monosodium L-glutamate. Am J Universitatis Upsaliensis. Thesis/Dissertation
Clin Nutr 1972; 25:140-146. 128. Weinstein ES, Spector ML, Adams EM et al.
116. Knapp WH, Helus F, Ostertag H et al. Up- Mitochondrial and myocardial performance.
take and turnover L-(13N)-glutamate in the Response to ischemia and reperfusion. Arch
normal human heart and in patients with Surg 1986; 121:324-329.
coronary artery disease. Eur J Nucl Med 129. Andersson LG, Ekroth R, Bratteby LE et al.
1982; 7:211-215. Acute renal failure after coronary surgery—a
117. Håkanson E, Svedjeholm R, Vanhanen I. study of incidence and risk factors in 2009
Physiologic aspects in postoperative cardiac consecutive patients. Thorac Cardiovasc Surg
patients. Ann Thorac Surg 1995; 59:S12-4. 1993; 41:237-241.
118. Svedjeholm R, Ekroth R, Joachimsson PO et 130. Frost L, Pedersen RS, Lund O et al. Progno-
al. High dose insulin improves the efficacy sis and risk factors in acute, dialysis-requir-
of dopamine early after cardiac surgery. A ing renal failure after open-heart surgery.
study of myocardial performance and oxygen Scand J Thorac Cardiovasc Surg 1991;
consumption. Scand J Thorac Cardiovasc 25:161-166.
Surg 1991; 25:215-221. 131. Jeppsson A, Ekroth R, Kirno K et al. Insulin
119. Svedjeholm R, Huljebrant I, Håkanson E et and amino acid infusion after cardiac opera-
al. Glutamate and high-dose glucose-insulin- tions: effects on systemic and renal perfusion.
potassium (GIK) in the treatment of severe J Thorac Cardiovas Surg 1997; 113:594-602.
cardiac failure after cardiac operations. Ann
Thorac Surg 1995; 59:S23-S30
120. Pissarek M, Goos H, Nohring J et al. Ben-
eficial effect of combined glucose-insulin-po-
tassium and mechanical support in acute
myocardial ischaemia. Biomedica Biochimica
Acta 1986; 45:629-636.
Section IV:
Surgical Techniques
A. Temperature of Blood Cardioplegia
CHAPTER 13

Cold/Tepid Cardioplegia
Hendrick B. Barner and Andrew C. Fiore

H
ypothermia has been an essential was best when the heart was maintained at
component of myocardial protection 10-15˚C, but Flaherty and co-workers found
for cardiac operations since the that 10-20˚C was no better than 27˚C for
beginning.1 The evolution of cardioplegia from protecting the myocardium during one hour
crystalloid solutions of intracellular composi- of crystalloid cardioplegia.9 Since the intro-
tion to those of extracellular composition and duction of cardioplegia it has been common
finally to blood based solutions has had the clinical practice to use systemic hypothermia
common theme of cardiac and systemic hy- of 25-30˚C and cardioplegia temperature of
pothermia.2 4-12˚C with topical cardiac cooling to achieve
The focus of many investigators has been a myocardial temperature of 10-20˚C. Many
that of reducing myocardial metabolism to the surgeons have believed and practiced the prin-
lowest possible level during the ischemic ciple that “the colder the heart the better”.
interval so that myocardial energy stores Thus, the introduction of normothermic
(adenosine triphosphate and glycogen) are (warm) blood cardioplegia in 1989 represented
maintained and tissue acidosis is avoided a sudden, dramatic shift in the strategy for
during the ischemic interval. Myocardial intraoperative myocardial protection and has
oxygen consumption of the working heart is resulted in a reassessment of the need for myo-
10-12 ml/100 g/min and of the nonworking cardial and systemic hypothermia during car-
vented heart 6-8 ml/100 g/min.3 The potas- dioplegia.10-12 Despite enthusiasm for and suc-
sium paralyzed heart has a myocardial oxygen cess with warm heart operations some clinical
consumption of 0.31 ml/100 g/min at 22˚C3 and experimental studies have indicated
which is reduced to 0.135 ml/100 g/min at potential limitations of normothermia and it
10-12˚C.4 has been proposed that an intermediate tem-
The optimal degree of hypothermia for perature (tepid) for the body and the heart may
intraoperative myocardial protection has never be preferable.12,13 “Tepid” was defined as a car-
been determined. Myocardial hypothermia to dioplegia temperature of 29˚C which resulted
0.5˚C in the absence of ischemia does not in a myocardial temperature of 28.4˚C13 or
appear to be harmful to the heart.5 St. Tho- 29.2˚C14 while systemic temperature drifted
mas’ solution provided optimal protection to to 33 ± 1˚C. Others have considered “tepid”
the isolated rat heart at 12˚C but the advan- as 32˚C for blood cardioplegia and systemic
tage over 25˚C was not statistically signifi- temperature. Therefore, this chapter broadly
cant.6,7 Tyers and associates8 found that myo- defines tepid as 28-32˚C for cardioplegia, car-
cardial recovery after crystalloid cardioplegia diac and systemic temperatures.

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Cold/Tepid Cardioplegia 159

Goals of Cardioplegia was comparable after 90 minutes of single


coronary occlusion followed by 60 minutes of
Myocardial Protection continuous retrograde blood cardioplegia at
The primary goal of cardioplegia is that of 18˚C or 28˚C with the same systemic tem-
preservation of the heart during conduct of a perature.27 Systolic ventricular function (maxi-
cardiac operation which requires a quiet field mum elastance) and diastolic function (stress-
(absence of mechanical activity) and good strain relationship) showed comparable
visualization (blood free). Until recently the recovery in the same two groups.27
major focus has been that of preserving myo- Left ventricular stroke work indices were
cyte contractility to prevent pump failure better in the tepid (29˚C) than in the cold
which includes conserving cell energy by (4˚C) blood cardioplegia groups after inter-
reducing metabolism to a low level which mittent antegrade and continuous retrograde
allows continued support of vital cell activi- infusion when measured at one and four hours
ties such as ion pumping to maintain the after cardiopulmonary bypass. Following
internal millieu. New knowledge about endo- 60 ± 3 minutes (cold) or 54 ± 4 minutes
thelial function and its pivotal role in the (tepid) of aortic cross clamping.14
behavior of the microvasculature as well as We measured left ventricular stroke work
reperfusion injury has shifted the investigative index after 78 minutes of 4˚C or 70 minutes
focus toward this area. Not only is there cur- of 29˚C intermittent antegrade blood car-
rent interest in the cardiac and systemic tem- dioplegia and found that at 12 hours postop-
perature during cardioplegia but also in the eratively tepid cardioplegia was associated with
effect of cardioplegia on the endothelium. a significantly (p< .05) better index and that
Thus, endothelial preservation may be as at 24 hours the stroke work index in the tepid
important as myocyte preservation. group had exceeded the pre-bypass value
The excellent clinical results attained with (p< .05) whereas the cold group had not.28
warm blood cardioplegia have suggested that In these same patients we assessed left ven-
earlier observations on impairment of some tricular function by volume loading after ter-
cell functions by hypothermia may be more mination of cardiopulmonary bypass and
relevant than previously thought. These measurement with transesophageal echo-
include reduced: membrane stability;15 abil- cardiographic sagittal and transverse images to
ity to utilize glucose,16 and fatty acids;17 mito- allow calculation of the ejection fraction which
chondrial generation of adenosine triphos- increased significantly over the control value
phate18 leading to depressed cell membrane in the tepid group (p< .03) but not in the cold
function;19 activity of the adenosine triphos- group.28
phatase system,20 leading to impaired cell vol-
ume regulation21,22 and decreased ability of the Myocardial Metabolism
sarcoplasmic reticulum to bind calcium;23
mitochondrial state respiration and activity of Oxygen Consumption
citrate synthetase;24 control of intracellular During clinical intermittent antegrade
pH;25 and activity of the sarcoplasmic reticu- combined with continuous retrograde blood
lum with regard to calcium uptake.23,26 cardioplegia myocardial oxygen consumption
during antegrade infusion was greater for tepid
Current Research (29˚C) than for cold (9˚C) (p < .05) cardiople-
gia (3.8 vs 1.5 ml/min).14 Immediately after
cross-clamp release there was an increase in
Ventricular Function
myocardial oxygen extraction which continued
Reperfusion recovery of global left ven-
to increase 10 minutes later and 10 minutes
tricular function as determined by the slope
after discontinuation of bypass with no dif-
of the preload recruitable strike work relation-
ference between the two groups.14
ship after 30, 60 and 90 minutes of reperfusion
160 Ischemia-Reperfusion Injury in Cardiac Surgery

We measured myocardial oxygen extraction minutes later and 10 minutes after discontinu-
immediately after cross-clamp release and also ation of bypass acid production was comparable
found no difference between 78 minutes with to that measured before aortic clamping.14
4˚C and 70 minutes with 29˚C intermittent We found that acid production with cross-
antegrade blood cardioplegia although both clamp release was significantly greater in hearts
groups had depressed oxygen uptake compared receiving tepid (29˚C) intermittent antegrade
to control.28 After five minutes of reperfusion blood cardioplegia than in these receiving 4˚C.
oxygen consumption had returned to pre-cross cardioplegia (4.5 ± 0.9 vs 0.46 ± 1.1 ∝mol/L,
clamp levels in both groups and exceeded these p < .05).28 The cold group released less acid than
values ten minutes after termination of car- prior to cross-clamping (control 6.1 ± 0.9 ∝mol/
diopulmonary bypass (p=ns). L, p < .05) whereas the warm group released a
Myocardial oxygen consumption during comparable amount (5.0 ± 0.6 ∝mol/L). At
the first hour of reperfusion after 90 minutes subsequent times (5 and 10 minutes after
of single coronary occlusion followed by 60 cross-clamp release and 10 minutes after ter-
minutes of continuous retrograde blood car- mination of cardiopulmonary bypass) acid
dioplegia at 18˚C or 28˚C was 5.4 ± 1.4 ml release was comparable for each group and not
02/min/100g and 4.7 ± 1.1 ml 02/min/100g significantly different from control.
(p=ns) respectively.27
Sarcoplasmic Reticulum
Lactate Release Antegrade cold blood (6-10˚C) cardio-
During clinical intermittent antegrade and plegia infused intermittently for two hours
continuous retrograde blood cardioplegia lac- resulted in impaired sarcoplasmic reticulum
tate release during antegrade infusion was calcium uptake (p < .05), and reduced activity
greater for tepid (29˚C) than for cold (9˚C) of calcium-adenosine triphosphatase (p < .05)
(0.1 vs 0.03 ∝mol/min, p<.05) hearts.14 With compared to continuous normothermic
cross-clamp release there was no difference in (37˚C) blood cardioplegia.26 This difference
lactate production between cold and tepid only became significant after 60 minutes of
cardioplegia. Ten minutes later and 10 min- reperfusion. The effect of tepid cardioplegia
utes after bypass was discontinued there was on sarcoplasmic reticulum was not determined.
comparable lactate uptake in both groups.
We observed significant lactate washout
with cross-clamp release in the tepid group, Myocardial Enzyme Release
when comparing 4˚C and 29˚C intermittent The total release of creatinine kinase—
antegrade blood cardioplegia (0.84 ± 014 vs myocardial band (CK-MB) in the 48 hours
2.2 ± 1.2 ∝mol/L, (p=0.0001).28 Ten minutes after operation was similar for intermittent
later hearts receiving 4˚C cardioplegia were antegrade/continuous retrograde 9˚C or 29˚C
extracting lactate while tepid hearts contin- blood cardioplegia.14 We found that total
ued to release lactate (p=ns). Ten minutes after CK-MB release in the 24 hours after opera-
discontinuation of cardiopulmonary bypass tion was greater for 4˚C than for 29˚C inter-
lactate consumption was present in the tepid mittent antegrade blood cardioplegia (1120
group and continued in the cold group (p=ns). ± 141 vs 767 ± 89 U x h/L; p < .04).28 In a
third report peak CK-MB was similar for in-
Acid Release termittent antegrade/continuous retrograde
Acid release during intermittent antegrade blood cardioplegia at 8-10˚C (cold) or 32˚C
and continuous retrograde blood cardioplegia (tepid).29
was greater for tepid (29˚C) than cold car-
dioplegia (9˚C) (p < .05).14 Immediately after
cross-clamp release acid production was simi-
lar for tepid and cold hearts but reduced over
pre-cross-clamp measurements while 10
Cold/Tepid Cardioplegia 161

Electrical Stability associates observed a higher incidence of


After continuous blood cardioplegia at neuropsychologic deficit after normothermic
18˚C or 28˚C there was a trend for more versus tepid or hypothermic cardiopulmonary
countershocks (1.8 ± 1.2 vs 0.6 ± 0.5, p = 0.07) bypass.32 To maintain a body temperature of
to restore an organized cardiac rhythm in the 35˚-37˚C requires active warming during car-
colder group.27 We also found that electrical diopulmonary bypass which may achieve a
defibrillation was more common after 4˚C perfusate temperature greater than 37˚C and
blood cardioplegia than after 29˚C blood car- initiate or exacerbate neuronal injury. Tem-
dioplegia (0.74 ± 0.9 vs 0.08 ± .27, defib- perature management strategy during cardiop-
rillations per patient p<.001).28 ulmonary bypass did not influence perfor-
mance on postoperative neuropsychological
tests.31
Inotropic Support At this time there is not sufficient infor-
We observed a greater need for inotropic mation to indicate the optimum perfusion
drug infusion in the postoperative interval in temperature to minimize the potential for
hearts protected with 4˚C versus 29˚C inter- adverse neurologic outcome. Hyperthermic
mittent antegrade blood cardioplegia (9/25 vs perfusion (active warming) during cardiopul-
2/27; p<.05).28 monary bypass is inappropriate and a tepid
temperature during perfusion may be more
promising.29,31
Neurologic Dysfunction
Hypothermia has been the traditional Fibrinolytic Potential
means of brain protection during cardiopul- Engelman and others determined that there
monary bypass since its inception. The cur- is an increased potential for fibrinolysis in
rent interest in warm heart surgery has patients receiving warm (37˚C) blood cardio-
prompted reassessment of neurologic function plegia as opposed to those receiving tepid
after cardiopulmonary bypass. In some reports (32˚C) or cold (20˚C) blood cardioplegia.29
maintenance of normothermia during car-
diopulmonary bypass has been associated with
increased risk of neurologic or neuro- Summary
psychologic dysfunction30-32 while others have Both clinical studies14,28 observed better left
reported no increased risk of adverse neuro- ventricular function after tepid blood car-
logic outcome.33-35 In one report tepid (32˚) dioplegia; one report was at one and four hours
cardioplegia and body temperature had the post cardiopulmonary bypass, which were the
lowest (2%) incidence of abnormal neurologic only intervals studied, and the other at 12 and
examinations prompting computed tomo- 24 hours but not at earlier intervals.
graphic scanning while normothermia had an The data14,28 on oxygen consumption, lac-
intermediate incidence (9.3%) and cold tate uptake (and release) and acid production
(8˚-10˚C) had the highest (18.9%) (p=ns).29 suggests that with tepid cardioplegia myocar-
Postoperative cognitive function was similar dial metabolic activity is greater than with cold
for patients maintained at 34˚-35˚C (n=30) cardioplegia. Intermittent infusion of car-
or at 28˚C (n=24) with both groups receiving dioplegia is associated with greater washout
intermittent cold (4˚C) blood cardioplegia.36 of lactate and acid with reperfusion in the tepid
The data of Mora et al suggest that if the group indicative of greater metabolic (anaero-
systemic temperature is maintained at 35˚C bic) activity, but with rapid return of meta-
or more by warming during cardiopulmonary bolic activity to control levels with reperfusion.
bypass there is an increased risk of neurologic Only our report28 found reduced release
events after coronary bypass grafting.31 Also of CK-MB following operation in the tepid
this study and the one by Regragui and group compared to 4˚C cardioplegia. Electrical
162 Ischemia-Reperfusion Injury in Cardiac Surgery

stability was greater and the need for inotropic preserved left ventricular function in the high
drugs reduced in clinical tepid cardioplegia.28 dose group.40 In coronary arteries isolated from
these hearts endothelium-dependent maxi-
Future Applications mum relaxation was completely preserved in
Since 1990 there have been striking the high dose group, 18% impaired in the low
changes in the utilization of cardioplegia dose group and 27% impaired in the control
involving route of delivery (antegrade/retro- group.40 Additionally, neutrophil accumula-
grade), mode (intermittent/continuous) tem- tion in the postischemic myocardium was
perature (tepid/normothermic) and to a lesser elevated in the control and low dose groups,
degree of composition. Some surgeons have but significantly reduced in the high dose
adapted normothermic or mildly hypothermic group. Using a similar model with 90 min-
(tepid) cardioplegia with a parallel systemic utes of regional ischemia but with inhibition
temperature. At this time it is unclear whether of basal endothelial nitric oxide release by add-
a consensus will be reached regarding the ideal ing L-nitro-arginine to the blood cardioplegia
temperature or optimal management of the there was increased postischemic/reperfusion
other parameters. It is likely that there will be injury associated with neutrophil adherence.41
continued assessment of the changes initiated Thus, cold blood cardioplegia does not appear
in this interval, with the result that new infor- to be associated with endothelial injury nor
mation will continue to be assimilated into does continuous antegrade normothermic
the arena of cardioplegia as we seek to achieve blood cardioplegia42 so that it is likely that
the best possible myocardial preservation with tepid blood cardioplegia is not harmful to the
the fewest systemic complications. endothelium.
The optimal temperature for myocardial
protection during intermittent cardioplegia
New Research and the ideal length of the intervals between
Current interest in the microcirculation infusions must be defined. Other parameters
and the apparent major role of the endothe- which might influence outcome are the vol-
lium in influencing cardiac tolerance to is- ume of infusate and composition. There is not
chemia/reperfusion will include examination consensus on the best temperature for central
of the role of temperature on endothelial func- nervous system protection which may prove
tion in relation to cardioplegia. It appears that difficult to determine because of the many
4˚C blood cardioplegia or crystalloid car- variables and the problems inherent in the
dioplegia with albumin is not associated with conduct of randomized clinical trials.
endothelial injury37 whereas crystalloid car-
dioplegia without albumin results in an im- Anticipated Developments
mediate injury which was reversible in one The authors believe that tepid cardiople-
study.38 gia with a comparable systemic temperature
Normothermic myocardial ischemia (45 will prove advantageous with regard to myo-
minutes) did not result in reduced vascular cardial protection and rapidity of cardiac and
responses to acetylcholine nor did the addi- systemic metabolic recovery from cardiople-
tion of one hour of intermittent 4˚C antegrade gia and mild hypothermia. Intermittent rather
blood cardioplegia, but if either event was than continuous cardioplegia facilitates con-
followed by reperfusion then vascular relax- duct of the operation (better visualization).
ation was impaired.39 Addition of nitric oxide Determining the optimal cardiac and systemic
donor agent (SPM-5185) in low (1 ∝mol/L) temperature for stroke prevention will be dif-
or high (10 ∝mol/L) dose to the blood car- ficult because of the many variables and the
dioplegia after 30 minutes of normothermic great numerical size needed for a randomized
myocardial ischemia followed by 60 minutes study to provide statistically useful data. It
of 4˚C intermittent cardioplegic infusion seems clear that tepid conduct of the operation
Cold/Tepid Cardioplegia 163

provides some margin of safety should systemic 14. Hayashida N, Weisel RD, Shirai T et al.
circulation be suddenly and unavoidably Tepid antegrade and retrograde cardioplegia.
Ann Thorac Surg 1995; 59:723-729.
interrupted (pump failure) and that the need 15. McMurchie EJ, Raison JK, Cairncross KD.
for hyperthermic perfusion may be less than Temperature-induced phase changes in mem-
that required for hypothermic or normother- branes of heart: A contrast between the
mic conduct of the operation. thermo response poikilotherms and homoth-
ermous. Comp Biochem Physiol (B) 1973;
44:1017-1026.
References 16. Fuhrman GJ, Fuhrman FA. Utilization of
1. Bigelow RWG, Lindsey WK, Greenwood glucose by the hypothermic rat. Am J Physiol
WF. Hypothermia: Its possible role in car- 1963; 295:181-183.
diac surgery. Ann Surg 1950; 132:849-856. 17. Teoh KH, Mickle DAJ, Weisel RD et al.
2. Barner HB. Historical aspects and current Decreased postoperative myocardial fatty acid
review of myocardial protection. in Salerno oxidation. J Surg Res 1988; 44:36-44.
TA, ed. Warm Heart Surgery, London: 18. Lyons JN, Raison JK. A temperature-induced
Arnold, 1995:1-15. transition in myocardial oxidation: Contrast
3. Buckberg GD, Brazier JR, Nelson RL et al. between cold and warm blooded animals.
Studies of the effects of hypothermia on re- Comp Biochem Physiol 1970; 37:405-411.
gional myocardial blood flow and metabolism 19. Vary TC, Angelakos ET, Schaffer SW. Rela-
during cardiopulmonary bypass. J Thorac tionship between adenine nucleotide metabo-
Cardiovasc Surg 1977; 73:87-94. lism and irreversible ischemic tissue damage
4. Iles RW, Silverman NA, Krukenkamp IB et in isolated perfused rat heart. Circ Res 1979;
al. Role of the red cell in oxygenated car- 45:218-224
dioplegia. FASEB J 1988; 2:A921-927. 20. Martin DR, Scott DF, Downes GL et al.
5. Shargge BW, Digerness SB, Blackstone EH. Primary cause of unsuccessful liver and heart
Complete recovery of myocardial function preservation: Cold sensitivity of the Atpase
following cold exposure. Circulation 1978; system. Ann Surg 1972; 175:111-117.
56(suppl II); 2:97-103. 21. Leaf A. Cell swelling: A factor in ischemic
6. Hearse DJ, Stewart DA, Chain CEB. Recov- tissue injury. Circulation 1973; 48:455-458.
ery from cardiac bypass and elective cardiac 22. Macknight AD, Leaf A. Regulation of cellu-
arrest. Circ Res 1974; 35:448-456. lar volume. Physiol Rev 1977; 57:510-573.
7. Hearse DJ, Stewart DA, Braimbridge MV. 23. Hess ML, Warner MF, Robbins AD et al.
Cellular protection during myocardial is- Postischemic deterioration of sarcoplasmic
chemia: The development and utilization of reticulum: Warm versus cold blood cardiople-
a procedure for the induction of revers- gia. Ann Thorac Surg 1994; 58:953-60.
ible ischemic arrest. Circulation 1976; 54: 24. See YP, Weisel RD, Mickle DAJ et al. Pro-
193-202. longed hypothermic cardiac storage for trans-
8. Tyers GFO, Williams EH, Hughes HC et al. plantation: The effects on myocardial metabo-
Effects of perfusion temperature on myocar- lism and mitachondrial function. J Thorac
dial protection from ischemia. J Thorac Cardiovasc Surg 1992; 104:817-824.
Cardiovasc Surg 1977; 73:766-771. 25. Rahn H, Reeves RB, Howell BJ. Hydrogen
9. Flaherty JT, Schaff HV, Goldman RA et al. ion regulation, temperature and evolution.
Metabolic and functional effects of progres- Am Rev Respir Dis 1975; 112:165-172.
sive degrees of hypothermia during global is- 26. Liu XL, Engelman RM, Wei Z et al. Postis-
chemia. Am J Physiol 1979; 236:H839- chemic deterioration of sarcoplasmic reticu-
H845. lum: Warm versus cold blood cardioplegia.
10. Lichtenstein SV, Ashe KA, El Dalati H et al. Ann Thorac Surg 1993; 56:1154-1159.
Warm heart surgery. J Thorac Cardiovasc 27. Buffkin BL, Mellitt RJ, Gott JP et al. Aero-
Surg 1991; 101:269-274. bic blood cardioplegia for revascularization of
11. Lichtenstein SV, Abel JG, Salerno TA. Warm acute infarct: Effects of delivery temperature.
heart surgery and results of operation for re- Ann Thorac Surg 1994; 58:953-960.
cent myocardial infarction. N Thorac Surg 28. Fiore AC, Swartz M, Nevett R et al. Tepid
1991; 52:455-460. blood cardioplegia for elective myocardial re-
12. Lichtenstein SV, El Dalati H, Panos A et al. vascularization. Ann Thorac Surg 1995;
Long cross-clamp times with warm heart sur- 65:1559-1560.
gery. Lancet 1989; 189; 1:144-144. 29. Engelman RM, Pleet AB, Rousou JA et al.
13. Hayashida N, Ikonomidis JS, Weisel RD et What is the best perfusion temperature for
al. The optimal cardioplegic temperature. N coronary revascularization? J Thorac
Thorac Surg 1994; 58:961-971. Cardiovasc Surg 1996; 112:1622-1633.
164 Ischemia-Reperfusion Injury in Cardiac Surgery

30. Martin TD, Craver JN, Gott JP et al. Pro- 37. Selke FW, Shafique T, Johnson RJ et al.
spective, randomized trial of retrograde warm Blood and albumen cardioplegia preserve
blood cardioplegia: Myocardial benefit and endothelian-dependent microvascular re-
neurologic threat. Ann Thorac Surg 1994; sponses. Ann Thorac Surg 1993; 55:977-985.
57:298-304. 38. Nilsson FN, Miller VN, Van Houtte PM et
31. Mora CT, Henson MB, Weintraub WS et al. Methods of cardiac preservation alter the
al. The effect of temperature management function of the endothelium in porcine coro-
during cardiopulmonary bypass on neurologic nary arteries. J Thorac Cardiovasc Surg
and neuropsychologic outcomes in patients 1991;102:923-930.
undergoing coronary revascularization. J 39. Nakanishi K, Zhao ZQ, Vinten-Johansen J
Thorac Cardiovasc Surg 1996; 112:514-522. et al. Coronary artery endothelial dysfunction
32. Regraqui I, Birdi I, Izzat MB et al. The ef- after global ischemia, blood cardioplegia and
fects of cardiopulmonary bypass temperature reperfusion. Ann Thorac Surg 1994; 58:
on neuropsychological outcome after coronary 191-199.
artery surgery. A prospective randomized trial. 40. Nakanishi K, Zhao ZQ, Vinten-Johansen J
J Thorac Cardiovasc Surg (in press) et al. Blood cardioplegia enhanced with ni-
33. McLean RF, Wong BI, Naylor CD et al. tric oxide donor SPM-5185 counteracts post-
Cardiopulmonary bypass temperature and ischemic endothelial and ventricular dysfunc-
central nervous system dysfunction. Circula- tion. J Thorac Cardiovasc Surg 1995; 109:
tion 1994; 90:250-255. 1146-1154.
34. Warm heart investigators. Randomized trial 41. Sato H, Zhao ZQ, Jordan JE et al. Basal ni-
of normothermic versus hypothermic coronary tric oxide depresses endogenous cardio pro-
bypass surgery. Lancet 1994; 343:559-563. tection during reperfusion by inhibition of
35. Singh AK, Bert AA, Feng WC. Neurologic neutrophil-mediated damage after surgical
complications during myocardial revascular- revascularization. J Thorac Cardiovasc Surg
ization using warm-body, cold-heart surgery. 1997; 113:399-409.
Eur J Cardiothorac Surg 1994; 8:259-264. 42. Lin PJ, Chang CH, Hsiao CW et al. Con-
36. Plourde G, Leduc AS, Morin JE et al. Tem- tinuous antegrade warm blood cardioplegia
perature during cardiopulmonary bypass for attenuates augmented coronary endothelium-
coronary artery operations does not influence dependent contraction after cardiac global is-
postoperative cognitive function: A prospec- chemia and reperfusion. J Thorac Cardiovasc
tive randomized trial. J Thorac Cardiovasc Surg 1997; 114:100-108.
Surg 1997; 114:123-128
CHAPTER 14

Surgical Techniques
for Warm Blood Cardioplegia
Syed T. Raza and Tomas A. Salerno

H
ypothermic techniques of myocardial Conceptual Framework
preservation (crystalloid or blood As mentioned earlier, the conceptual frame-
cardioplegia) have been utilized in work of warm blood cardioplegia (WBC) is
recent years and represent an important prevention of ischemia during the period of
development in cardiac surgery. It can be stated aortic cross-clamping. Utilized by Gott et al1,2
that results of cardiac surgery are largely due who used continuous retrograde warm blood
to precision in surgical techniques and myo- perfusion for aortic valve surgery, the method
cardial protective strategies. Among strategies was further developed by Bomfim et al3 who
available to cardiac surgeons are the use of administered continuous cold blood cardiople-
either crystalloid (delivered cold) or blood (de- gia and by our group in Toronto who redis-
livered either warm or cold) cardioplegic covered the technique, modified it, and applied
solutions. The mode of delivery, i.e., antegrade, it clinically.4-7 Over the years, there have been
retrograde, alternately antegrade/retrograde, or important developments regarding the tech-
simultaneously delivered antegrade/retrograde nique, some of which are related to the car-
are all important routes of administration dioplegia composition, route of administra-
available in any cardiac procedure. The use of tion and changes in the temperature of the
intermittent delivery of cardioplegia implies cardioplegia. More importantly, for precision
that the heart is subjected to a period of is- during coronary artery surgery, the intermit-
chemia which, if for short periods of time (< tent method of WBC was reported8,9 and its
10 minutes each), may be inconsequential. efficacy and principles were reported by
However, longer periods of aortic clamping Deslauriers and her group at the National
and/or the presence of ventricular hypertro- Research Council10 Canada. What follows is
phy or acute ischemic syndromes, may lead to a current view of the warm cardioplegic
an adverse outcome, should the intermittent method, with emphasis on some of the most
method be utilized. Warm blood cardioplegia important aspects of the technique.
was developed in search of prevention of is-
chemia during the period of aortic clamping.
The implication of the original concept was Techniques
that the heart would be continuously perfused
during the period of aortic clamping. Antegrade Warm Blood
Cardioplegia (AWBC)
Although there are concerns in the litera-
ture regarding the flow distribution of car-
dioplegia distal to coronary stenoses, AWBC

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


166 Ischemia-Reperfusion Injury in Cardiac Surgery

is one of the simplest methods of myocardial Retrograde Continuous Warm


protection. It can be utilized in all forms of Blood Cardioplegia (RCWBC)
cardiac procedures, including coronary and
As mentioned, this technique was rediscov-
valvular surgery and other procedures. The
ered, and it was first utilized by Gott et al.1,2
current technique is usually via the “mini-
The development of the retrograde catheter
plegia” route, i.e., from the oxygenator blood
by Buckberg et al, in which insertion is via
is extracted and is infused into the aorta with
the transatrial approach, simplified the
the aid of a roller pump; the “miniplegia” is
method. After aortic clamping, an infusion of
simply KCl and Mg, which are infused directly
antegrade cardioplegia arrests the heart alter-
into the cardioplegic line.11 The advantages of
natively. Aortic clamping is followed by retro-
this method is that hemodilution is avoided
grade delivery of cardioplegia without
and higher hemoglobin and oxygen carrying
antegrade infusion. The infusion pressure,
capacity are achieved. When crystalloid solu-
measured at the distal port of the coronary
tions are used and mixed with blood in a 4:1,
sinus catheter, should be < 50 mmHg,
8:1 or other dilutions, severe hemodilution
which usually corresponds to flows of
may occur if the period of aortic clamping is
250-300 ml/min of cardioplegia. Blood in the
prolonged. Left ventricular distention may be
operative field is minimized, i.e., when the
avoided either by intermittently venting the
coronary artery is opened, venous blood exits
aorta during very short periods of cardioplegic
from the arteriotomy. This is easily controlled
interruptions, or by venting the left heart.
by the blower, flowrester or suture devices.
Patients with moderate or severe aortic insuf-
Ventricular distention does not occur and the
ficiency pose problems during antegrade
presence of aortic insufficiency does not pre-
cardioplegic delivery as most of the solution
clude its use. There have been excellent stud-
goes into the left ventricle via an incompetent
ies in the literature dealing with flow distribu-
aortic valve, leading to inadequate myocardial
tion during RCWBC, including elegant MRI
protection. Furthermore, in coronary artery
studies in pigs by Deslaurier’s group at the
surgery, the presence of blood in the coronary
National Research Council.12 In these studies
artery makes visualization at times difficult
the RV region is not adequately perfused dur-
unless the artery is snared, shunted, or a blower
ing RCWBC, although there have been no
device is used. For these reasons, intermittent
human studies showing that the right ventricle
warm blood cardioplegia (IWBC) was devel-
is poorly protected.
oped and was shown experimentally8,10 to be
safe. Periods of less than or equal to 10 min-
utes are used, with reinfusion of cardioplegia Alternate Retrograde/Antegrade
at these intervals. This replenishes the energy Cardioplegia
stores and is well tolerated by the normal heart.
Elegant studies by Buckberg et al13 suggest
Recently, Calafiore et al utilized this method
that antegrade versus retrograde cardioplegia
in hypertrophied hearts (aortic stenosis) with
perfuses different vascular beds. It is therefore
excellent results. Due to other technical diffi-
reasoned that for “ideal” cardioplegic protec-
culties such as LV distention during construc-
tion, the cardioplegia must be administered
tion of a circumflex anastomosis, the concern
antegradely followed by the retrograde route.
of maldistribution of flow and other technical
This method was defined for and used by uti-
problems during surgery, retrograde continu-
lizing cold intermittent blood cardioplegia.
ous warm blood cardioplegia was rediscovered
and was applied clinically.
Simultaneous Antegrade/
Retrograde Cardioplegia
During a complex cardiac procedure, the
aortic clamp was released while retrograde car-
Surgical Techniques for Warm Blood Cardioplegia 167

dioplegia was being administered. This lead cerns regarding right ventricular protection in
to the clinical observation that the pressure in hypertrophied hearts and in patients with pul-
the distal port of the coronary sinus catheter monary hypertension, right ventricular failure
did not change. We therefore began to deliver has not been observed clinically.
continuously simultaneous antegrade (into the
grafts or coronary ostia) and retrograde (into Normothermic Perfusion
the coronary sinus) cardioplegia during all During the cardiac procedure, the systemic
types of cardiac surgery. Experimental work14 temperature is maintained at normothermia
and clinical data confirmed the safety of the (37˚C). This usually requires warming the
technique, which, in our view, represents one patient as soon as cardiopulmonary bypass is
of the most effective method of myocardial established. Although there were initial con-
protection, addressing all concerns of mal- cerns regarding the adverse effects of normo-
distribution of flow during cardioplegic thermia on the central nervous system, recent
administration. Again, in all these methods, studies have failed to confirm this observation
“miniplegia” is used without any additives such and normothermia appears to be safe. 15
as Asparate/Glutamate, delivered at 37˚C and Peripheral dilatation usually occurs and
without interruption. Experimental and clini- requires administration of alpha constrictors
cal work14 attests to the safety and efficiency during and sometimes postoperatively. Some
of this modality of myocardial protection. believe that the beneficial effects of warm heart
surgery are due to normothermic systemic per-
Warm Cardioplegia Techniques fusion and its sequellae, i.e., peripheral dilata-
for Valvular Surgery tion. Shivering is avoided postsurgery allow-
Currently we have modified the cardiople- ing for more stability hemodynamically and
gia technique for valvular surgery with or with- for early extubation of the patient.
out combined coronary surgery. We simply
clamp the aorta and deliver blood under pres-
sure (line resistance 130 mmHg and distal Intermittent Techniques
coronary sinus port pressure < 50 mmHg) of Warm Blood Cardioplegia
which usually equates to mean flows of These have been championed by Calafiore
300 ml/min. The heart beats throughout the et al.8 There is no doubt that for coronary
operation and, if needed, boluses of KCl are artery surgery this technique has many advan-
infused for brief periods to ensure continued tages, such as simplicity, decreased cost, and
cardiac arrest. During aortic valvular surgery, excellent early results. Although warm blood
after aortic clamping and delivery of blood into cardioplegia (WBC) was not designed to be
the coronary sinus, the aortotomy is performed delivered intermittently, i.e., the principle is
and a PolystanTM cannula is placed in the left prevention of ischemia and not to subject the
and right coronary ostia and simultaneous heart to normothermic ischemia, this method
coronary sinus and ostia blood flow are estab- is being extensively used in coronary artery
lished. If the right ostium is small, only the surgery in Europe. Its scientific basis has been
coronary sinus and the left ostium are perfused established16 but surgeons are still skeptical
simultaneously. As each coronary artery bypass about the safety of warm ischemia in high risk
is performed, blood flow is delivered into patients and acute syndromes.
the coronary sinus and into each vein as the
distal anastomosis is completed. The whole
procedure is performed during a single aortic Myocardial Resuscitation
clamping period. For mitral valve replacement The goal of the WBC method was to allow
or repair only retrograde perfusion of blood for maintenance of aerobic metabolism and,
or “miniplegia” is infused while the procedure hopefully resuscitation of the damaged heart
is performed on a clamped aorta. Despite con- by continuous perfusion of WBC, based on
168 Ischemia-Reperfusion Injury in Cardiac Surgery

the principle that only WBC resuscitates the of warm versus cold cardioplegia techniques
heart. Unfortunately, this has not been con- to actual resuscitation of the myocardium with
firmed and studies on Asparate/Glutamate in WBC particularly in shock hearts and in hy-
the cardioplegia failed to confirm the incor- pertrophic hearts (patients undergoing valve
poration of these additives in the TCA cycle surgery).
and failure to prove a beneficial effect. This Use of retrograde warm blood perfusion
has lead us to abandon the use of these addi- of the myocardium was first reported by Gott2
tives in a clinical solution. The idea of resusci- et al in 1957. This was later replaced by direct
tation of the injured heart to our knowledge coronary ostial perfusion, until crystalloid
remains elusive. hyperkalemic arrest and the hypothermic
blood cardioplegic techniques become widely
Monitoring of Cardioplegia used in the 1970s. Warm blood cardioplegia
Delivery was rediscovered by the group in Toronto and
reported in 1989.16
Nowhere is monitoring more important
It has been shown in a prospective, ran-
than during WBC techniques. As mentioned,
domized study of 20 patients undergoing car-
the principle of the method is continuous
diac surgery that oxidative stress is minimized
delivery of WBC into the coronary sinus. Fail-
with warm blood cardioplegia, 17 but this
ure to monitor flow and/or distal coronary
reduction in oxidative stress did not corre-
sinus catheter port pressure, may lead to
late with postoperative hemodynamic
excessive pressure thereby the risk of rupture
measurements.
of the coronary sinus or alternatively, dislodg-
In another study, the beta adrenergic
ment of the coronary sinus catheter into the
receptor function of the heart was compared
right atrium without warning. This may lead
in a group of 20 patients, each undergoing
to a false sense of security when in actual fact
CABG with either cold intermittent or inter-
flow of WBC has been interrupted and the
mittent warm cardioplegia. 18 This study
heart is subjected to prolonged periods of nor-
showed improved preservation of the auto-
mothermic ischemia. Therefore, WBC is more
nomic sympathetic function of the heart with
cumbersome to the perfusionist, who must be
warm cardioplegia.
constantly attendant during the period of aor-
Myocardial ultrastructure was compared in
tic clamping, while the surgeon is busy with
the two cardioplegia groups19 and was noted
the technical details of the procedure. This is
that only mild to moderate and reversible
in contrast with intermittent techniques of car-
ultrastructure changes occur during continu-
dioplegia delivery, in which case the perfusion-
ous retrograde warm blood cardioplegia
ist only monitors the time period of the
(WBC) and cold blood cardioplegia (CBC)
cardioplegic interruption. One of the most
for CABG operation. In another study of
important pitfalls of the RCWBC method is
ultrastructure, WBC was shown to be supe-
coronary sinus injury and catheter displace-
rior to CBC techniques.20
ment leading to normothermic ischemia.
Numerous clinical studies, both retrospec-
tive8,21-30 and prospective randomized31-37 have
Advantages and Disadvantages reported that WBC is at least as efficacious or
of Warm Blood Cardioplegia better than CBC techniques for CABG. Of
note are the two large prospective, random-
Theoretically the single greatest advantage
ized clinical trials comparing WBC versus
of WBC is to reduce or prevent myocardial
CBC in patients undergoing CABG.32,37 The
ischemia during aortic cross-clamping for per-
first of these trials32 was conducted at Emory
formance of cardiac operations. A review of
University in Atlanta and randomized 1001
the clinical studies reported over the last 7 years
patients undergoing CABG surgery to either
(since the introduction of WBC in 1989) show
continuous WBC with systemic normother-
that opinions vary from no minimal advantage
mia or intermittent cold oxygenated, crystal-
Surgical Techniques for Warm Blood Cardioplegia 169

loid cardioplegia and moderate systemic hy- Visual Enhancement


pothermia. The preoperative variables were During WBC
similar in the two groups. The postoperative
Three techniques and innovations have
results showed that the mortality (warm 1.0%
been used to improve conditions for coronary
and cold 1.6%), Q wave infarction (warm
anastomoses during CABG surgery with
1.4%, cold 0.8%) and need for intra-aortic
WBC. These include use of Flowresters and
balloon pump (IABP) (warm 1.4% and cold
use of blowers which blow saline with a mist
2.0%) were similar in the 2 groups. Total neu-
of O2 to blow away blood from coronary ar-
rologic events (warm 4.5% and cold 1.4%;
teries. The third technique is to use intermit-
p<0.005) and perioperative strokes (warm
tent WBC, where cardioplegia delivery is
3.1% and cold 1.0%; p<0.02) were signifi-
interrupted for up to 10 minutes while
cantly higher in the warm group. In the
performing the anastomoses. In a series of very
Toronto prospective trial37 1732 patients
elegant articles, Calafiore et al from Chieti,
undergoing CABG were randomized to warm
Italy have demonstrated that safety and effi-
and cold blood cardioplegia for myocardial
cacy of using intermittent WBC9,28,38 in vari-
preservation. The baseline demographics were
ous cardiac operations. In one study8 compar-
similar. The all-cause mortality was 2.5% in
ing intermittent antegrade WBC (IAWBC)
the cold group and 1.4% in the warm group
used in the first 250 patients undergoing
(p < 0.12), as was the incidence of nonfatal
CABG with this technique to the last 250
Q-wave infarction (warm 10.1%, cold
patients who had CABG using intermittent
11.1%). In this study, however, there were no
antegrade CBC, Calafiore et al showed that
differences in the rate of stroke, reoperation
the mortality was lower in the warm group
for bleeding or tamponade, or sternal compli-
(0.8% versus 3.6%; p < .05). Further, in-hos-
cations. They concluded that warm heart sur-
pital mortality for high risk patients was 0/53
gery is a safe and effective alternative to con-
in the warm group compared to 2/28 (p <
ventional hypothermic techniques for patients
0.05) in the cold group. Circulatory assistance
undergoing CABG surgery.
was needed in 0 versus 4 patients in Group A
and B. The rates of postoperative myocardial
Technical Considerations infarction and strokes were similar in the two
in WBC groups.
Obviously the most important consider- To determine if intermittence in WBC was
ation in any myocardial preservation technique harmful, the Toronto Warm Heart Investiga-
is the ability to perform a perfect anastomosis tors studied 720 CABG patients with particu-
while performing CABG, which requires a still lar attention to the longest single time off
heart and clear blood vessels. To obtain a still (LTOC) and the time off cardioplegia as per-
heart in WBC, larger amounts of cardioplegic centage of the cross-clamp time (PTOC).39
solution is required to be infused antegrade or Longer LTOC was found to be harmful
retrograde continuously. This resulted in large whereas longer PTOC was protective. It was
volume of cardioplegia solution to be deliv- further shown that repeated interruptions of
ered resulting in hypervolemia as well as warm blood cardioplegia are unlikely to lead
hyperkalemia. These problems were largely to adverse clinical results as single interrup-
resolved with the introduction of “miniplegia” tions are greater than or equal to 13 minutes.
techniques.11 In this method, blood from the
arterial pump is infused into the aortic roof or
the coronary sinus, and small amounts of crys- Inadequate RV Protection
talloid solution containing K+ and Mg++ are with WBC
added to maintain a still heart. That the right ventricle is inadequately
protected in WBC has been a concern, par-
ticularly when the delivery of WBC is through
170 Ischemia-Reperfusion Injury in Cardiac Surgery

the retrograde approach via the coronary sinus. Postoperative Neurological


However, this has not been found to be a clini- Deficits and Stroke Rates
cal problem. Two studies need be cited in this
regard. The first by Menasche et al21 where 75 after WBC and Normothermic
patients were carefully studied to determine Cardiopulmonary Bypass
the RV function after cardiac operation using Concern regarding increased incidence of
RWBC. In the first part of this three-part neurological deficits and stroke rates follow-
study, they showed that the anaerobic metabo- ing CABG surgery with normothermic car-
lism was similar in RV and LV. In a second diopulmonary bypass and WBC were first
part, the RV stroke work index was compared raised by the Emory University group. In their
in 15 patients where RWBC was used to case randomized trial32 of warm versus cold car-
match 15 patients where cold antegrade crys- dioplegia which was summarized earlier, they
talloid cardioplegia was used. Again the results reported a significantly higher incidence of
were similar in the two groups. In part three, total neurologic events (4.5% versus 1.4%)
function was determined in 30 patients and perioperative strokes (3.1% versus 1.0%)
undergoing mitral valve procedures where RV in the warm cardioplegia group. Since then,
was at higher risk of ischemia (15 each in warm many additional studies have been reported
and cold groups). Again there were no differ- looking at this problem.42-45 In a review article,
ences in postoperative RV function. Christakis et al46 analyzed all available con-
The second study was reported by trolled studies of warm versus cold and
Christakis et al from Toronto.40 They ran- antegrade versus retrograde cardioplegia, to
domized 52 patients to either intermittent assess the incidence of perioperative stroke and
warm or cold blood cardioplegia during adverse neuropsychological outcomes. Nine
CABG surgery. The RV ejection fraction was randomized trials and two studies with im-
greater at six hours (warm 0.46 ± 0.06; cold mediate historical controls were included. The
0.37 ± 0.08; p < 0.05) and at 8 hours (warm pooled event rates for perioperative stroke were
0.43 ± 0.08; cold 0.37 ± 0.08; p < 0.05) in the 1.5% for warm antegrade, 3.14% for warm
warm group. The RV end diastolic volume retrograde, 1.7% for cold antegrade and
index was less in the warm group at eight hours 0-1.2% for cold retrograde. In further analy-
postoperatively. sis, they determined that the differences found
Thus RV dysfunction after WBC is not a are unlikely to be due to temperature, but may
clinical problem. be related to antegrade versus retrograde coro-
nary perfusion.
Coronary Sinus Injuries
Coronary sinus injuries during placement Tepid Cardioplegia
of retrograde coronary sinus catheter have been Warm cardioplegia implies perfusate tem-
reported by Panos et al.41 These are of course perature of 37˚C. Some groups have taken the
not related to warm or cold cardioplegia, but middle of the road approach in order to get
to the route of delivery. One must be cautious the maximum benefits of both warm and cold
in placing of these catheters. The catheters blood cardioplegia, while simultaneously
should never be pushed forcefully. The guide reducing the disadvantages of the two
should be pulled back about a centimeter extremes. They are proponents of the tepid
before advancing the catheter in the coronary (or lukewarm) cardioplegia.45,47,48 Good results
sinus, so as to have the soft tip of the catheter are reported with lukewarm (32˚C) tempera-
on the leading edge as it is advanced. After an ture of cardioplegia.
initial learning curve, this maneuver becomes
easier and injuries to the coronary sinus very
rare.
Surgical Techniques for Warm Blood Cardioplegia 171

Cold Agglutinins and Warm References


1. Gott VL, Gonalez JL, Paneth M et al. Car-
Blood Cardioplegia diac retroperfusion with induced asystole for
The technique of normothermic cardio- open heart surgery upon the aortic valve or
pulmonary bypass with warm blood cardiople- coronary arteries. Proc Soc Exp Biol Med
gia is an advantageous technique for perform- 1957; 94(4):689-692.
ing cardiac surgical procedures in patients with 2. Gott VL, Gonzalez JL, Zuhdi MN et al. Ret-
cold autoimmune disease. Several case reports rograde perfusion of the coronary sinus for
have documented this advantage49-51 direct vision aortic surgery. Surg Gynecol
Obstet 1957; 104:319-328.
3. Bomfim V, Kaijser L, Bendz R et al. Myo-
cardial protection during aortic valve replace-
Future Perspectives ment. Cardiac metabolism and enzyme release
As with any technique of myocardial pro- following continuous blood cardioplegia. Scan
tection, WBC received great attention as we J Thorac Cardiovasc Surg 1981; 15:141-147.
searched for the “ideal solution” to the 4. Salerno TA. Continuous blood cardioplegia:
Option for the future or return to the past?
cardioplegic method. The initial crystalloid J Mol Cell Cardiol 1990; 22(Suppl V):S49.
solutions added to blood in different mixtures 5. Panos A, Ashe K, El-Dalati H et al. Clinical
(4:1 or 8:1) caused many problems such as comparison of continuous warm (37˚C) ver-
hemodilution, hyperglycemia, hyperkalemia sus continuous cold (10˚C) blood cardiople-
and others. “Miniplegia” solved these issues gia in CABG surgery. Clin Invest Med 1989;
12(5 suppl):C55.
and, more recently, perfusion of the coronary 6. Panos A, Abel J, Slutsky AS et al. Warm aero-
sinus with blood seems to us to be even more bic arrest: A new approach to myocardial
advantageous. We are now performing com- protection. J Mol Cell Cardiol 1990; 22(suppl
plete revascularization on a beating heart with- V):S31.
7. Panos A, Kingsley SJ, Hong AP et al. Con-
out cardiopulmonary bypass. The role of car- tinuous warm blood cardioplegia. Surg Fo-
diopulmonary bypass and cardioplegia in rum 1990; 41:233-235.
coronary artery surgery is being reassessed by 8. Calafiore AM, Teodori G, Mezzetti A et al.
our group, who has spent the last 20 years Intermittent antegrade warm blood cardiople-
working on myocardial protection. Some of gia. Ann Thorac Surg 1995; 59:398-402.
9. Mezzetti A, Calafiore AM, Lapenna D et al.
the techniques and developments in cardiople- Intermittent antegrade warm cardioplegia re-
gia are already of historical value. All of them duces oxidative stress and improves metabo-
have made cardiac surgery safer but, with ad- lism in the ischemic-reperfused human myo-
vancement of technology, are being replaced cardium. J Thorac Cardiovasc Surg 1995;
109:787-795.
by innovative techniques aimed at a simpler 10. Tian GH, Xiang B Butler KW et al. Effects
and safer operations. There will always be the of intermittent warm blood cardioplegia on
need for cardioplegia in cardiac surgery for as myocardial high energy phosphates, intracel-
long as we have to perform intracardiac repair lular pH and contractile function—a 31P
and/or complex ascending aorta and arch sur- NMR study in isolated rat and pig hearts. J
Thorac Cardiovasc Surg (in press 1995; 109).
gery. Simultaneous antegrade/retrograde infu- 11. LeHouerou D, Pargaonkar S, Lessana A. Car-
sion of warm blood (or WBC) comes close to dioplegia delivery systems for warm blood
being the “ideal” method of myocardial pro- cardioplegia. Warm Heart Surgery 1995;
tection. Surgeons can now utilize a variety of 46-54.
12. Ye J, Sun J, Shen J et al. Does retrograde
myocardial protective strategies which warm blood cardioplegia provide equal pro-
compliment each other during cardiac surgery. tection to both ventricles? A magnetic reso-
There should be no adversarial position of one nance spectroscopy study in pigs. Circ (Suppl
method against the other. Each technique is II) 1997; 96(9):II210-II215.
tailored to each patient’s needs. 13. Partington MT, Acar C, Buckberg GD et al.
Advantages of antegrade/retrograde cardiople-
gia to optimize distribution in jeopardized
myocardium. J Thorac Cardiovasc Surg 1989;
97:613-622.
172 Ischemia-Reperfusion Injury in Cardiac Surgery

14. Buckberg GD, Salerno TA. Simultaneous 28. Calafiore AM, Teodori G, Di Giammarco G
antegrade/retrograde cardioplegia: Experimen- et al. Intermittent antegrade cardioplegia:
tal/clinical study of a new cardioprotective warm blood vs cold crystalloid. A clinical
technique. Warm Heart Surgery 1995; 90-98. study. J Cardiovasc Surg 1994 Dec; 35(6
15. Pleet AB, Engelman RM. Cerebral protection Suppl 1):179-184.
during warm heart surgery. Warm Heart Sur- 29. Mikaeloff P, Jegaden O, Montagna P et al.
gery 1995; 178-188. Is continuous warm retrograde blood car-
16. Lichtenstein SV, El Dalati H, Panos A et al. dioplegia completely safe for coronary artery
Long cross-clamp times with warm heart sur- surgery? Europ J Cardiothorac Surg 1994;
gery. Lancet 1989; 1:1443-1444. 8(11):569-574.
17. Biagioli B, Borrelli E, Maccherini M et al. 30. Ali IM, Kinley CE. The safety of intermit-
Reduction of oxidative stress does not affect tent warm blood cardioplegia. Europ J
recovery of myocardial function: Warm con- Cardiothorac Surg 1994; 8(10):554-556.
tinuous versus cold intermittent blood car- 31. Pelletier LC, Carrier M, Leclerc Y et al. In-
dioplegia. Heart 1997; 77(5):465-473. termittent antegrade warm versus cold blood
18. Chello M, Mastroroberto P, De Amicis V et cardioplegia: a prospective, randomized study.
al. Intermittent warm blood cardioplegia pre- Ann Thorac Surg 1994 Jul; 58(1):41-48.
serves myocardial beta-adrenergic receptor 32. Martin TD, Craver JM, Gott JP et al. Pro-
function. Ann Thorac Surg 1997 Mar; spective, randomized trial of retrograde warm
63(3):683-688. blood cardioplegia: Myocardial benefit and
19. Rainio P, Sormunen R, Lepojarvi M et al. neurologic threat. Ann Thorac Surg 1994
Ultrastructural changes during continuous Feb; 57(2):298-302.
retrograde warm and mild hypothermic blood 33. Gerola LR, Oliveira SA, Moreira LF et al.
cardioplegia for coronary bypass operations. Blood cardioplegia with warm reperfusion
J Thorac Cardiovasc Surg 1995 Jul; 110(1): versus intermittent aortic crossclamping in
81-88. myocardial revascularization. Randomized
20. Tasdemir O, Katircioglu SF, Kucukaksu DS controlled trial. J Thorac Cardiovasc Surg
et al. Warm blood cardioplegia: Ultrastruc- 1993 Sep; 106(3):491-196.
tural and hemodynamic study. Ann Thorac 34. Lajos TZ, Espersen CC, Lajos PS et al. Com-
Surg 1993 Aug \; 56(2):305-311. parison of cold versus warm cardioplegia.
21. Menasche P, Fleury JP, Droc L et al. Meta- Crystalloid antegrade or retrograde blood?
bolic and functional evidence that retrograde Circ 1993 Nov; 88(5 Pt 2):II344-349.
warm blood cardioplegia does not injure the 35. Landymore R, Murphy JT, Hall R et al. Ran-
right ventricle in human beings. Circ 1994 domized trial comparing intermittent ante-
Nov; 90(5 Pt 2):II310-II315. grade warm blood cardioplegia with multidose
22. Hanafy HM, Allen BS, Winkelmann JW et cold blood cardioplegia for coronary artery
al. Warm blood cardioplegic induction: an bypass. Europ J Cardiothorac Surg 1996;
underused modality. Ann Thorac Surg 1994 10(3):179-184.
Dec; 58(6):1589-1594. 36. Rashid A, Fabri BM, Jackson M et al. A pro-
23. Vaughn CC, Opie JC, Florendo FT et al. spective randomized study of continuous
Warm blood cardioplegia. Ann Thorac Surg warm versus intermittent cold blood car-
1993 May; 55(5):1227-1232. dioplegia for coronary artery surgery: Prelimi-
24. Gundry SR, Wang N, Bannon D et al. Ret- nary report. Europ J Cardiothorac Surg 1994;
rograde continuous warm blood cardioplegia: 8(5):265-269.
Maintenance of myocardial homeostasis in 37. Naylor CD, Lichtenstein SV, Fremes SE et
humans. Ann Thorac Surg 1993 Feb 55(2): al. Randomized trial of normothermic versus
358-361. hypothermic coronary bypass surgery. The
25. Bel A, Aznag H, Faris B et al. Warm blood warm heart investigators. Lancet 1994 Mar;
cardioplegia in high risk patients. Europ J 343(8897):559-563.
Cardiothorac Surg 1997 Jun; 11(6):1118- 38. Calafiore AM, Teodori G, Bosco G et al.
1123. Intermittent antegrade warm blood cardiople-
26. Borman JB, Arbell D, Izhar U et al. Luke gia in aortic valve replacement. J Card Surg
warm blood cardioplegia for CAB surgery in 1996 Sep-Oct; 11(5):348-354.
patients with severely impaired LV function. 39. Lichtenstein SV, Naylor CD, Feindel CM et
J Cardiovasc Surg 1995 Dec; 36(6):545-550. al. Intermittent warm blood cardioplegia.
27. Tolis GA, Astras G, Sfyras N et al. Experi- Warm Heart Investigators. Circ 1995 Nov;
ence with warm blood cardioplegia in 480 92(9 Suppl):II341-346.
patients. Cardiovasc Surg 1995 Apr; 3(2):
175-180.
Surgical Techniques for Warm Blood Cardioplegia 173

40. Christakis GT, Buth KJ, Weisel RD et al. 46. Christakis GT, Abel JG, Lichtenstein SV.
Randomized study of right ventricular func- Neurological outcomes and cardiopulmonary
tion with intermittent warm or cold car- temperature: A clinical review. J Card Surg
dioplegia. Ann Thorac Surg 1996 Jan; 61(1): 1995 Jul; 10(4 Suppl):475-480.
128-134. 47. Hayashida N, Ikonomidis JS, Weisel RD et
41. Panos AL, Ali IS, Birnbaum PL et al. Coro- al. The optimal cardioplegic temperature. Ann
nary sinus injuries during retrograde continu- Thorac Surg 1994 Oct; 58(4):961-971.
ous normothermic blood cardioplegia. Ann 48. Hayashida N, Weisel RD, Shirai T et al.
Thorac Surg 1992; 54(6):1137-1138. Tepid antegrade and retrograde cardioplegia.
42. Craver JM, Bufkin BL, Weintraub WS et al. Ann Thorac Surg 1995 Mar; 59(3):723-729.
Neurologic events after coronary bypass graft- 49. Muehreke DD, Torchianna DF. Warm heart
ing: further observations with warm cardiople- surgery in patients with cold autoimmune
gia. Ann Thorac Surg 1995 Jun; 59(6): disorders. Ann Thorac Surg 1993 Feb;
1429-1433. 55(2):532-533.
43. Guyton RA, Mellitt RJ, Weintraub WS. A 50. Hearnsberger J, Ziomek S, Tobler G et al.
critical assessment of neurological risk dur- Management of cold agglutinemia with warm
ing warm heart surgery. J Card Surg 1995 heart surgical intervention: a case report. J
Jul; 10(4 Suppl):488-492. Thorac Cardiovasc Surg 1993 Oct; 106(4):
44. Guyton RA, Gott JP, Brown WM et al. Cold 756-757.
and warm myocardial protection techniques. 51. Donatelli F, Mariani MA, Triggiani M et al.
Advances in Card Surg 1996; 7:1-29. Warm heart surgery in cold haemagglutinin
45. Engelman RM, Pleet AB, Rousou JA et al. disease. Cardiovasc Surg 1995 Apr; 3(2):
What is the best perfusion temperature for 191-192.
coronary revascularization? J Thorac
Cardiovasc Surg 1996 Dec; 112(6):1622-
1632.
CHAPTER 15

Interrupting Warm Blood


Cardioplegia
Harold L. Lazar

W
arm blood cardioplegia has lower areas of necrosis than in the antegrade
emerged as an alternative method warm blood cardioplegia group, it did not
of myocardial protection. Initial achieve any superior protection to that
retrospective clinical studies using warm blood obtained with antegrade/retrograde cold blood
techniques showed good myocardial protec- cardioplegic techniques. During the period of
tion; however, prospective studies comparing cardioplegic arrest, both warm cardioplegic
warm blood cardioplegia with cold blood techniques had significantly lower tissue pH
cardioplegia were lacking. 1,2 Furthermore, values in both the area at risk and in unob-
the ability of warm blood cardioplegia to structed myocardium than the cold cardiople-
adequately protect acutely ischemic myocar- gia groups. Upon reperfusion, pH values
dium was unknown. This prompted us to returned to preischemic values in the unob-
undertake an experimental study to compare structed myocardium in all cardioplegic
the effectiveness of warm blood cardioplegia groups. In addition, wall motion scores were
versus cold blood cardioplegia in protecting not significantly different in the unobstructed
areas of ischemic myocardium.3 In 40 adult myocardial segments, suggesting that tempera-
pigs, the second and third diagonal coronary ture and the route of delivery are not as criti-
arteries were occluded with snares for 90 min- cal in myocardial tissue with normal coronary
utes, followed by 45 minutes of cardioplegic anatomy. In contrast, the method of cardio-
arrest and 180 minutes of reperfusion during plegic delivery was an important determinant
which time the coronary snares were released. in the degree of necrosis seen in the area at
During the period of cardioplegic arrest, risk. All retrograde cardioplegic techniques,
10 pigs received antegrade continuous regardless of temperature, had much lower
warm blood cardioplegic solution (37˚C) at areas of necrosis when compared with
100 ml/min; 10 animals received retrograde antegrade delivery. This suggested that it was
continuous warm blood cardioplegic solution the adequacy of distribution and not the tem-
at 100 ml/min; 10 received intermittent, perature of the cardioplegic solution that is
antegrade cold blood cardioplegic solution most critical in achieving optimal myocardial
(4˚C), and 10 received intermittent, antegrade/ protection.
retrograde cold blood cardioplegic solution. A potential disadvantage of warm blood
Our results showed that antegrade continu- cardioplegia techniques is that the continuous
ous warm blood cardioplegia resulted in the infusion of cardioplegia will obscure the
least optimal myocardial protection in the area operative field necessitating that the cardiople-
at risk. Although retrograde continuous warm gia be interrupted. However, the effects of
blood cardioplegia resulted in significantly interrupting warm blood cardioplegia during

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Interrupting Warm Blood Cardioplegia 175

the revascularization of acutely ischemic myo- prolonged regional ischemia.5 However, inter-
cardium were unknown. We, therefore, added rupting warm blood cardioplegia after global
another group to our previous study of 90 ischemic arrest differs from a beating heart sub-
minutes of coronary occlusion followed by 45 jected to repeated episodes of regional
minutes of cardioplegic arrest and 180 min- ischemia. Several important issues needed to
utes of reperfusion.4 In 10 pigs, the retrograde be resolved before the efficacy of interrupting
infusion of warm blood cardioplegia (37˚C at warm blood cardioplegia could be established.
100 ml/min) was interrupted for three con- These included determining
secutive 7 minute periods. This group of ani- 1. The flow rates that would be necessary
mals were compared to our previous groups to meet the metabolic needs of the myo-
of continuous retrograde warm blood car- cardium in both normal and hypertro-
dioplegia and intermittent, antegrade/retro- phied hearts, and in obstructed and
grade cold blood cardioplegia.3 After 45 min- unobstructed territories of the myocardium,
utes of cardioplegic arrest, pH increased in 2. how long the cardioplegia could be in-
hearts receiving antegrade/retrograde cold terrupted in normal and acutely is-
blood cardioplegia, remained the same in the chemic muscle,
continuous retrograde warm blood cardiople- 3. what clinical methods could be used to
gia group, and actually decreased in the ani- monitor ongoing ischemia during
mals when the retrograde warm blood car- cardioplegic interruption, and
dioplegia was interrupted. This was reflected 4. what technique could be used to reverse
in the wall motion scores in the area at risk ischemic changes following periods of
which were significantly lower in the inter- intermittent warm blood cardioplegic
rupted retrograde warm blood cardioplegia delivery.
group. Furthermore, wall motion scores in The aim of this chapter will be to review
non-ischemic areas of the myocardium were the current status of interrupting warm blood
also significantly lower in the interrupted cardioplegia in the clinical practice of cardiac
group. This resulted in an area of necrosis surgery. Recent experimental and clinical stud-
which was significantly higher when warm ies will be highlighted in an attempt to fur-
blood cardioplegia was interrupted. We con- ther defined the limits and role of this tech-
cluded that interrupting warm blood car- nique as a method of myocardial protection.
dioplegia accentuates damage in acutely
ischemic myocardium and may also be detri- Experimental Studies
mental to areas of myocardium without Carrier and his co-workers6 sought to
obstructed vessels. determine the effects of interrupting cold and
Our experimental work raised serious ques- warm blood cardioplegia on myocardial pH
tions about the safety of interrupting warm and the release of metabolic markers in a
blood cardioplegia. While our results showed canine model. Following sequential arrest
that regional ischemia was accentuated by in- periods of 5, 10, 20, and 40 minutes, hearts
terrupting warm blood cardioplegia, the effects were reperfused with either cold (12˚C) or
of interrupting warm blood cardioplegia in warm (37˚C) blood cardioplegia at 100 ml/min
myocardium supplied by unobstructed myo- for 10 minutes. Although pH decreased
cardium was still unknown. Lichtenstein, Abel, significantly in both groups there was no
and Salerno suggested that interrupting warm difference in the absolute pH or )pH between
blood cardioplegia actually preconditioned the warm and cold blood cardioplegia. The
heart to subsequent periods of ischemia.1 This amount of lactate production and creatine ki-
was based on studies of regional ischemia in nase release was also similar in both groups.
the beating heart which showed that brief The release of troponin T was higher in the
intermittent episodes of normothermic warm blood cardioplegia group but this did
ischemia reduces myocardial stunning and lim- not reach statistical significance. Their results
its necrosis after subsequent periods of
176 Ischemia-Reperfusion Injury in Cardiac Surgery

also showed that periods of warm ischemic (37˚C) for 90 minutes. Another group received
arrest > 10 minutes and periods of cold is- six 5 minute periods of warm blood cardiople-
chemic arrest > 20 minutes were more likely gia infusion followed by six 20 minute periods
to result in greater pH changes and more re- of ischemia. The last group had no ischemia
lease of lactate, creatine kinase, and troponin. during the 150 minutes of perfusion. NMR
This suggests that the margin of safety for is- studies showed that a 10 minute interruption
chemic arrest is prolonged when hypothermic of warm blood cardioplegia decreased creat-
rather than warm blood techniques are used. ine phosphate levels and intracellular pH by
In another canine model, Ko and his col- approximately 47% (p < 0.01) and 0.12 units
leagues7 arrested three groups of seven dogs (p < 0.05). Resumption of cardioplegia for 5
with antegrade warm blood cardioplegia. The minutes resulted in almost complete recovery
control group received continuous warm blood of creatine phosphate and intracellular pH
cardioplegia while the other two groups re- values. More importantly, subsequent inter-
ceived multidose warm blood cardioplegia ad- ruptions did not result in any cumulative
ministered every 5 or 10 minutes during a 30 changes in creatine phosphate levels or intrac-
minute period of ischemic arrest. Systolic and ellular pH beyond those changes observed after
diastolic recovery was impaired after arrest in the initial cardioplegic interruption. During
animals that had received intermittent warm reperfusion, there were no significant differ-
blood cardioplegia every 10 minutes, and mild ences in ATP and creatine phosphate levels
stunning was observed when warm blood car- among the groups. Recovery of contractility
dioplegia was administered every 5 minutes. as assessed by balloon function curves did not
Landymore and co-workers8 extended the differ amongst the groups. Hence, in this
period of interruption to 15 minutes in a dog experimental study using normal, isolated
model comparing intermittent antegrade hearts, interruption of warm blood cardiople-
warm blood cardioplegia to intermittent gia for up to 10 minutes appeared to be well
antegrade cold blood cardioplegia. Hearts were tolerated. Similar results were reported by Tonz
arrested by giving an initial dose of 30 ml/kg and co-workers10 in an intact pig model.
delivered at a rate of 150 ml/min. Subsequent Antegrade warm blood cardioplegia was given
infusions were given every 15 minutes for 15 minutes followed by a 5 minute inter-
(10 ml/kg) at a rate of 150 ml/min during the ruption or for 10 minutes followed by a 10
90 minute arrest period. Left ventricular sys- minute period of interruption during a 180
tolic elastance and end-diastolic elastance was minute period of ischemic arrest. Interrupt-
assessed using pressure-volume loops. Systolic ing antegrade warm blood cardioplegia for 10
function was well preserved whereas diastolic minute intervals resulted in no significant dif-
function decreased slightly in both groups. ference in systolic or diastolic function com-
High energy phosphates decreased signifi- pared to 5 minutes of interruption or to those
cantly in both groups during reperfusion. This animals which received 180 minutes of con-
study used global parameters to assess LV func- tinuous cardioplegic perfusion. Torracca and
tion in normal animals without coronary co-workers11 also concluded that 10 minutes
artery lesions or ventricular hypertrophy. of antegrade warm blood cardioplegia inter-
Landymore could not demonstrate any supe- ruption was safe in an isolated blood-perfused
riority for interrupting warm blood cardiople- rabbit model. Antegrade warm blood car-
gia over established cold blood cardioplegia dioplegia was stopped for 10 minutes followed
techniques in this model. by 5 minutes of reperfusion. The sequence was
Further insight into the metabolic effects repeated three times for a total of 45 minutes.
of interrupting warm blood cardioplegia was Compared to continuous warm blood car-
provided by Tian and co-workers9 using NMR dioplegia, the intermittent group had no sig-
techniques. Isolated blood perfused pig hearts nificant changes in ATP content or contrac-
were divided into three groups. One group tility as assessed by balloon function curves.
received continuous warm blood cardioplegia However, there was a transient but significant
Interrupting Warm Blood Cardioplegia 177

release of creatine kinase and lactate after sion was delivered into the aortic root. Myo-
the first minute of reperfusion suggesting cardial temperatures were maintained between
anaerobic glucose metabolism. This may be 12 and 18˚C. Patients were systemically cooled
potentially harmful in areas of acutely ischemic to 25-28˚C. Warm blood cardioplegia was
myocardium or when cross-clamp times are delivered during 60 ± 3% of the arrest period;
longer than 45 minutes which is frequently while cold blood cardioplegia was delivered
seen in clinical practice. only 28 ± 5% of the cross-clamp time
In summary, experimental studies seem to (p = 0.009). There were no significant differ-
suggest that antegrade warm blood cardiople- ences between operative mortality, sternal
gia can be interrupted for as long as 10 min- infections, perioperative myocardial infarc-
utes without significant changes in high energy tions, strokes, and the need for inotropic or
phosphate levels or contractility after a period intra-aortic balloon support between the
of reperfusion. However, it should be noted groups. Immediately after removal of the cross-
that these studies were performed in normal clamp, warm hearts showed more washout of
hearts without coronary lesions or periods of accumulated lactate (p < 0.05 versus cold).
ongoing ischemia, or ventricular hypertrophy. This lactate production was thought to be re-
Based on these experimental results, clinical lated to the inadequate delivery of cardioplegic
studies were conducted to further define the solution in the LAD distribution which was
efficacy of the intermittent warm blood car- grafted with an internal mammary artery and
dioplegia technique in clinical practice. was the last territory to be grafted. Unfortu-
nately, regional function in the LAD distribu-
Clinical Studies tion was not assessed in this study but end-
Yau and co-workers12 performed a random- systolic elastance and preload recruitable stroke
ized trial comparing continuous antegrade work index were not depressed in the warm
warm blood cardioplegia with intermittent blood group. Furthermore, the MB fraction
cold blood cardioplegia in patients undergo- of creatine kinase was lower in the warm hearts.
ing elective coronary artery bypass graft sur- Since the incidence of infarcts was similar in
gery performed by a single surgeon. The cases both groups, this suggests that the increased
were of low risk; over 80% of patients had an release of creatine kinase in the cold hearts may
ejection fraction ∃ 40%. Proximal and distal have been due to reversible disruption of
anastomoses were performed under a single sacrolemnal integrity, allowing transient trans-
cross-clamp period. After an initial arresting membrane loss of these high-molecular weight
dose of 500 ml; antegrade, warm blood car- proteins. In summary, interrupting antegrade
dioplegia was infused at 80 ml/min and inter- warm blood cardioplegia appeared to have
rupted “whenever necessary” to allow adequate been well tolerated in these low risk patients.
visualization of each distal anastomosis. After Delivering cardioplegia through the completed
the interruption, a “catch-up” infusion was vein grafts undoubtedly decreased ischemic
given through the aortic root and in each com- damage in some areas of the heart with criti-
pleted vein graft. The volume of this infusion cal coronary stenoses. Nevertheless, concern
was calculated by the perfusionist to precisely is raised about the increased lactate washout
compensate for the period of cessation of flow in the warm blood group; especially in the
and to maintain the preset cardioplegic flow LAD distribution. This potentially could have
rate. These patients were not actively cooled; resulted in regional dysfunction in more
systemic temperatures were allowed to drift acutely ischemic myocardium.
toward 30-32˚C. In the cold blood group, after A similar prospective, randomized study
an arresting dose of 500 ml; 100 ml of low K+ was performed by Pelletier and co-workers13
(10 mEq/L) cardioplegia was infused intermit- comparing intermittent, warm blood car-
tently into each vein graft after completion of dioplegia and intermittent cold blood car-
each distal anastomosis, and a 400 ml infu- dioplegia in 200 low risk CABG patients. After
an initial arresting dose of 300 ml of high K+
178 Ischemia-Reperfusion Injury in Cardiac Surgery

cardioplegia, a low K+ antegrade, warm blood interval averaged 11.4 ± 4.0 minutes but ex-
cardioplegic solution was administered tended from 6.8 ± 5.4 to 15.3 ± 5.0 minutes
(200-300 ml) after each distal anastomosis. according to the individual surgeon. In this
The maximal single time of interruption was study, repeated interruptions of antegrade
12-15 minutes; cardioplegia was delivered warm blood cardioplegia were less likely to lead
30-40% of the cross-clamp period. In 13% of to adverse clinical results if single interruptions
the intermittent warm blood group, a sus- were ∀ 13 minutes. Their results were similar
tained electromechanical arrest could not be to earlier experimental studies which suggested
achieved and these patients crossed over to the that the longest ischemic interval was more
cold blood group. The cold blood group had important than the cumulative ischemic
similar rates of infusion through the aortic time.9,10 Nevertheless, their results also showed
root. The mortality (1% vs 1%) and infarct that the chance of an adverse outcome in-
rates (2% warm, 4% cold) were similar in each creased when the period of interruption ex-
group. There was no difference in the need ceeded 13 minutes; especially in patients with
for pharmacologic or mechanical support left main disease, decreased LV function, and
between the two groups. The serum levels of those requiring more urgent surgery.
postoperative creatine kinase were again sig- Chritakis and his co-workers15 studied the
nificantly lower in the warm blood group sug- effects of intermittent antegrade, warm blood
gesting that hypothermia may result in tem- cardioplegia on postoperative right ventricu-
porary membrane dysfunction resulting in a lar (RV) function in 52 isolated CABG
higher leak of creatine kinase which is not patients. Patients were randomized to receive
indicative of permanent injury or myocardial either intermittent antegrade, warm blood car-
necrosis. These results appear to confirm dioplegia (37˚C) with systemic pump flow at
Yau’s12 observation that warm blood cardiople- 35-37˚C or intermittent antegrade, cold blood
gia can be safely interrupted in low risk surgi- cardioplegia (5-8˚C) with systemic tempera-
cal patients. However, it should be noted that tures ranging from 25-30˚C. All hearts re-
Pelletier and his co-workers had to alter their ceived an arresting high K+ dose of 200-300
cardioplegia technique in 13% of patients ml/min followed by a continuous low K+ in-
because a sustained electromechanical arrest fusion of 100-150 ml/min. Cardioplegia was
could not be achieved. interrupted only during the construction of
In a further attempt to define the safe the distal anastomoses. After the completion
period that warm blood cardioplegia can be of each distal, a 500 ml bolus of cardioplegia
interrupted, Lichtenstein and co-workers14 was given at 200-300 ml/min after which the
conducted a randomized study of 720 patients rate was decreased to 100-150 ml/min.
comparing cold blood cardioplegia with warm Cardioplegia was also given through each
blood cardioplegia. Following an initial arrest- completed vein graft and both groups received
ing dose of high K+ cardioplegia, a low K+ a terminal infusion of 350 ml of warm (37˚C)
solution was delivered at 50-150 ml/min. After cardioplegia prior to removing the cross-clamp.
periods of interruption, warm blood cardiople- Total ischemic time was longer in the cold
gia was delivered at 200-300 ml/min after group (42 ± 8 vs 31 ± 14 min; p < 0.001).
which the rate was reduced to 50-150 ml/min. Right ventricular ejection fraction, RV end-
Cardioplegia was also administered through diastolic volume index, and RV end-systolic
completed vein grafts. No retrograde infusions volume index, as derived from a thermodilu-
were given. Seventeen percent of patients in tion REF-1 catheter, were found to be signifi-
the warm group crossed over to the cold blood cantly higher in the warm blood group at 6
group because of excessive coronary “flooding” and 8 hours after bypass. However, these mea-
or difficulty achieving or maintaining a cardiac surements are affected by changes in RV
arrest. In this study, warm blood cardioplegia afterload, and their significant improvement
was interrupted for approximately 50% of the over the cold group most likely reflect the
cross-clamp time. The longest single ischemic decreased pulmonary vascular resistance and
Interrupting Warm Blood Cardioplegia 179

warmer systemic temperatures in the warm gia. Cardioplegia was also administered
blood group. This may be a beneficial effect directly into each completed vein graft in both
of normothermic cardiopulmonary bypass and groups. In the patients receiving antegrade/
not warm blood cardioplegia. Furthermore, at retrograde cardioplegia, a continuous infusion
6 and 8 hours postop, nearly one-third to one- of retrograde cardioplegia was interrupted only
quarter of the patients in both groups were for visualization of the anastomosis. Intermit-
excluded because they required either tent infusions (250 ml) of antegrade cardiople-
inotropes or afterload reducing agents, or were gia were given after completion of each proxi-
shivering and had a tachycardia. Christakis mal anastomosis. Infusion of cardioplegia was
noted in his discussion that ischemic times never done simultaneously in an antegrade and
were longer in the cold group “because all sur- retrograde manner. Cardioplegia was least in-
geons felt more confident with the luxury of terrupted in the combined group (17 ± 3%
hypothermia” and that “surgeons were more combined vs 28 ± 4% retrograde vs 36 ± 4%
expeditious when constructing anastomoses in antegrade p < 0.05). The combined group also
the warm group.” Their data actually showed received significantly more cardioplegic solu-
that the increased ischemic time taken to per- tion (6.3 ± 0.4 L combined vs 4.8 ± 0.3 L
form the distal anastomoses in the cold group retrograde; vs 4.6 ± 0.2 L antegrade; p <
had no detrimental effects on right ventricu- 0.001). No differences in clinical outcomes
lar or left ventricular function. were found among the groups. Myocardial lac-
Our earlier experimental studies showed tate concentrations were highest in the LAD
that the presence of an LAD occlusion limits territory during antegrade cardioplegia deliv-
cardioplegia delivery when given in an ery. Retrograde cardioplegia had greater lac-
antegrade fashion and that this damage may tate production in the right and left ventricles
be accentuated when warm antegrade car- and decreased ATP concentrations. The com-
dioplegia is used for myocardial protection.3,4 bination of antegrade and retrograde car-
Although continuous retrograde cardioplegia dioplegia resulted in the best preservation of
results in better delivery of cardioplegia beyond ATP, least lactate production and accumula-
a coronary occlusion,3 it may still provide lim- tion, and the best global LV and RV function.
ited perfusion to the posterior septum and Hence, the technique which resulted in the
right ventricle. 16 Hayashida and his col- most cardioplegic delivery and least interrup-
leagues17 therefore postulated that a combi- tions gave the best myocardial protection.
nation of antegrade and retrograde delivery of
cardioplegia might be required to provide bet- Summary and Conclusions
ter perfusion of the myocardium when warm This chapter has reviewed the experimen-
blood cardioplegia techniques are used. They tal and clinical studies in the development of
undertook a prospective, randomized clinical techniques used to interrupt warm blood car-
study in low risk patients undergoing CABG dioplegia. Most experimental studies have been
comparing intermittent, warm antegrade performed in animals without coronary artery
blood cardioplegia, intermittent, retrograde disease or ventricular hypertrophy. Many in-
warm blood cardioplegia, and a combination volve isolated preparations which may not be
of antegrade/retrograde warm blood cardiople- analogous to clinical practice. The majority of
gia. Both antegrade and retrograde groups clinical studies have been performed on low
received an initial arresting antegrade dose of risk patients with normal ejection fractions,
500 ml of high K+ cardioplegia. Cardioplegia no previous infarcts, and without any on-go-
was then administered either retrograde or ing ischemia. Assessment of left ventricular
antegrade at 200 ml/min. The cardioplegia was performance has generally been global with
interrupted when necessary for visualization load dependent indices. Since many of the cold
of the anastomosis. A “catch-up” infusion was blood cardioplegia patients were cooled to
then given, averaging 150-200 ml/min to com- 25-28˚C, their systemic vascular resistance
pensate for the period of cessation of cardiople-
180 Ischemia-Reperfusion Injury in Cardiac Surgery

tended to be higher in the earlier postop lated with ongoing ischemia in the arrested
period, thus resulting in lower stroke work heart and could not be used as an on-line
indices compared to patients treated with measurement of active ischemia. Myocardial
warm blood cardioplegia. Despite these limi- pH has been used to reliably detect regional
tations, certain conclusions can be drawn ischemia during cardiac surgery and can give
regarding the technique of interrupting warm on-line data.18 Surgeons using warm blood car-
blood cardioplegia. dioplegia in patients where the solution must
How long can warm blood cardioplegia be be interrupted for greater than 10 minutes may
safely interrupted? It appears that interrupt- wish to consider monitoring pH values.
ing warm blood cardioplegia for up to 10 min- What techniques could be used to reverse
utes in low risk patients without active ischemic changes following periods of inter-
ischemia is well tolerated and results in no in- mittent warm blood cardioplegic delivery? It
creased mortality or infarct rate and that glo- is the adequacy of distribution of cardioplegia
bal left ventricular function appears to be pre- and not the temperature which is the most
served.6,9-11,14 The length of the period of important factor in limiting ischemia during
interruption of cardioplegia and not the total aortic cross-clamping. In the presence of sig-
ischemic time is the most important param- nificant coronary stenoses, antegrade car-
eter in determining myocardial performance dioplegia alone may not provide adequate
and patient outcome.9,14 The safe period for myocardial protection.3,19 This may result in
interrupting warm blood cardioplegia in further ischemic injury when warm blood car-
actively ischemic patients is still undetermined. dioplegia is interrupted.4 Infusing cardiople-
It appears however, that when cardioplegia is gia through completed vein grafts is one
interrupted for longer than 13 minutes, there method by which cardioplegic distribution
is a greater likelihood of an adverse outcome may be improved. Furthermore, continuous
in patients with left main disease, decreased infusion of cardioplegia through completed
left ventricular function, and those requiring vein grafts during the time that antegrade flow
urgent surgery.14 is interrupted may limit problems with visu-
What are the optimal flow rates to meet alization in the operative field while still
the needs of the myocardium when warm adequately perfusing other areas of the myo-
blood cardioplegia is interrupted? The exact cardium. Another technique to maximize
flow rates for normal, ischemic, and hyper- cardioplegic distribution is the combination
trophied myocardium have yet to be deter- of antegrade/retrograde delivery. Hayashida
mined. Nevertheless, experimental and clini- and his colleagues17 demonstrated that inter-
cal studies using antegrade techniques suggest mittent antegrade infusions of warm cardio-
that an arresting dose of high K+ cardioplegia plegic solution during continuous warm ret-
(25-30 mEq) at 100-150 ml/min should be rograde cardioplegic delivery provided more
adequate for the nonischemic, nonhyper- homogenous distribution and prevented
trophied myocardium. Following the interrup- ischemic myocardial injury during warm heart
tion of cardioplegia, a “pay-back” dose of operations. It would seem that the combina-
200 ml/min is usually given for 2-3 min- tion of antegrade/retrograde cardioplegia plus
utes followed by the baseline flow rate infusion of cardioplegia through the completed
(100-150 ml/min).15,17 vein grafts would provide the most optimal
What clinical methods can be used to myocardial protection in those instances when
monitor ongoing ischemia during the period warm blood cardioplegia must be interrupted.
of cardioplegic interruption? NMR is imprac- What is the role of interrupting warm
tical in the operating room and may not be blood cardioplegia in the current practice of
able to localize abnormalities in regional myo- cardiac surgery? Randomized clinical studies
cardial metabolism. Lactate washout is also have yet to show a clear-cut superiority of
non-specific and impractical in clinical cardiac warm blood cardioplegia over cold blood
surgery. ATP levels may not always be corre- cardioplegic techniques. It is important to
Interrupting Warm Blood Cardioplegia 181

remember that cardioplegia is merely an 9. Tian G, Xiang B, Butler KW et al. A 31P-


adjunct to the surgeons’ main goal which is to nuclear magnetic resonance study of intermit-
tent warm blood cardioplegia. J Thorac
perform a technically perfect operation. This Cardiovasc Surg 1995; 109:1155-1163.
is even more important in the current prac- 10. Tonz M, Krogmann ON, Hess OM et al.
tice of cardiac surgery where optimal visual- Effect of intermittent warm blood cardiople-
ization is needed to bypass vessels which are gia on functional recovery after prolonged
cardiac arrest. Ann Thorac Surg 1996;
smaller with more diffuse disease, to recon- 62:1146-1151.
struct valves, and to replace valves with 11. Torracca L, Pasini E, Curello S et al. Con-
advanced deterioration. Surgeons must rely on tinuous versus intermittent warm blood car-
cardioplegic techniques which provide an dioplegia: Functional and energetic changes.
unobscured field to construct a perfect tech- Ann Thorac Surg 1996; 62:1172-1179.
12. Yau TM, Ikonomidis JS, Weisel RD et al.
nical anastomosis as well as afford excellent Ventricular function after normothermic ver-
myocardial protection for acutely ischemic sus hypothermic cardioplegia. J Thorac
hearts with lower ejection fractions. Surgeons Cardiovasc Surg 1993; 105:833-844.
employing intermittent warm blood cardiople- 13. Pelletier LC, Carrier M, Leclerc Y et al. In-
termittent antegrade warm versus cold blood
gia must understand the limitations of this cardioplegia: A prospective, randomized study.
technique and be able to alter their method- Ann Thorac Surg 1994; 58:41-49.
ology when necessary in order to safely achieve 14. Lichtenstein SV, Naylor CD, Feindel CM et
both these goals. al. Intermittent warm blood cardioplegia. Circ
1995; 92:341-346.
15. Christakis GT, Buth KJ, Weisel RD et al.
References Randomized study of right ventricular
1. Lichtenstein SV, Abel JG, Salterno TA. function with intermittent warm or cold
Warm heart surgery and results of operation cardioplegia. Ann Thorac Surg 1996; 61:
for recent myocardial infarction. Ann Thor 128-135.
Surg 1991; 52:455-460. 16. Caldarone CA, Krukenkamp IB, Misare BD
2. Salerno TA, Houck JP, Barrozo CAM et al. et al. Perfusion deficits with retrograde warm
Retrograde continuous warm blood cardiople- blood cardioplegia. Ann Thorac Surg 1994;
gia: a new concept in myocardial protection. 57:403-406.
Ann Thorac Surg 1991; 51:245-247. 17. Hayashida N, Ikonomidis JS, Weisel RD et
3. Matsuura H, Lazar HL, Yang X et al. Warm al. Adequate distribution of warm cardioplegic
versus cold blood cardioplegia—Is there a solution. J Thorac Cardiovasc Surg 1995;
difference? J Thorac Cardiovasc Surg 1993; 110:800-812.
105:45-51. 18. Khuri SF, Josa M, Martson W et al. First
4. Matsuura H, Lazar HL, Yang XM et al. Det- report of intramyocardial pH in man. II.
rimental effects of interrupting warm blood Assessment of adequacy of myocardial pres-
cardioplegia during coronary revascularization. ervation. J Thorac Cardiovasc Surg 1983;
J Thorac Cardiovasc Surg 1993; 106:357-361. 86:667-678.
5. Cohen MV, Liu GS, Downey JM. Precondi- 19. Haan C, Lazar HL, Bernard S et al. Superi-
tioning by a brief coronary occlusion preserves ority of retrograde coronary sinus cardiople-
wall motion in ischemia/reperfusion. Circ gia over antegrade cardioplegia following re-
1990; 82:271-275. vascularization of an acute coronary occlusion.
6. Carrier M, Tourigny A, Thoribe N et al. Ann Thorac Surg 1991; 51:408-412.
Effects of cold and warm blood cardioplegia
assessed by myocardial pH and release of
metabolic markers. Ann Thorac Surg 1994;
58:764-767.
7. Ko W, Zelano J, Fahey L et al. Warm is-
chemic tolerance of the arrested heart. Euro-
pean J Cardiothorac Surg 1993; 7:295-299.
8. Landymore RW, Marble AE, Fris J. Effect
of intermittent delivery of warm blood car-
dioplegia on myocardial recovery. Ann Thorac
Surg 1994; 57:1267-1272.
Section IV:
Surgical Techniques
B. Protection Strategies
CHAPTER 16

Myocardial Protection Strategies


in Routine Coronary and Valve
Operations
Kiyozo Morita and Michio Yoshitake

Integrated Myocardial This method is based on the following


Management principles:
1. Adequate visualization for surgical pre-
The goal of every cardiac operation must be a
cision is optimized by a still, bloodless
technically perfect anatomic result contribut-
field, accomplished by intermittent
ing to functional improvement that requires
administration of cold potassium blood
adequate visualization in a quiet, bloodless
cardioplegia.
operative field without postoperative deterio-
2. Uniform myocardial distribution of
ration in cardiac function. The essential pre-
cardioplegic solution is ensured by the
requisite for a successful cardiac operation is
combination of antegrade and retro-
the use of simple and safe cardioprotective
grade cardioplegic delivery.
techniques to avoid myocardial damage, while
3. Warm blood cardioplegia is used for
maintaining a quiet, bloodless field.
active resuscitation of preoperatively
A variety of myocardial protection strate-
impaired myocardium and/or avoid-
gies have been developed, and the clinical
ance of reperfusion injury, in addition
effectiveness of each strategy has been con-
to cold multidose blood cardioplegia.
firmed.1-4
4. Ischemia is unnecessary under the cir-
This chapter describes an integrated tech-
cumstances of the procedure when
nique of myocardial protection with warm and
visualization is nonproblematic and
cold blood cardioplegia in conjunction with
continuous coronary perfusion is pos-
the combined use of antegrade and retrograde
sible in an arrested myocardium.
delivery for routine adult cardiac operations.
5. Continuous blood perfusion of the cold
This approach of integrated myocardial man-
arrested heart does not require car-
agement, developed by Buckberg and associ-
dioplegic solution. Noncardioplegic
ates,5 combines the advantages of various
normal cold blood can be used to maintain
cardioprotective strategies to compensate for
arrest.
their individual shortcomings and incorporates
the strategies of warm/cold blood cardiople-
gia, antegrade/retrograde delivery, continuous/
intermittent infusion, and blood/blood
cardioplegic perfusion during a single period
of aortic clamping.

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


184 Ischemia-Reperfusion Injury in Cardiac Surgery

Background and Logic supplementation of depleted substrates, while


of Integrated Myocardial keeping the heart arrested to lower its oxygen
demands.8 Thus, ATP is channeled to repara-
Management tive processes in myocardial metabolism. The
formulation of warm cardioplegic solution
Combined Cold must be modified (hypocalcemic, alkalotic and
and Warm Blood Cardioplegia enriched with amino acids, glutamate and
The primary advantage of cold blood car- aspartate) to limit calcium influx during reflow,
dioplegia is that it couples the myocardial buffer acidosis9 and replenish key Krebs cycle
nourishment by intermittent oxygenation and intermediates depleted during ischemia.10
substrates supply with the capacity to lower The clinical study by Teoh and associates10
myocardial oxygen demands and the rate and demonstrated that warm cardioplegic reper-
development of ischemic damage in the fusion (terminal warm blood cardioplegia)
hypothermic arrested heart.1 accelerates myocardial metabolic recovery after
Despite the theoretical advantage of con- reperfusion. It is now generally accepted that
tinuous warm blood perfusion to prevent warm, controlled reperfusion provides a pow-
ischemia,2,3 the use of cold blood cardiople- erful tool to limit reperfusion injury and nul-
gia and subsequent myocardial hypothermia lify the adverse effects of prolonged aortic
may be essential to prevent myocardial dam- clamping.12 Additionally, the clinical benefits
age during intentional ischemia when blood of warm induction in energy-depleted hearts
supply must be interrupted for surgical preci- were demonstrated in coronary patients with
sion. The capacity of cold intermittent blood cardiogenic shock13,14 and advanced valvular
cardioplegia to prevent ischemic damage in the disease.15 In the integrated management strat-
normal myocardium is adequaet for up to 4 egy, multidose cold blood cardioplegia to allow
hours of the aortic clamping, as shown experi- interrupting coronary perfusion for surgical
mentally.6 Intermittent cold blood cardiople- precision is incorporated with warm blood
gia techniques, however, can only prevent fur- cardioplegic induction and reperfusion in
ther deterioration but cannot correct metabolic order to combine the individual benefits of
defects that may compound the underlying warm and cold cardioplegia to maximize the
mechanical cardiac disorder requiring correc- cardioprotective effects of blood cardioplegia.
tion. The method may be insufficient in the
energy- and substrate-depleted heart, and Antegrade/Retrograde Cardioplegic
impaired function may persist, as demon- Delivery
strated experimentally.7 The benefits of cardioplegia are provided
Warm blood cardioplegia was initially only if the solutions are distributed to all
introduced as “warm blood cardioplegic myocardial regions in sufficient amounts.
reperfusion” to limit reperfusion injury by a Antegrade cardioplegia is used by most sur-
brief period of administration of warm blood geon and produces prompt cardiac arrest, but
cardioplegia at the initial phase of reperfusion is distributed poorly in coronary patients with
(before aortic unclamping).8-10 Subsequently, severely stenotic or occluded coronary arter-
the concept of “warm cardioplegic induction”, ies unless it is delivered through the vein
delivery of warm blood cardioplegia to induce graft.4,16,17 These strategies are ineffective in
cardiac arrest after aortic clamping, was intro- patients who receive arterial grafts or who have
duced to actively resuscitate the ischemically diffuse coronary artery disease. Further limi-
damaged, energy-depleted heart and improve tation of antegrade cardioplegia include:
its tolerance to subsequent intervals of cold poor distribution in patients with aortic
ischemia.7 Administration of warm blood regurgitation unless the aorta is opened and
cardioplegic solution restores aerobic ATP pro- the coronary ostia are perfused directly; risk
duction by maximizing myocardial metabolic of potential ostial injury during aortic valve
activity by normothermic blood perfusion and procedure from direct perfusion; and the need
Myocardial Protection Strategies in Routine Coronary and Valve Operations 185

to interrupt the continuity of mitral valve op- Intermittent Blood Cardioplegia


erations to remove the retractors and avoid and Continuous Perfusion
aortic distortion during cardioplegic replen- of Noncardioplegic Cold Blood
ishment. These limitations of antegrade car- A dry, quiet operative field is a prerequi-
dioplegia can be overcome by the use of retro- site for a technically precise cardiac procedure.
grade cardioplegic delivery.4,16 In the presence Most surgeons arrest the heart with high
of complete coronary artery occlusion, this dose potassium blood cardioplegia (20 mEq/L)
method would appear to result in a better per- and use multidose low dose potassium
fusion of the ischemic myocardial area.4 (8-10 mEq/L) as interrupting coronary per-
The development of transatrial coronary fusion for the remainder of the operation. In
sinus cannulation technique has made simple, a routine adult cardiac operation, however,
safe, and rapid access to the coronary sinus ischemia is unnecessary under the circum-
feasible,18 and retrograde cardioplegia is now stances, when visualization is nonproblematic,
generally accepted as a routine method for and therefore continuous coronary perfusion
intraoperative myocardial protection. Never- is feasible if the hearts can be kept arrested.
theless, experimental17 and clinical19-21 studies For that purpose, continuous blood perfu-
have shown that the right ventricle and poste- sion of the cold arrested heart does not require
rior septum are not adequately perfused with cardioplegic solution (noncardioplegic normal
retrograde cardioplegia. Inconsistent right ven- cold blood is possible).
tricular protection may be problematic in Our recent experimental study26 showed
patients with marked right ventricular hyper- that cold arrested hearts remain quiescent and
trophy or pulmonary hypertension. This rec- both ventricles recovered completely when
ognized limitation of right ventricular perfused with cold 4-10c retrograde non-
hypoperfusion can be counteracted partially cardioplegic blood. Consequently, the advan-
by cold cardioplegic perfusion, inasmuch as tages of continuous perfusion and nourish-
coronary sinus retroperfusion provides right ment to shorten real ischemic interval can be
ventricular hypothermia as the effluent achieved without the drawbacks of excessive
traverses conductance vessels, and therefore hemodilution and cardioplegic overdose.
confers hypothermic lowering of oxygen These benefits are possible only with “cold”
demands to counteract this limited nutritive noncardioplegic blood since electromechani-
oxygen supply due to veno-veno shunts. This cal activity (i.e., beating or fibrillation) returns
benefit is achieved only if hypothermia is used and myocardial oxygen demands rise when
in conjunction with retrograde cardioplegic warm blood is delivered.
delivery.
Aforementioned limitations of antegrade
and retrograde perfusion have stimulated the
Method of Integrated Myocardial
use of combined antegrade/retrograde infusion Management and Surgical
of cardioplegia (alternating between antegrade Techniques
and retrograde20-24 or simultaneous antegrade/
retrograde perfusion25). In an editorial com- The Cardioplegic Solution
ment, Menasche4 suggested that a combined The blood cardioplegic formulations for
antegrade and retrograde approach may be the standard 4:1 ratio (Blood:Cardioplegia) is
safer and more effective in obtaining a more listed in Table 16.1. The formulation is
uniform myocardial distribution of blood car- modified for the other ratio (i.e., 8:1
dioplegia. Blood:Cardioplegia), as reported previously.5
For standard protocol, four cardioplegic
solutions are prepared: a high and low K+
amino acid enriched solution for warm induc-
tion and reperfusion, and a high and low K+
nonamino acid enriched solution for cold
186 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 16.1. The blood cardioplegic formulations for the standard 4:1 ratio
(Blood:Cardioplegia) in the integrated myocardial management. The formulation is
modified for the other ratio (i.e., 8:1 Blood:Cardioplegia), as reported previously.5

Cardioplegic Volume Component Concentration


Additive Added(mL) Modified Delivered

Cold Induction

Tham (0.3M) 200 pH 7.7-7.8


CPD 50 Ca++ 0.5-0.6 mM/L
5% dextrose & H2O 550 Osm 340-360 mOsm
KCL (2mEq/mL) 30 K+ 18-20 mEq
Warm induction
Tham (0.3M) 225 pH 7.5-7.6
CPD 225 Ca++ 0.15 mM/L
50%dextrose 40 Glucose > 400mg
KCL (2mEq/mL) 40 K+ 20-25 mEq
Glutamate 13 mM/L
250 substrate
Aspartate 13 mM/L
5% dextrose & H2O 220 %Osm 380-400 mOsm
Cold Maintenance
Tham (0.3M) 200 pH 7.7-7.8
CPD 50 Ca++ 0.5-0.6 mM/L
5% dextrose & H2O 550 Osm 340-360 mOsm
KCL (2mEq/mL) 10 K+ 8-10 mEq
Warm Reperfusion

Tham (0.3M) 225 pH 7.5-7.6


CPD 225 Ca++ 0.15-0.25 mM/L
50%dextrose 40 Glucose > 400mg %
KCL (2mEq/mL) 15 K+ 8-10 mEq
Glutamate 13 mM/L
250 substrate
Aspartate 13 mM/L
5% dextrose & H2O 200 Osm 380-400 mOsm

induction and maintenance doses. Currently, occur when large volumes of amino acids are
only two cardioplegic solutions: a high K+ used.
amino acid enriched solution and a low K+
nonamino acid enriched solution are made up Cardioplegic Delivery System
for each procedure, as the solution for warm Blood cardioplegia delivery system used for
induction is used also for cold induction and the integrated myocardial management con-
warm reperfusion in a routine procedure to tains a single roller pump, heat exchanger for
simplify the preparation. Glutamate and altering cardioplegic temperature, and coro-
aspartate are not added to the maintenance nary perfusion- monitoring set for alternating
solution, as peripheral vasodilatation may between antegrade and retrograde routes
(Fig. 16.1). The blood component of cardio-
Myocardial Protection Strategies in Routine Coronary and Valve Operations 187

plegic solution is withdrawn from the coro- Antegrade Cannula


nary port of the oxygenator. The blood is An aortic cannula containing ports of
mixed in a ratio of 4:1 (blood:cardioplegia) monitoring perfusion pressure and aortic vent-
by fixing the calibers of the tubing coming ing is required for delivery of antegrade car-
from the cardioplegic solution and coronary dioplegia, when used in combination with ret-
port of the oxygenator. The tubes are then rograde cardioplegia. The aortic vent line is
routed through a single occlusive roller pump, outfitted with a one-way pop-off valve which
with an insert to accommodate the different is kept on low suction throughout the proce-
sizes of the tubing and into a heat exchanger dure. Alternatively, the vent line can be con-
with the capacity of rapid alteration of blood nected to a Y-connector in the venous line to
temperature so that the cardioplegic solution provide gravity drainage.
can be delivered either warm (37˚C) or cold Aortic pressure monitoring is crucial to
(8-10˚C) as required under different ensure cardioplegic delivery and it allows:
circumstances. A) detection of aortic regurgitation; B) inad-
As a heat exchanger, standard Advance vertent failure to close the aortic vent line;
BCDTM or more recently developed Buckberg C) determination of optimal flow rate in
BCD VanguardTM (Sorin Biochemical Inc., hypertrophied ventricle; and D) regulation of
Irvine, CA) is exclusively utilized. the pressure during controlled reperfusion.
Two heater/coolers (one for the cardio- Clinical cardioplegic perfusion pressure of
plegic solution, and the other for systemic 80-100 mmHg are safe during cardioplegic
perfusion) are required in order to control tem- induction. Conversely, keeping perfusion pres-
perature of the heart and body independently. sure at or below 50 mmHg during reinfusion
A heat exchanger, Buckberg BCD and reperfusion will be desired to protect
VanguardTM (Fig. 16.2) has efficient heat edema formation and endothelial integrity.
exchange performance, rapid priming capac-
ity and low priming volume (35 mL device Retrograde Cannula
only), integral filter, bubble trap and pressure The cannula for retrograde delivery of
monitoring port. choice is used routinely to deliver retrograde
The pressure port is essential so the perfu- cardioplegia via coronary sinus and simulta-
sionist can determine if there is obstruction neously monitoring coronary sinus pressure.
in the system during cardioplegic administra- We exclusively use a RetroplegiaTM cannula
tion, and make adjustments to avoid disrup- with the self-inflated textured balloon (RC-
tion of the heat exchanger by excessive 014-T, Research Medical, Inc., Midvale, Utah)
pressure. containing a specially designed stylet with a
The outlet arm of the heat exchanger is low pressure, self inflating and deflating bal-
connected to a cardioplegic infusion line. For loon and an integral second lumen to permit
alternating between antegrade and retrograde pressure monitoring. The flow lumen is
delivery, we use antegrade/retrograde designed to have the small holes distal to the
infusion and pressure monitoring set balloon and larger holes present within the
(MIS-001-CP) (Research Medical Inc., balloon. This design allows the balloon to
Midvale, Utah) containing two arms of each become self inflated as the cardioplegic solu-
infusion and pressure monitoring line specially tion flows through the cannula, and therefore
designed to allow rapid switching from occludes back flow from the coronary sinus as
antegrade to retrograde cardioplegia and simul- the remainder of the solution flows through
taneous monitoring of aortic and coronary the smaller distal holes. This also allows the
sinus pressure. The side arm attached to the balloon to be self deflated spontaneously when
retrograde cardioplegic delivery arm permits the infusion is completed. The inflated bal-
perfusion via vein graft or coronary ostial can- loon is only 1.8 cm in diameter and contains
nula (Fig. 16.3). allow intraluminal pressure, so that it cannot
188 Ischemia-Reperfusion Injury in Cardiac Surgery

Fig. 16.1. Cardioplegic delivery system containing a single occlusive roller pump, tubing system, heat exchanger
with the capacity of rapid alteration of blood cardioplegic temperature and mixture of blood and cardioplegic
solution, heater/cooler, and the specially designed cardioplegic infusion line for alternating between antegrade and
retrograde delivery. As a heat exchanger, standard Advance BCD TM or Buckberg BCD Vanguard TM (Sorin
Biochemical Inc., Irvine, CA) is routinely utilized.

Fig. 16.2. Buckberg BCD VanguardTM (Sorin Biomedi-


cal Inc. Irvine, CA) used for warm/cold blood
cardioplegic delivery. This heat exchanger has efficient
heat exchange performance, rapid priming capacity and
low priming volume (35 mL device only), integral filter,
bubble trap and pressure monitoring port.
Myocardial Protection Strategies in Routine Coronary and Valve Operations 189

Fig. 16.3. Schematic for delivering antegrade/retrograde blood cardioplegia and pressure monitoring. Antegrade/
retrograde infusion and pressure monitoring set (MIS-001-CP) (Research Medical Inc., Midvale, Utah) is utilized
for cardioplegic infusion, and Retroplegia TM with self-inflated textured balloon (RC-014-T, Research Medical,
Inc., Midvale, Utah) is used for retroperfusion

damage the coronary sinus by over inflation Retroplegia cannulation is accomplished most
and produce barotrauma during infusion. readily before starting extracorporeal circula-
The coronary sinus pressure must be dis- tion or can be done on partial bypass. A 3-0
played so that the surgeon and perfusionist can prolene suture is placed in the low right atrium,
be aware of it during infusion. near the IVC junction, and is withdrawn
through a rubber tourniquet. This low posi-
Placement of Retroplegia tion for placement is selected to avoid dislodg-
The Retroplegia cannula is always placed ment of the retroplegia cannula during retro-
after venous cannulation to avoid inadvertent perfusion or retraction of the heart for
dislodgment when the IVC cannula is inserted.
190 Ischemia-Reperfusion Injury in Cardiac Surgery

exposure of the circumflex and distal branches that the self-inflating balloon is not occluding
of the right coronary artery. the coronary sinus completely, or that the cath-
The stylet and cannula are introduced into eter is dislodged.
the right atrium through a small puncture Under these circumstances, the position of
anterior to the two-stage cannula, and inserted the catheter tip must be tested by palpation of
far enough to advance the balloon into an the coronary sinus. The adequacy of retro-
intra-atrial position. The suture and tourni- perfusion can be assumed when the catheter
quet are snagged to prevent bleeding. Place- has been retracted, but remains in the coro-
ment of the index finger at the IVC-right atrial nary sinus, by simply compressing the coro-
junction will allow palpation of the tip of the nary sinus-right atrial junction against the IVC
cannula, guide its entrance into the coronary cannula. This always results in a prompt rise
sinus and verify its correct placement position. in coronary sinus pressure and avoids the need
By this maneuver, advancement of cannula for repositioning the cannula. Sometimes the
into coronary sinus can be accomplished cannula is retracted and slipped away from the
readily. Normally coronary sinus course is in coronary sinus due to the back pressure in the
a direction that extends at a approximate 45˚ coronary sinus at the initiation of retrograde
angle directly toward the left shoulder from infusion. A gradual increase of the flow rate
its orifice to its position beneath the left atrial over 12-15 seconds will minimize catheter
appendage. Verification of the correct place- retraction during infusion. Failure to en-
ment of the stylet is made by palpating the sure adequate coronary sinus pressure
course of the coronary sinus behind the heart. (20-50 mmHg) during cardioplegic infusions
When the proper positioning is confirmed, the will result in inadequate retrograde myocar-
cannula is advanced little further over the stylet dial protection and therefore retrograde car-
until it meets any resistance, while holding the dioplegia must be abandoned.
stylet in place. This maneuver will place can-
nula at the coronary sinus great coronary vein Standard Protocol for Integrated
junction beneath the left atrial appendage. The Blood Cardioplegia
stylet is then withdrawn while cannula is held
The cardioplegic dose is generally divided
in place. The pressure port is connected to
by alternating between antegrade and retro-
ensure coronary sinus pressure. A high coro-
grade delivery in a standard protocol for a
nary sinus pressure (i.e., > 20 mmHg) indi-
routine adult open heart surgery. Flow guide-
cates the catheter is wedged.
lines used for alternative administration of
antegrade and retrograde blood cardioplegia
Pressure Monitoring During for induction, maintenance, reperfusion and
Retrograde Cardioplegia continuous noncardioplegic cold blood per-
The coronary sinus pressure ranges usually fusion are listed in Table 16.2.
30-50mmHg when retrograde cardioplegia is
delivered at 200-250 mL/min. The surgeon Cardioplegic Induction
and perfusionist must be aware of the coro- Antegrade cardioplegia is always given first
nary sinus pressure during infusion so that the to produce prompt arrest, followed by
appropriate correction can be made if the coro- retroperfusion of the remainder of the
nary sinus pressure is either too high or too cardioplegic dose, except aortic valve insuffi-
low. A coronary sinus pressure > 50 mmHg ciency patients. Failure to achieve prompt
means the catheter is lodged too far into the arrest (i.e., 1-2 minutes) suggests that either
coronary sinus or is in the posterior descend- there is incomplete aortic clamping, aortic
ing coronary vein and this can be corrected by regurgitation, or inadequate venous drainage
reducing the flow rate immediately, with- with admixture of cardioplegia with blood that
drawing the catheter and restarting retro- escapes the venous return cannula. Reappli-
grade flow. Conversely, a coronary sinus pres- cation of the aortic clamp will ensure that the
sure < 20 mmHg during infusion indicates aorta is occluded completely. The presence of
Myocardial Protection Strategies in Routine Coronary and Valve Operations 191

Table 16.2. Flow guidelines used for alternative administration of antegrade and
retrograde blood cardioplegia for induction, maintenance, reperfusion and continuous
noncardioplegic cold blood perfusion. The flow rates are increased by 50-100mL/min
for hypertrophied hearts.

Cardioplegia Antegrade Retrograde

Cold Induction 300 mL/min (2 min) 200mL/min (2min)


Warm induction 300 mL/min until arrest 150 mL/min (2.5 min)
then 150 mL/min (2.5 min)
Maintenance 200 mL/min (1 min) 200 mL/min (1 min)
Noncardioplegic Blood 150 mL/min
Warm reperfusion 150 mL/min (2 min) 150 mL/min (2 min)

aortic regurgitation will be noted in case of a nary perfusion is interrupted when there is a
low aortic perfusion pressure during cardio- need to ensure a bloodless operative field, oth-
plegic induction, and the most common cause erwise retrograde perfusion of noncardioplegic
of this is distortion of the noncoronary cusp cold blood is continued. The conversion from
by venous cannula. This can be corrected antegrade to retrograde cardioplegia is made
readily by retracting the venous cannula down- by turning the stopcock and activating the
ward and to the right. Clinically unsuspected aortic vent line.
mild aortic regurgitation can be handled by The aortic vent line is kept open until the
increasing the flow rate of the antegrade car- next antegrade cardioplegia to provide cardiac
dioplegia to 500 mL/min, and by compress- decompression and to evacuate air from the
ing the right ventricular outflow with a sponge aorta when the next antegrade infusion is
stick to push the septum against the posterior started.
left ventricular wall beneath the aortic valve.
Failure of these maneuvers to restore satisfac- Warm Induction
tory aortic perfusion pressure at the increased The heart is stopped with a warm (37˚C)
flow rate immediately should lead to abandon- substrate enriched (glutamate/aspartate) high
ment of the antegrade cardioplegic infusion potassium (20-25 mEq/L) cardioplegic solu-
and to complete reliance upon retrograde tion (Table 16.1) when there is clinically sig-
cardioplegia as the only method of myocar- nificant preoperative impairment of cardiac
dial protection. function (i.e., cardiogenic shock, advanced
valve disease, extending myocardial infarction).
Cold Induction The purpose of warm induction is to “resusci-
Cold cardioplegic induction with 8-10˚C tate” the heart and improve its tolerance to
blood cardioplegia is used during all elective subsequent ischemia. Warm induction is
operations where preoperative cardiac function achieved by circulating warm water from the
is reasonable (i.e., LVEF > 40%). The heater/cooler into a heat exchanger. Warm
antegrade dose is given at 300-350 mL/min induction is delivered antegradely first. The
for 2 minutes to stop the heart and the initial infusion is started at 300-350 mL/min
remainder of the induction dose is delivered until the heart stops, and the infusion rate is
retrograde at 200-250 mL/min for 2 minutes. then slowed to 150-200 mL/min. The total
The cardioplegic volume and infusion rate are dose is divided relatively equal between
increased in hypertrophied hearts (2-minute antegrade and retrograde delivery. The total
antegrade at 350 mL/min, and 2-minute ret- period of warm infusion is 5 minutes (2.5
rograde at 250 mL/min). After the comple- minutes antegrade and 2.5 minutes retrograde)
tion of retrograde cardioplegic perfusion, coro-
192 Ischemia-Reperfusion Injury in Cardiac Surgery

Warm induction is followed always by a 3-4 sutures into the prosthetic valve or annulo-
minute infusion of cold cardioplegia (8-10˚C) plasty ring and closure of the atriotomy or
at 200-250 mL/min (1/2 antegrade and 1/2 aortotomy).
retrograde) with the low K+ cardioplegic solu- A one-minute infusion of the cardioplegic
tion to ensure cardioplegic distribution and maintenance solution is delivered every
cooling. For this purpose, one to two minutes 10-20 minutes during prolonged cold
before the completion of initial warm induc- noncardioplegic blood perfusion or if electro-
tion, the heater/cooler is switched to the mechanical activity returns spontaneously.
maximum cooling mode to lower the tempera- This ensures maintenance of arrest and avoids
ture of cardioplegic solution rapidly down to cold noncardioplegic blood perfusion of the
10˚C. heart with electromechanical activity.
Perfusion is always stopped if blood
Multidose Administration of Cold obscures vision, as visualization is never com-
Blood Cardioplegia (Cold promised to maximize surgical precision.
Maintenance Cardioplegia)
Multidose administration of cold low K+ Warm Reperfusion
blood cardioplegia are repeated to replenish The objective of warm reperfusion is to
the solution washed away by noncoronary limit reperfusion injury by lowering energy
collateral flow at approximately 20-25 min- demands. This is done by keeping the heart
utes intervals. Reinfusion are delivered at arrested during the initial period of reflow, and
200-250mL/min for 2 minutes, with 1-minute optimizing metabolic rate of repair by raising
antegrade and 1-minute retrograde, whether the temperature to normothermia, while add-
or not electromechanical activity returns for ing substrates, buffering acidosis, and limit-
the following reasons: maintains arrest; restores ing the calcium load.27 Warm reperfusate
hypothermia; buffers acidosis; washes away (Table 16.1) is delivered before aortic
acid metabolites; and replenishes substrates. unclamping in all patients at 150 mL/min for
In coronary operations, all reinfusions are 3-5 minutes, while monitoring perfusion pres-
divided equally between antegrade and retro- sure and is divided equally by alternating
grade routes of delivery (1 minute antegrade between antegrade and retrograde delivery.
and 1 minute retrograde). Conversely, all During application of warm reperfusion, it is
reinfusions during mitral and aortic valve crucial to maintain persistent cardiac arrest
operations are delivered retrograde for 2 min- without any electromechanical activity and to
utes so that the procedure can proceed unin- avoid distention of the left ventricle. Further-
terrupted, unless the right ventricle starts to more keeping perfusion pressure at or below
contract. If this occurs, 1 minute infusion is 50 mmHg during antegrade warm reperfusion
delivered at 20-25 minutes intervals via the by controlling flow rate will be desired to pro-
antegrade route in mitral operations or the tect edema formation and endothelial integ-
right coronary ostia in aortic valve procedures rity. Rewarming of the blood cardioplegic
in order to ensure right ventricular protection. solution is started approximately 2-3 minutes
before starting the warm reperfusate, and is
accomplished by switching the heater-cooler
Noncardioplegic Cold Blood
to warming mode. The duration of warm
Retroperfusion cardioplegic reperfusion is prolonged to 20
After the completion of antegrade and ret- minutes when the operation is for acute evolv-
rograde reinfusion of cold blood cardioplegia, ing myocardial infarction, since prolonged
retrograde cold noncardioplegic blood is con- controlled reperfusion enhances early func-
tinuously perfused usually at 150 mL/min, tional recovery after regional ischemia, as
when it does not compromise visualization experimentally shown in a report by Allen and
during the aspects of the procedure (i.e., proxi- associates.27
mal anastomosis of coronary graft, placing
Myocardial Protection Strategies in Routine Coronary and Valve Operations 193

Special Consideration Valve Operations


for the Type of Surgical
Procedure Aortic Valve Operation
In aortic stenosis, an antegrade cardioplegic
induction is started antegradely, followed by
Coronary Operation retrograde delivery as the aorta is opened. With
In general, the aforementioned standard aortic regurgitation, cardiopulmonary bypass
protocol for integrated management is applied is initiated normothermically to prevent
to all coronary patients. In coronary opera- ventricular distention and ventricular fibrilla-
tions, all infusions of cardioplegia are divided tion; cardioplegia is administered retrograde
equally between antegrade and retrograde at a higher flow rate than the standard proto-
routes of delivery, inasmuch as the adequate col, while the aorta is opened. The coronary
distribution of cardioplegic solution is of cru- perfusion cannula connected to the side arm
cial importance in patients with coronary dis- of cardioplegic infusion set is available for
ease. The current strategies for coronary bypass selective coronary ostial perfusion. The left and
grafting include constructing all anastomoses right coronary ostia is cannulated promptly
during a single period of aortic clamping, in patients with aortic regurgitation and car-
delivering the cardioplegic solution through dioplegia is delivered until the standard dose
either aortic root or coronary sinus, and per- for antegrade induction is completed and elec-
fusing cardioplegic solution through the graft tromechanical activity completely disappears.
after each distal anastomosis if needed. Graft A coronary perfusion cannula must be intro-
perfusion is particularly important if the right duced into the right coronary ostia to ensure
coronary artery is revascularized with the vein right ventricular protection.
graft, since the right ventricle may not be suf- Retrograde cold cardioplegia is given for 2
ficiently perfused and protected with either minutes each 20-25 minutes exclusively at
antegrade aortic root perfusion or retro- 200 mL/min to avoid ostial perfusion for valve
perfusion. The side arm of cardioplegic infu- excision, placement of sutures in the annulus,
sion set is used for graft perfusion. Alterna- and securing the prosthesis.
tively, cardioplegic delivery is ensured by Cold noncardioplegic blood is infused con-
antegrade perfusion provided that each proxi- tinuously at 150 mL/min while sutures are
mal anastomosis is accomplished immediately placed from the annulus to the valve ring, and
after each distal anastomosis. while the aortotomy is closed. Particularly in
After antegrade and retrograde cardioplegic complicated aortic and mitral operation (i.e.,
induction, cold ischemia is often required double valve replacement with annular
during distal anastomosis, but continuous enlargement procedure), a relatively extended
retroperfusion of noncardioplegic cold blood period of continuous perfusion is technically
is used when it does not compromise visual- feasible to shorten ischemic interval.
ization (i.e., dissection of coronary arteries, The color of the effluent blood from the
preparation of arterial grafts, or proximal anas- coronary ostia is inspected during retro-
tomosis of vein grafts). The obligatory perfusion. A well oxygenated effluent is inter-
prolongation of aortic clamping is counter- preted to reflect adequate ventricular perfu-
balanced by the improved cardioplegic sion (sufficient flow to meet the low demands
distribution. of arrest during hypothermia) and operation
Anterograde warm reperfusion is started ret- is performed exclusively with retroperfusion
rogradely prior to the completion of proximal via the coronary sinus with blood cardiople-
anastomosis and followed by antegrade warm gia and noncardioplegic blood. Conversely,
reperfusion. During antegrade warm intermittent right coronary ostial perfusion is
reperfusion, the clamps of vein grafts are released mandatory if the right coronary effluent is very
so that antegrade perfusion is delivered to the desaturated, as this is considered to indicate
segments that were revascularized.
194 Ischemia-Reperfusion Injury in Cardiac Surgery

that right ventricular perfusion is potentially Combined Valve and Coronary


deficient. Procedures
The retrograde warm blood cardioplegic The distal coronary anastomoses are always
reperfusate is initiated approximately 4-5 min- constructed first in all combined valve and
utes before it is anticipated that the aortotomy coronary procedures using cold intermittent
incision will be closed and is followed by doses of antegrade and retrograde blood car-
antegrade perfusion after the aorta is de-aired dioplegia as described previously. This allows
completely. all subsequent cardioplegic infusions to be
The clamp is removed and replaced imme- delivered from the graft (if needed).
diately 1/3-1/2 way across the site of clamp- The vein introducers in the coronary graft
ing to create a dome to purge retained air. are connected to the cardioplegic infusion set
containing one or more side arms so that the
Mitral Valve Operation cardioplegic solution can be delivered via the
After antegrade and retrograde cardioplegic grafts during the valve procedure. Myocardial
induction (1/2 antegrade and 1/2 retrograde), management during aortic and mitral proce-
cold cardioplegic or blood perfusion is exclu- dures are as described previously. Retro-
sively retrograde for the remainder of the perfusion of cold noncardioplegic blood is
operation with the exception of the period of continued as the proximal anastomoses are
warm reperfusion. Primary reliance is placed started. During antegrade warm reperfusion,
upon retrograde perfusion, since mitral retrac- the clamps of vein grafts is released so that
tors may make the aortic valve incompetent antegrade perfusion is delivered to the seg-
and make antegrade perfusion ineffective. ments that had antegrade revascularization.
Cold cardioplegia is given for 2 minutes at
20-25 minute interval exclusively via retro-
grade route at 200 mL/min for valve repair, References
valve excision, placement of sutures in the 1. Follette DM, Mulder DG, Maloney JVJ et
annulus, and securing the prosthesis. During al. Advantages of blood cardioplegia over con-
tinuous coronary perfusion and intermittent
mitral procedures, however, the coronary ischemia. J Thorac Cardiovasc Surg 1978;
effluent from the retroperfusion drainage 76:604-619.
which comes from the coronary ostia is suc- 2. Lichtenstein SV, Ash KA, El-Dalati H et al.
cessfully harvested from suction placed into Warm heart surgery. J Thorac Cardiovasc
the left ventricle, and continuous perfusion of Surg 1991; 101:269-274.
noncardioplegic cold blood is maintained 3. Salerno TA, Houck JP, Barrozo CAM et al.
Retrograde continuous warm blood cardiople-
without compromising visualization in most gia: a new concept in myocardial protection.
of the aspects of mitral procedure. Perfusion Ann Thorac Surg 1991; 51:245-247.
is always stopped if blood obscures vision, as 4. Menasche’ P, Subayi JB, Veyssie’ L et al.
visualization is never compromised to maxi- Efficacy of coronary sinus cardioplegia in
mize surgical precision. patients with complete coronary artery occlu-
sions. Ann Thorac Surg 1991; 51:423-428.
Intermittent doses of the cardioplegic 5. Buckberg GD, Beyersdorf F, Allen BS. Inte-
maintenance solution are administered for one grated myocardial management in valvular
minute every 10-20 minutes of prolonged cold heart disease. J Heart Valve Disease 1995; 4
noncardioplegic blood retroperfusion or when- (suppl. II):S 198-213.
6. Robertson JM, Vinten-Johansen J, Buckberg
ever electromechanical activity returns. GD et al. Safety of prolonged aortic clamp-
The retrograde warm blood cardioplegic ing with blood cardioplegia. I Glutamate
reperfusate is initiated as the atriotomy clo- enrichment in normal hearts. J Thorac
sure is begun. The antegrade portion of the Cardiovasc Surg 1984; 88:395-401.
warm reperfusate is then delivered
Myocardial Protection Strategies in Routine Coronary and Valve Operations 195

7. Rosenkranz ER, Okamoto F, Buckberg GD 18. Buckberg GD, Drinkwater DC, Laks H. A
et al. Safety of prolonged aortic clamping new technique for delivering antegrade/retro-
with blood cardioplegia. III Aspartate enrich- grade blood cardioplegia without right heart
ment of glutamate-blood cardioplegia in en- isolation. Eur J Cardiothorac Surg 1990;
ergy-depleted hearts after ischemic and 4:163-168.
reperfusion injury. J Thorac Cardiovasc Surg 19. Allen BS, Winkelmann JW, Hanafy H et al.
1986; 91:428-435. Retrograde cardioplegia does not adequately
8. Follette DM, Steed DL, Foglia RP et al. Re- perfuse the right ventricle. J Thorac
ducing postischemic myocardial damage by Cardiovasc Surg 1995; 109:1116-1126.
maintaining arrest during initial reperfusion. 20. Ardehali A, Gates RN, Laks H et al. The
Surg Forum 1977; 28:281-283. regional capillary distribution of retrograde
9. Follette D, Fey K, Buckberg GD et al. Re- blood cardioplegia in explanted human hearts.
ducing postischemic damage by temporary J Thorac Cardiovasc Surg 1995; 109:935-940.
modification of reperfusate calcium, potas- 21. Carrier M, Gre’goire J, Khalil A et al. Myo-
sium, pH, and osmolarity. J Thorac cardial distribution of retrograde cardioplegic
Cardiovasc Surg 1981; 82:221-238. solution assessed by myocardial thallium-210
10. Lazar HL, Buckberg GD, Manganaro AM et uptake. J Thorac Cardiovasc Surg 1994;
al. Myocardial energy replenishment and re- 108:1115-1118.
versal of ischemic damage by substrate en- 22. Buckberg GD. Antegrade/ retrograde blood
hancement of secondary blood cardioplegia cardioplegia to ensure cardioplegic distribu-
with amino acids during reperfusion. J Thorac tion: operative techniques and objectives. J
Cardiovasc Surg 1980; 80:350-359. Card Surg 1989; 4:216-238.
11. Teoh KH, Christakis GT, Weisel RD et al. 23. Jegaden O, Eker A, Montagma P et al.
Accelerated myocardial metabolic recovery Antegrade/retrograde cardioplegia in arterial
with terminal warm blood cardioplegia. J grafting: metabolic randomized clinical trial.
Thorac Cardiovasc Surg 1986; 91:888-895. Ann Thorac Surg 1995; 59:456-461.
12. Kirklin JW. Technical and scientific advances 24. Aldea GS, Hou D, Fonger JD et al. Inho-
in cardiac surgery over the past 25 years. Ann mogeneous and complementary antegrade and
Thorac Surg 1990; 49:26-31. retrograde delivery of cardioplegic solution in
13. Rosenkranz ER, Buckberg GD, Mulder DG the absence of coronary artery obstruction. J
et al. Warm induction of cardioplegia with Thorac Cardiovasc Surg 1994; 107:499-504.
glutamate-enriched blood in coronary patients 25. Ihnken K, Morita K, Buckberg GD et al. The
with cardiogenic shock who are dependent on safety of simultaneous arterial and coronary
inotropic drugs and intra-aortic balloon sup- sinus perfusion: experimental background and
port: Initial experience and operative strategy. initial clinical results. J Cardiac Surg 1994;
J Thorac Cardiovasc Surg 1983; 86:507-518. 9:15-25.
14. Beyersdorf F, Kirsh MM, Buckberg GD et 26. Ihnken K, Morita K, Buckberg GD. New
al. Warm glutamate/aspartate-enriched blood approaches to blood cardioplegic delivery to
cardioplegic solution for perioperative sudden reduce hemodilution and cardioplegic over-
death. J Thorac Cardiovasc Surg 1992; dose. J Card Surg 1994; 9:26-36.
104:1141-1147. 27. Allen BS, Okamoto F, Buckberg GD et al.
15. Cunningham J, Rosenkranz ER, Okamoto F Studies of controlled reperfusion after is-
et al. Benefits of normothermic induction of chemia. XV. Immediate functional recovery
blood cardioplegia in energy-depleted hearts after 6 hours of regional ischemia by careful
with maintenance of arrest by multidose cold control of conditions of reperfusion and com-
blood cardioplegia. J Thorac Cardiovasc Surg position of reperfusate. J Thorac Cardiovasc
1982; 84:676-683. Surg 1986; 92:621-635.
16. Noyez L, van Son JAM, van der Werf T et
al. Retrograde versus antegrade delivery of
cardioplegic solution in myocardial revascu-
larization. J Thorac Cardiovasc Surg 1993;
105:854-863.
17. Partington MT, Acar C, Buckberg GD et al.
Studies of retrograde cardioplegia. II. Nutri-
tive blood flow distribution in normal and
jeopardized myocardium. J Thorac Cardiovasc
Surg 1989; 97:613-.
CHAPTER 17

Acute Myocardial Infarction


and Cardiogenic Shock
Francesco Donatelli, Marco Pocar and Adalberto Grossi

T
he surgical approach to myocardial achieved within 4-6 hours from the onset of
infarction during the acute evolving symptoms.4
phase has been controversial and its In parallel, the other technique that has
role has changed during the last decades. In been developed for the treatment of acute
fact emergency coronary bypass surgery has evolving myocardial infarction is percutane-
been adopted in some centers since the ous transluminal angioplasty (PTCA) of the
1970s.1-3 Results were acceptable, but far from diseased vessel. Reperfusion of the ischemic
satisfactory. However, this applied to medical area can be achieved in about 90% of cases,
therapy as well. Thus, research was directed in-hospital mortality is around 10% and sys-
towards improvement of both, surgical and tolic left ventricular function improves in more
nonsurgical, strategies. What is certainly well than 50%. On the other hand, an early
documented is that revascularization proce- reocclusion rate up to 30% has been docu-
dures should preferably be accomplished mented and results are not satisfactory in case
within approximately 6 hours from the onset of elderly patients, women, multivessel coro-
of myocardial ischemia. This fact rises logistic nary artery disease, poor left ventricular func-
problems in the standardization of in-hospi- tion and acute infarction associated with car-
tal treatment of such a widely diffused and life- diogenic shock.5,6 PTCA as an adjunctive
threatening disease. procedure associated with successful throm-
Initial reports of surgically treated patients bolysis does not appear to provide clear ben-
with acute evolving myocardial infarction efits compared with isolated thrombolysis and
documented mortality rates as low as 2%,1,2 is correlated with higher morbidity.7,8
but selection certainly played an important Most data concerning surgical revascular-
role: factors such as three-vessel disease or ization in acute myocardial infarction are
transmural infarction, as well as cardiogenic nonrandomized and selection of patients is
shock, adversely affected outcome raising over- likely to be linked to encouraging results. A
all mortality to about 25%. The advent of randomized trial has demonstrated low mor-
thrombolytic therapy for acute coronary tality rates (2.9%) when bypass surgery was
occlusion has added knowledge to the effect accomplished at a mean of 4.3 hours from the
of early reperfusion on myocardial function onset of ischemia.9 On the contrary, mortal-
and survival, and has gradually become the ity was 8.8% in the medically treated patients.
most widely adopted treatment for acute evolv- This trial, however, excluded patients over 65
ing infarction, due to its simplicity. Many tri- years of age, or with a history of cardiogenic
als have shown improvement in left ventricu- shock or prior myocardial infarction, but it is
lar function and survival if reperfusion is interesting to notice that mortality, at an 18

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Acute Myocardial Infarction and Cardiogenic Shock 197

months follow-up, raised to 20% in the medi- in the current era, in the absence of complica-
cal series, while all surgical survivors where tions and additional risk factors. Patients are
alive. generally selected for surgery when they
It appears that some form of revasculariza- present with diffuse coronary artery disease, a
tion, simple thrombolysis of the acutely history of prior myocardial necrosis often with
occluded coronary vessel, PTCA, or surgical left ventricular dysfunction of varying sever-
bypass, during the acute phase of evolving ity, and especially when the hemodynamic
myocardial infarction, i.e., during the first 6 state is unstable or cardiogenic shock is already
hours from the onset of ischemia, is beneficial present. Surgical treatment for acute myocar-
in reducing the necrotic area of myocardium dial infarction is also widely considered the
and hospital mortality.10 In particular, surgi- treatment of choice in case of failed PTCA
cal revascularization offers some advantages to procedures or unsuccessful thrombolytic
the alternative and less invasive procedures: therapy. In the latter case coronary bypass sur-
1. it allows complete revascularization in gery has been performed with mortality rates
virtually all patients (in 100% of pa- of about 15%, but often bleeding complica-
tients that could benefit from PTCA or tions represent a major concern.11,16,17 Obvi-
thrombolysis) reducing the incidence of ously, patients with mechanical complications
recurrent infarction and angina pecto- of myocardial infarction are surgical candi-
ris, and reducing mortality at 1 year af- dates, almost invariably on an urgent basis or
ter infarction from 10% to about 2- as true emergencies. Encouraged by the good
3%;11 results obtained with the myocardial protec-
2. specific protocols of myocardial protec- tive strategies suggested by Buckberg and as-
tive measures can be aggressively insti- sociates, and taking into account that the high
tuted to allow recovery of optimal meta- mortality and morbidity regarding the surgi-
bolic conditions of the acutely ischemic cal treatment of patients with acute evolving
and energy-depleted myocardium: this myocardial infarction reported in the litera-
obviously applies to the nonperfused ture are possibly determined by selection of
segment depending from the acutely poor candidates for revascularization proce-
occluded coronary artery, but also, and dures in general and not to coronary bypass
very importantly, to the viable, and surgery itself, we started since 1994 to per-
hypercontractile, remaining remote form coronary operations in selected patients
myocardium; with acute myocardial infarction.18,19
3. despite suboptimal mortality rates,
around 15% for elderly patients with Indications for Operation,
multivessel disease or left ventricular Preoperative Management
dysfunction and often well over 20-
25% for patients presenting in cardio- and Surgical Technique
genic shock,10,12-15 it gives the possibil-
ity to improve results in high-risk Indications for Operation
situations compared to conventional and Preoperative Management
therapy. In this respect, it is worth re- Patients are selected for coronary artery
membering that mortality in medically bypass grafting during acute evolving myocar-
treated patients in cardiogenic shock dial infarction not only in emergency condi-
due to acute evolving myocardial inf- tions or after failed thrombolysis or PTCA,
arction approaches 100% and that but also in case of hemodynamically stable
pump failure is the most common cause patients with ongoing ischemia and extensive
of death in this population. areas of jeopardized myocardium. In other
Despite the considerations cited above, words, surgery is performed not only in the
coronary artery bypass grafting for acute evolv- case no other therapeutic option is available,
ing myocardial infarction is seldom performed, but is primarily selected in some patients. The
198 Ischemia-Reperfusion Injury in Cardiac Surgery

extension of the myocardial area at risk of of cases, the majority of these patients become
necrosis is the critical issue in the indication balloon-dependent and eventually die in the
for immediate operation: when the infarct hospital: the one-year mortality rate is around
results in a loss of approximately 40% or more 95%.15
of the left ventricular mass, cardiogenic shock On admission, primary diagnosis is estab-
results. In this respect, the extension of the lished by electrocardiography and echo-
critically ischemic area is dependent also on cardiography. If the patient’s clinical picture
other important factors such as the presence corresponds to one of the conditions listed
of prior myocardial infarction or left ventricu- above, no attempt to perform successful
lar dysfunction (not necessarily on an ischemic thrombolysis is made and the coronary
basis) and the gradual dysfunction of the anatomy is immediately outlined by angiog-
hypercontractile remote ischemic muscle, the raphy. The patient is then transferred to the
severity of which is generally well correlated operating room as quickly as possible. Preop-
with the presence of multivessel coronary erative intra-aortic balloon counterpulsation
artery disease. is instituted when necessary, namely, in case
Summarizing, we select for surgery patients of hemodynamic instability or cardiogenic
in the following conditions: shock. A widespread use of aortic counterpul-
sation in this setting would be reasonable from
Acute Evolving Myocardial Infarction a theoretical and pathophysiologic standpoint,
(Within 6 Hours from the Onset but may often necessitate additional time to
of Myocardial Ischemia) insert the balloon (especially if percutaneous
techniques are unsuccessful, e.g., in the pres-
1. anterior infarction with ST segment el- ence of severe peripheral vascular disease);
evation in at least 4 leads; therefore we do not recommend its routine
2. infarction with concomitant inferior, use.
posterior and lateral extension; Not all patients that present in the afore-
3. infarction with inferior, but biven- mentioned clinical conditions are surgical can-
tricular involvement; didates. Although absolute and relative
contraindications for operation may be present
Cardiogenic Shock Secondary to Acute (as for any other surgical candidate), they are
Myocardial Infarction (Within 3 Hours not related to the severity of the hemodynamic
from the Onset of Shock) status, except for moribund patients. Patients
It is to be noted, however, that indication over 75 years of age have not yet been consid-
for operation may still be present despite an ered for immediate coronary surgery, but this
interval longer than 6 hours from the onset of limit is probably arguable.
ischemia because the rationale for surgery in What must finally be emphasized is the
this setting is primarily based on the principle importance of logistic and organizational as-
that survival is dependent on reperfusion of pects of such a therapeutic strategy. Coordi-
the reversibly ischemic peri-infarct segments nation between cardiologists, cardiac surgeons,
and remote areas of hypercontractile myocar- anesthesiologists, the emergency department
dium supplied by critically stenotic coronar- and, eventually, physicians or cardiologists
ies. Cardiogenic shock is clinically defined as from other hospitals in the surrounding area,
the presence of hypotension (mean arterial is crucial in avoiding time loss and is greatly
pressure < 60 mmHg), anuria (urine output responsible in determining a successful out-
< 0.5 ml/Kg/hr) and metabolic acidosis. As come: more in particular, it is essential to per-
outlined previously, mortality of medically form coronary angiography as soon as possible.
treated cardiogenic shock approaches 100%.
Although intra-aortic balloon counter- Surgical Technique
pulsation can reverse this state in about 80% The general planning of the operation is
similar to standard coronary artery bypass
Acute Myocardial Infarction and Cardiogenic Shock 199

grafting. However, some details in surgical acutely ischemic region first in order to pro-
technique and especially in myocardial pro- vide maximal cardioplegic protection in the
tective strategies are, in our opinion, impor- most vulnerable area. In contrast, in the pres-
tant and should be emphasized. In fact, we ence of cardiogenic shock secondary to myo-
started this surgical protocol encouraged by cardial infarction, emergency surgery is indi-
the good results obtained adopting the method cated primarily to rescue the stunned remote
of myocardial management suggested by viable muscle as this situation often ensues after
Buckberg and associates. In our experience, 6 hours from the onset of ischemia: conse-
this consideration applies, in particular, to quently, the bypass graft to be constructed last
coronary artery surgery in patients with should be the one supplying the acutely
unstable angina or severe left ventricular dys- occluded vessel.
function, and to energy-depleted hearts in In the clinical practice, we have slightly
general. modified this operative protocol. This relates
Cardiopulmonary bypass is maintained at in particular to a wider use of retrograde car-
moderate levels of systemic hypothermia dioplegia. In fact (not only in this subset of
(32˚C). In patients with acute myocardial in- patients) we frequently infuse maintaining
farction, as in patients with left ventricular doses for 2 minutes retrograde rather than 1
dysfunction, the left ventricle is vented minute antegrade and 1 minute retrograde, as
through the right superior pulmonary vein. originally suggested, because this does not in-
An aortic root vent is also placed as it is part terrupt the flow of the operation (it is
of the cardioplegia circuitry. As already stated undoubtedly an ideal route for continuous car-
above we follow the Buckberg protocol for all dioplegia). In addition to several technically
cardiac operations. More in particular, in this convenient situations (aortic valve surgery and
population the strategy for energy-depleted regurgitation; severely calcified or porcelain
hearts 20-22 is applied in all cases. This con- ascending aorta; mitral valve operations after
sists of the infusion of antegrade and retro- exposure of the left atrium with a distorted
grade (coronary sinus) normothermic blood aortic valve; aortic dissection), retrograde
(blood:cardioplegia ratio 4:1) substrate-en- infusion, and occasionally induction, may
riched cardioplegic induction (KCl 20-25 actually provide superior protection in a num-
mEq/L, glutamate and aspartate 13 mM/L, ber of patients with diffusely stenotic or
37˚C, 2.5 minutes antegradely and retro- occluded coronary arteries, and in case of left
gradely respectively), followed by hypother- main disease.23-28
mic induction (KCl 8-10 mEq/L, 4-8°C, In this respect we perform the 20-minute-
1.5 minutes antegradely and retrogradely), period of controlled reperfusion in a retrograde
maintaining infusions (KCl 8-10 mEq/L, fashion at the end of all distal anastomoses.
nonsubstrate-enriched, 4-8°C, 1 minute This technique has, in our opinion, several
antegrade and retrograde) and controlled advantages:
antegrade reperfusion (500 ml, KCl 1. it provides controlled and homoge-
8-10 mEq/L, 37°C, substrate-enriched; then neous controlled reperfusion to all myo-
unmodified blood for 2 minutes to wash-out cardial segments. Some degree of stun-
the cardioplegic solution prior to aortic ning in myocardial regions other than
unclamping). When surgery is performed for the infarct (remote myocardium) is
acute myocardial infarction, the protocol likely to occur in the presence of
includes 20 minutes of additional controlled multivessel disease or prior myocardial
reperfusion of the infarcted area through a infarction;
saphenous vein graft after aortic unclamping, 2. even if this technique necessitates aor-
with the heart beating and unloaded. It is to tic clamping, the heart is perfused and,
be remarked that, if the indication for opera- theoretically, optimally protected. Thus,
tion is acute evolving myocardial infarction, the aortic cross-clamp time, tradition-
grafts should be performed to reperfuse the ally correlated with myocardial damage,
200 Ischemia-Reperfusion Injury in Cardiac Surgery

relates to a somewhat different condi- hemorrhage 5 days after surgery) and these
tion if compared to standard data compare favorably with surgical series pre-
cardioplegic arrest; viously reported in the literature,2,17 especially
3. this reperfusion period is ideal (also because, in the latter, surgical candidates were
from a technical point of view) to con- not primarily selected on the basis of the
struct the proximal anastomoses of the extension of the ischemic area.
vein grafts with the heart in a flaccid On the contrary, mortality was 50% (4/8)
and empty state, and avoiding side-bit- in patients with cardiogenic shock. This out-
ing clamps on the ascending aorta. In come is strongly affected by preoperative con-
fact we now perform such anastomoses ditions and confirms the dramatic advantages
during a single aortic cross-clamp pe- of surgical treatment compared with conven-
riod for all coronary operations; tional measures. All patients (2/8) requiring
4. finally, the internal mammary artery cardiopulmonary resuscitation before opera-
may more easily be used to revascularize tion died.
the left anterior descending coronary ar- Intraaortic balloon counterpulsation is very
tery (LAD) in selected cases. This op- useful in this subset of patients. We have placed
tion avoids the necessity to place a vein a balloon after operation in 20% of patients
graft on the LAD to assure controlled with acute myocardial infarction and in all
reperfusion. The construction of two patients with cardiogenic shock. Preoperative
separate bypass grafts on the LAD has balloon counterpulsation has been necessary
been advocated,29 but it is probably less in 75% of cases with cardiogenic shock. More-
recommendable being time-consum- over, 25% of patients with cardiogenic shock
ing. In this regard, the choice to use the required preoperative cardiopulmonary resus-
internal mammary artery must obvi- citation. Similarly, inotropic support before
ously be carefully evaluated and the operation was needed in 20% of patients with
pedicle should be dissected as expedi- acute myocardial infarction and in all patients
tiously as possible, eventually after the with cardiogenic shock. Postoperatively, most
institution of cardiopulmonary bypass, cases required intravenous catecholamines
preferably by a senior surgeon. In our (80% of patients with acute myocardial inf-
experience, a judicious use of the inter- arction and in all patients with cardiogenic
nal thoracic artery has been possible in shock). Occasionally we performed a delayed
about 50% of patients with acute myo- sternal closure in patients showing hemody-
cardial infarction. It has not been used namic deterioration while attempting to close
in case of cardiogenic shock because the sternal edges.30
long-term prognosis is not the critical Peak CK-MB enzyme serum levels in pa-
issue in this subset of patients. tients operated for acute evolving infarction
averaged about 70 mg/L and were recorded at
Personal Experience arrival in the intensive care unit in the major-
and Conclusive Considerations ity of cases. Left ventricular function appears
to improve after coronary bypass. Although
As we have reported,19 results can be di-
our data are not statistically significant, mean
vided into two groups of patients reflecting
left ventricular ejection fraction markedly in-
the two different indications for operation. In
creased in patients with cardiogenic shock,
particular, results are much more disappoint-
especially during the first months following
ing in the presence of cardiogenic shock, as it
the operation (22% preoperatively; 25% at
is with conventional medical therapy. In our
discharge from the hospital; 35% at 6-months
preliminary experience on 23 patients, in-hos-
follow-up), while only mild variations were
pital mortality was very low in patients who
encountered, in case surgery was performed
underwent operation for acute evolving
for extensive acute myocardial infarction (40%
myocardial infarction (1/15 due to cerebral
Acute Myocardial Infarction and Cardiogenic Shock 201

preoperatively; 45% at hospital discharge and 5. O’Keefe JH, Rutherford BD, McConahay DR
follow-up). et al. Early and late results of coronary
angioplasty without antecedent thrombolytic
Finally, from an economic standpoint, this therapy for acute myocardial infarction. Am
technique may appear expensive. It is certainly J Cardiol 1989; 64:1221-1230.
a subset of critically ill patients compared to 6. Stack RS, Califf RM, Hinohara T et al. Sur-
elective coronary surgical candidates, but it vival and cardiac event rates in the first year
after emergency coronary angioplasty for acute
must be emphasized that the acute ischemic myocardial infarction. J Am Coll Cardiol
event and the underlying coronary artery dis- 1988; 11:1141-1149.
ease are treated during a single hospitalization, 7. Salem DN, Desnoyers MR, Berman AD et
thus reducing costs. In addition, this particu- al. Coronary angioplasty and thrombolysis for
lar population is more likely to require a pro- acute myocardial infarction: is two a crowd?
Am J Med 1989; 86:259-261.
longed hospitalization and a higher probabil- 8. Ryan TJ. Revascularization for acute myocar-
ity to develop congestive heart failure on the dial infarction. Strategies in need of revision.
medium-to-long term, if patients are treated Circulation 1990; 82(suppl. II):II-110-II-116.
in a more conventional manner. 9. Koshal A, Beanlands DS, Davies RA et al.
Urgent surgical reperfusion in acute evolving
In conclusion, we are convinced that emer- myocardial infarction. Circulation 1988;
gency surgical therapy can play a substantial 78(suppl. I):I-171-I-178.
role in improving results in patients with acute 10. Flameng W, Sergeant P, Vanhaecke J et al.
evolving myocardial infarction and cardiogenic Emergency coronary bypass grafting for evolv-
shock. If coronary bypass is performed within ing myocardial infarction. Effects on infarct
size and left ventricular function. J Thorac
6 hours from the onset of ischemia, we feel Cardiovasc Surg 1987; 94:124-131.
that the majority of patients with extensive 11. Barner HB, Lea JW IV, Naunheim KS et al.
ongoing infarction, i.e., patients at risk for Emergency coronary bypass not associated
secondary cardiogenic shock or, less dramati- with preoperative cardiogenic shock in failed
angioplasty, after thrombolysis, and for acute
cally, for the development of congestive heart myocardial infarction. Circulation 1989;
failure on the medium-long term, have a bet- 79(suppl. I):I-152-I-159.
ter prognosis. Myocardial protective strategies 12. Goldman BS, Weisel RD. Surgical reperfusion
are a crucial issue in performing such surgical of acute myocardial ischemia: a clinical re-
procedures and, in fact, emergency coronary view. J Cardiac Surg 1986; 1:167-199.
13. Jones EL. Surgical revascularization during
bypass grafting in this setting may well be con- acute evolving myocardial infarction. Circu-
sidered a “myocardial protective operation.” lation 1987; 76(suppl. III):III-146-III-148.
14. Athanasuleas CL, Geer DA, Arciniegas JG et
References al. A reappraisal of surgical intervention for
acute myocardial infarction. J Thorac
1. Berg R, Kendall RW, Duvoisin GE et al.
Cardiovasc Surg 1987; 93:405-414.
Acute myocardial infarction. A surgical emer-
15. Laks H, Rosenkranz E, Buckberg G. Surgical
gency. J Thorac Cardiovasc Surg 1975;
treatment of cardiogenic shock after myocar-
70:432-439.
dial infarction. Circulation 1986; 74(suppl.
2. De Wood MA, Spores J, Notske RN et al. III):III-11-III 16.
Medical and surgical management of myocar- 16. Bolooki H. Emergency cardiac procedures in
dial infarction. Am J Cardiol 1979; 44:1356- patients in cardiogenic shock due to compli-
1364. cations of coronary artery disease. Circulation
3. Berg R Jr, Selinger SL, Leonard JJ et al. Im- 1989; 79(suppl.I):I-1137-I-148.
mediate coronary artery bypass for acute 17. Phillips SJ, Kongtahworn C, Skinner JR et
evolving myocardial infarction. J Thorac al. Emergency coronary artery reperfusion: A
Cardiovasc Surg 1981; 81:493. choice therapy for evolving myocardial inf-
4. Yusuf S, Sleight P, Held P et al. Routine arction. Results in 339 patients. J Thorac
medical management of acute myocardial in- Cardiovasc Surg 1983; 86:679-688.
farction. Lessons from overviews of recent 18. Donatelli F, Benussi S, Triggiani M et al.
randomized controlled trials. Circulation Emergency surgical revascularization in exten-
1990; 82(suppl. II):II-117-II-134. sive acute myocardial infarction: a prospec-
tive protocol. Circulation 1996; 94(suppl.
I):I-476.
202 Ischemia-Reperfusion Injury in Cardiac Surgery

19. Donatelli F, Benussi S, Triggiani M et al. 25. Diehl JT, Eichhorn EJ, Konstam MA et al.
Surgical treatment for life-threatening acute Efficacy of retrograde coronary sinus car-
myocardial infarction: a prospective protocol. dioplegia in patients undergoing myocardial
Eur J Cardio-Thorac Surg 1997; 11:228-233. revascularization: A prospective randomized
20. Rosenkranz ER, Buckberg GD. Myocardial trial. Ann Thorac Surg 1988; 45:595-602.
protection during surgical coronary reper- 26. Eichhorn EJ, Diehl JT, Konstam MA et al.
fusion. J Am Coll Cardiol 1983; 1:1235- Protective effects of retrograde compared with
1246. antegrade cardioplegia on right ventricular
21. Rosenkranz ER, Okamoto F, Buckberg GD systolic and diastolic function during coro-
et al. Safety of prolonged aortic clamping nary bypass surgery. Circulation 1989; 79:
with blood cardioplegia. III. Aspartate enrich- 1271-1281.
ment of glutamate-blood cardioplegian energy 27. Fiore AC, Naunheim KS, Kaiser GC et al.
depleted hearts after ischemic and reperfusion Coronary sinus versus aortic root perfusion
injury. J Thorac Cardiovasc Surg 1986; with blood cardioplegia in elective myocar-
91:428-435. dial revascularization. Ann Thorac Surg 1989;
22. Allen BS, Rosenkranz E, Buckberg G et al. 47:684-688.
Studies on prolonged acute regional ischemia. 28. Haan C, Lazar HL, Bernard S et al. Superi-
Myocardial infarction with left ventricular ority of retrograde cardioplegia after acute
power failure: a medical/surgical emergency coronary occlusion. Ann Thorac Surg 1991;
requiring urgent revascularization with maxi- 51:408-412.
mal protection of remote muscle. J Thorac 29. Myocardial management during cardiac sur-
Cardiovasc Surg 1989; 98:691-703. gery with cardiopulmonary bypass. In: Kirklin
23. Gundry GR, Kirsh MM. A comparison of JW, Barratt-Boyes BG, eds. Cardiac surgery,
retrograde cardioplegia versus antegrade car- 2nd edition, New York: Churchill Livingstone
dioplegia in the presence of coronary artery 1993:155-156.
obstruction. Ann Thorac Surg 1984; 38: 30. Donatelli F, Triggiani M, Benussi S et al.
124-127. Advantages of delayed sternal closure in car-
24. Guiraudon GM, Campbell CS, McLellan DG diac-compromised adult patients. J Cardiac
et al. Retrograde coronary sinus versus aortic Surg 1995; 10:632-636.
root perfusion with cold cardioplegia: Ran-
domized study of enzyme levels in 40 pa-
tients. Circulation 1986; 74(suppl. III):III-
105-III-115.
CHAPTER 18

Protection Strategies in Reoperations


Fumiyuki Okamoto and Keisuke Sakai

A
s the life-expectancy in patients surgery. There may be, however, danger in ster-
undergoing cardiac surgery nal re-entry, e.g., injury to substernally adher-
has increased, the frequency of cardiac ent structures such as the free wall of the right
reoperations for acquired heart disease has ventricle, anteriorly extending aorta, patent
correspndingly increased. Reoperation is vein grafts overlying heart, in situ internal tho-
accompanied by difficult problems deriving racic artery (ITA) grafts crossing the aorta or
from substernal and intrapericardial tissue ad- pulmonary artery, and so forth.1,4 Often it is
hesions, severely compromised heart function helpful to assess the substernal anatomy by pre-
with advanced cardiac lesions, and older and operative CT scan. In addition, preoperative
sicker patients with other organ dysfunc- coronary angiograms—including vein, ITA
tions.1,2 These patients often require pro- and other arterial grafts—are a prerequisite
longed cardiopulmonary bypass and aortic regardless of whether patent or occluded in
cross clamping. In such circumstances, we order not only to plan the newly constructed
must maximally preserve cardiac function by coronary bypass, but to avoid unnecessary
means of myocardial protection. injury against these conduits.1,4 If a graft is
The majority of adult cardiac reoperations occluded, for instance, there is no need to
involve coronary bypass; for example, in our make special efforts to avoid injuring it. But if
hospital over the past several years 70.1-77.0% patent, compression trauma or direct injury
are coronary cases. Valve surgery and thoracic during dissection may cause intraoperative
aortic aneurysms are the second and third most myocardial infarction before cardiopulmonary
common indications for reoperation, respec- bypass is established either by interruption of
tively. Myocardial protection techniques dif- blood supply or embolization of easily detach-
fer more or less based upon the problems in- able atheromatous debris.1-4 Therefore, exten-
herent in the underlying pathology, e.g., the sive intrapericardial dissection, especially
embolization of atherosclerotic debris from around bypass conduits. should be delayed
previously placed vein grafts, annular defects until cardiopulmonary bypass is established or
caused by infectious endocarditis after pros- until cardioplegic arrest occurs.3-5
thetic valve replacement, and severe aortic re- Sometimes arterial and venous access for
gurgitation secondary to proximal extension cardiopulmonary bypass should be attained
of aortic dissection. 1-5 In this chapter, prior to sternal re-entry, usually by femoral ex-
myoprotective strategies will be described in posure (artery and vein)1,2,5 or axillary arterial
accordance with these three major categories. approach.4 Once sternal re-entry with subse-
quent prebypass dissection confined to aorta
Coronary Bypass Reoperations and right atrium is safely established, the con-
The median sternotomy is the standard ventional cannulation technique can be un-
access in coronary reoperation as with primary dertaken and femoral or axillary access can be

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


204 Ischemia-Reperfusion Injury in Cardiac Surgery

discontinued. If sternal re-entry is not easily embolization is expected, functioning grafts


accomplished, cardiopulmonary bypass via should be divided1-3,5 and new anastomoses
femoro-femoral access or axillary approach constructed. Once functioning, but severely
should be established to decompress the heart, diseased vein grafts are transected, the com-
thereby keeping cardiac structures, including bined antegrade and retrograde cardioplegic
patent graft conduits, away from the ster- infusion may be indicated as in routine car-
num.2,4,5 This substantially enhances the safety diac surgery without risk of embolization.
of sternal re-entry with an oscillating saw and Another option is continuous retrograde cold
the subsequent intrapericardial dissection. In blood perfusion following aortic cross clamp-
addition, hemodynamic stability can be main- ing, which gives rise to cardiac quiescence ad-
tained by cardiopulmonary bypass, even if se- equate to ensure accurate intrapericardial dis-
rious injury to substernal cardiac structures oc- section.1-5
curs.4 On some occasions, however, dissection Functioning arterial grafts, such as ITA or
around heart should be limited to minmize gastroepiploic artery (GEA), are problematic2,3
the risk of intravascular detachment of athero- since they tend to reduce the myoprotective
matous debrises in diseased, but still patent effect and to hamper the operative procedure
vein grafts, even though cardiopulmonary by- by continuously draining noncardioplegic
pass is established. Moreover cardiac quies- blood into the myocardium, delaying
cence induced by cardioplegia facilitates intra- cardioplegic arrest and early return of electro-
pericardial dissection.1-5 The technical aspects mechanical activity. The careful dissection of
of sternal re-entry and a more detailed descrip- pedicled arterial grafts and temporary place-
tion of cannulation strategies for cardiopul- ment of occlusion clamps to interrupt
monary bypass are described elsewhere.4 noncardioplegic blood flow into the myocar-
Systemic hypothermia below 25˚C used to dium overcomes these problems.1,3 In such
be an indispensable adjunct in cardiac surgery, cases, retrograde infusion is extremely useful
but current advancements in myocardial pro- to provide relatively uniform distribution of
tection have rendered it redundant.3,7,21 In rou- cardioplegic solution and to optimize myocar-
tine cardiac operations, we keep the tempera- dial protection, particularly when these arte-
ture at subnormothermic levels, around 34˚C; rial conduits supply a considerable amount of
this can be applied to coronary reoperations blood flow to the myocardium.3,4 Coronary
as well. However, whenever a prolonged aor- anastomoses are usually constructed under in-
tic cross clamp time is expected, the systemic duced cardioplegic arrest followed by proxi-
temperature should be lowered to increase the mal anastomoses performed while cold retro-
margin of safety. For the purpose of placing grade noncardioplegic blood is continuously
ice slush, complete isolation of the heart from infused.3,24
pericardial adhesions is ideal. Yet more and If pedicled arterial grafts are difficult to
more institutions, including ours, have aban- dissect, aortic cross-clamping and cardiople-
doned topical hypothermia as an adjunct to gia often are effective so the pedicles can be
myoprotection.6,7 freed-up under relative cardiac quiescence,
Cardioplegic delivery, either antegrade via even though electromechanical activity may
the ascending aorta or retrograde via the right resume. With functioning arterial grafts, hy-
atrium, or both, should be accomplished af- pothermic systemic circulatory arrest8 that
ter cardiopulmonary bypass. Recently com- does not require clamping arterial pedicles may
bined antegrade and retrograde cardioplegia be indiccated, but this considerably prolongs
has become the method of choice,2,3,5,7 but is cardiopulmonary bypass and increases the risk
not always indicated in re-do coronary sur- of cerebral ischemia. The distal coronary anas-
gery since antegrade cardioplegia sometimes tomoses also can be constructed under induced
causes embolization of atheromatous debris ventricular fibrillation as conducted in the
from severely diseased vein grafts and possibly past,9 but this tends to underperfuse the
from advanced native coronary lesions.4 If subendocardium with subsequent myocardial
Protection Strategies in Reoperations 205

necrosis.7,10 Intermittent aortic cross-clamp- elevated.2,17,20 If the functional class of patients


ing for each distal anastomosis, with ischemic is NYHA-IV, the risk of reoperation is higher
arrest or antegrade cardioplegic infusion via than in NYHA-I to -III patints in whom
patent native coronary arteries, is another op- operative mortality and morbidity are com-
tion although the pedicle has to be clamped parable to that seen in primary surgery.2 There-
temporarily and the proximal anastomoses fore, it is important that reoperative valve sur-
constructed under tangential aortic clamping.3 gery be undertaken before the patient’s
Neither technique, however, fulfills the hemodynamic status deteriorates.
myoprotection obtained in primary surgery. Infectious endocarditis is another serious
Right or left thoracotomy11 thoracotomy4,12 problem in re-do valve operations18 since it
are sometimes safer approaches for re-enter- produces impaired left ventricular function,
ing the mediastinum in coronary reoperations, e.g., perivalvular leakages cause aortic or mitral
but they are suited only to the right coronary regurgitation, and destroys periannular
system or mainly to posterolateral portions of anatomy with the potential of cerebrovascu-
the left ventricle (sometimes to left anterior lar embolism.19,20 In these cases, the meticu-
descending coronary artery-LAD). In these lous surgical repair of periannular defects along
cases, either ventricular fibrillation, topical with the careful re-reimplantation of a new
cardiac hypothermia or intermittent valve are needed, often requiring long cardio-
interruption of coronary blood flow by occlu- pulmonary bypass and aortic cross-clamp
sion snare is needed while the anastomoses are times. For this reason, innovative myo-
constructed. Nonetheless, these techniques are protection is required.
not as effective as conventional myocardial Whenever difficulties in sternal re-entry are
protection. urgical benefits have to be com- expected, prebypass exposures of femoral
promised considerably by technical feasibility. artery and vein are recommended to readily
On the other hand, minimally invasive car- initiate cardiopulmonary bypass and to
diac surgery likely can be applied to coronary decompress the heart by inferior vena caval
reoperations, for example, a left ITA to LAD drainage; this facilitates meticulous intra-
anastomosis through a limited anterior thora- pericardial dissection.2 The substernally adher-
cotomy or a right GEA to distal right coro- ent failed right ventricle may be easily injured
nary artery through a divided xyphoid and because it tends to dilate and its wall thins sec-
small upper abdominal incision.13,14,32 ondary to left ventricular failure, often neces-
sitating complicated repair. Therefore, the pre-
Valve Reoperations operative chest x-ray2 and CT are helpful in
Although recent advances in artificial valve avoiding these dangers. Even if sternal re-entry
design have led to excellent long-term clinical is accomplished without installing femoro-
results, there are still reasons for reoperation: femoral bypass, dissection to establish cardiop-
structural defects of mechanical valves, degen- ulmonary bypass should sometimes be con-
erative alterations of bioprosthetic valves (por- fined to limited areas of the ascending aorta
cine aortic or bovine pericardium) or hemo- and right atrium—just enough to permit
dynamic impairments of arising from cannulation. Further intrapericardial dissec-
perivalvular thrombi or fibrotic tissue prolif- tion can be ndertaken after the heart is
eration.2,15,16,20 In these circumstances, the decompressed completely during cardiopul-
patient may have adapted with left and/or right monary bypass. Because topical hypothermia
ventricular distention secondary to pulmonary has decreased in popularity in recent years,6,7
hypertension which can interfere with sternal isolation of the heart from the pericardium is
re-entr y. 2 In patients who require not necessarily needed to optimize myo-
reoperative valve surgery repair is sometimes protection. In any case, this disection is asso-
deferred until the hemodynamic status alread ciated with persistent intrapericardial bleed-
is deteriorated; accordingly the incidence of ing, particularly when the patients are
reoperative mortality and morbidity is anticoagulated preoperatively. Nevertheless,
206 Ischemia-Reperfusion Injury in Cardiac Surgery

care must be taken during deairing when the annular suture placements et cetera, the retro-
aorta and/or left atrium are closed after valve grade perfusion of cold plain blood can be con-
replacement. The intraoperative trans- tinuous, thereby virtually offsetting the
esophageal echocardiogram is useful to detect ischemic insult despite prolonged aortic cross-
residual air frequently existing in the left clamping. Most surgical procedures (e.g.,
atrium, which otherwise often enters the right suture placement to a prosthetic valve ring,
coronary orifice, leading to postoperative right refixation of a valve by securing sutures,
ventricular dysfunction and resultant low car- aortorrhaphy) can be accomplished without
diac output despite appropriate myocardial creating a further ischemic burden in func-
protection.2 tionally impaired hearts. As a consequence, in
The combined use of antegrade and retro- most aortic valve reoperations, the strict use
grade cardioplegia is the method of choice to of combined antegrade and retrograde
protect the heart in re-do valve surgery because cardioplegic perfusion may no longer be
of the uniform distribution of cardioplegic so- absolutely necessary, and the simultaneous
lution.2,21 Although cardiac quiescence can be antegrade to right coronary combined with
more readly achieved by antegrade than retro- retrograde cardioplegia would be a better
grade cardioplegia, aortic regurgitation delays choice to reduce technical complexity.
cardioplegic arrest considerably, and antegrade In mitral re-do operations, minimal
cardioplegia should be used only at the outset intrapericardial dissections mainly around the
with subsequent immediate retrograde right atrium (i.e., trans-septal approach) or
cardioplegic perfusion. 2,3,21 The direct right-sided left atrium allow suffcient access
intracoronary infusion of cardioplegia follow- to the mitral valve, unless a more complicated
ing aortotomy, either as cardioplegic induc- procedure like “Cox-maze operation” is
tion or subsequent to initial infusion of cold required or visualization of mitral valve is
retrograde cardioplegia, may be another alter- insufficient.25,26 As performed in other cardiac
native to induce immediate cardioplegic arrest re-dos, the initial cardiac quiescence can be
in aortic valve reoperations with severe aortic provided by antegrade cardioplegic perfusion
regurgitation.3,21 But care must be taken not whenever possible, and then followed by ret-
to inadvertently injure the coronary orifices, rograde perfusion.2,3,24 If the operative proce-
particularly when the aorta is severely calcified. dure requires a brief period of aortic cross-
Once cardiac quiescence is achieved in aor- clamping—as in a single mitral valve re-re-
tic valve re-do surgeries, we prefer to utilize placement—, repeated retrograde cardiople-
simultaneous selective antegrade to right coro- gia may be adequate to protect the heart. If
nary artery and retrograde cardioplegic perfu- reoperation requires a long period of aortic
sion to ensure the uniform distribution of car- cross-clamping, the combined use of antegrade
dioplegia solution. Since one of the biggest and retrograde cardioplegia is recommended,
disadvantages of retrograde perfusion is inad- particularly in functionally-compromised
equate nutritional flow to the right ventricle hearts associated with left ventricular dysfunc-
with poor myoprotection despite internal tion. In this case, however, air must not be
cooling through the venovenous shunt,22-24 the pushed inadvertently into coronary arteries
additive perfusion of retrograde cardioplegia since an open left atrium and empty left ven-
simultaneously directed to the right coronary tricle usually accompanies an air-filled aorta.
artery can yield better distribution of Regardless of the method of cardioplegia, it
cardioplegic solution. This can be achieved may be useful to perfuse the myocardium ret-
easily by connecting the intracoronary cannula rogradely with cold noncardioplegic blood
to the side-arm of the retrograde perfusion line throughout surgery where technical precision
as advocated by Buckberg et al.3,24 Unless is not strictly required, e.g., in valve fixation
extraordinary surgical precision is required as with securing sutures, closure of an atrial sep-
in repair of periannular defects, removal of a tum or left atriorrhaphy.3,21,24 This may also
previously replaced malfunctioning valve, be helpful in flushing back any intracoronary
Protection Strategies in Reoperations 207

air or emboli toward the coronary orifices in impairment of the left ventricle making it less
the aorta; this prevents postoperative impair- tolerant to ischemic insult. Although antegrade
ment of cardiac function.2,3,21,24,33 In excep- cardioplegic induction via the aortic root pro-
tional cases of re-do mitral valve operations, duces prompt cardiac arrest despite aortic
as with multiple previous surgeries, a right an- regurgitation, the left ventricle has to be vented
terior thoracotomy with femoro-femoral immediately to avoid overdistension, and ret-
bypass can be employed for access to the mi- rograde cardioplegia should be given to con-
tral valve in order to avoid massive bleeding fer more efficient myocardial protection.
from incision by means of a median sterno- Alternatively, the direct intracoronary infusion
tomy.2,27 Since the conventional myoprotective of cardioplegia is a delivery option after
strategy of combined antegrade and retrograde aortotomy to ensure reliable myoprotection.
cardioplegia is difficult to conduct in this In these cases, where left ventricular hyper-
approach, systemic hypothermia to 20˚C is trophy exists, the total amount of cardiople-
concomitantly utilized to protect the heart. gia should be increased3,21 as much as 1.5 times
with anticipation of more complete distribu-
Thoracic Aortic Aneurysm tion of cardioplegia.
Reoperations Reoperations of thoracic aortic aneurysms
require extremely difficult and time-consum-
Re-do thoracic aortic aneurysm surgery
ing reconstruction which should be performed
involving the aortic root is the most difficult
in a relatively bloodless field. We, therefore,
type of reoperative cardiac surgery because
preferentially utilize retrograde, continuous
large, adherent ascending aortic aneurysms
noncardioplegic blood perfusions to reduce
sometimes make sternal re-entry extremely
ischemia duration whenever. Absolutely
dangerous and distort adjacent cardiac struc-
bloodless operative fields are mandatory only
tures including aortic valves and coronary ar-
in aortic root reconstructions which include
teries in unexpected directions.2,28 In order to
coronary orifices. As with aortic valve
ensure safer sternal re-entry, exposure of the
reoperations, repeated cardioplegic infusions
femoral artery and vein for initiation of retro-
subsequent to initial induction can be substi-
grade cardiopulmonary bypass is routine
tuted by simultaneous selective antegrade (to
although there is the risk of fatal cerebral or
right coronary artery) and retrograde perfu-
even myocardial embolizations from debris
sion even while noncardioplegic blood is being
dislodged from the descending aorta and/or
given.3,24 This avoids the cumbersome place-
proximal aortic dissection.29 Therefore, care-
ment of intracoronary cannulae at each
ful monitoring by a transesophageal echo is
cardioplegic interval and prevents traumatic
extremely useful to detect abnormalities in the
injury to the coronary ostia. Additionally, any
descending aorta and if such devastating com-
debris and/or air bubbles can be evacuated
plications are likely to happen, antegrade car-
from coronary arteries retrogradely.2,3,24,33
diopulmonary bypass via the ascending aorta
Almost all forms of myoprotection used in
or arch should be taken into account. Deep
re-do coronary and valve surgery are applicable
hypothermic systemic arrest is almost always
in reoperations of thoracic aortic aneurysms.
required to protect multiple organs, particu-
larly brain as well as heart. The various
modalities for brain protection have inherent Summary
advantages and disadvantages, and the tech- The current protection strategies for
nical aspects, along with cannulation strate- reoperations have been reviewed with regard
gies, are described in detail elsewhere.30,31 to the three major indications for adult car-
diac surgery. Although the fundamental pre-
Often ascending aortic aneurysms are cepts of myocardial protection in redo surgery
associated with aortic regurgitation because of are almost identical to those in primary sur-
annular dilatation or direct involvement of dis- gery, there are numerous factors which com-
section, 2 which then leads to functional plicate re-do operations. The complexities in
208 Ischemia-Reperfusion Injury in Cardiac Surgery

any individual case cannot be overcome by any 14. Benetti F, Mariani MA, Sani G et al. Video-
single, universal method. Comprehensive assisted minimally invasive coronary opera-
tions without cardiopulmonary bypass: A
integration of myoprotective techniques is multicenter study. J Thorac Cardiovasc Surg
required in various re-do surgical situations. 1996; 112(6):1478-1484.
15. Cohn LH, Koster JK Jr, Van de Vanter S et
References al. The in-hospital risk of rereplacement of
dysfunctional mitral and aortic valves. Cir-
1. Loop FD. The value and conduct of
culation 1982; 66(Suppl I):I-153-I-156.
reoperations for coronary atherosclerosis.
16. Bortolotti U, Milano A, Mossuto E et al.
Semin Thorac Cardiovasc Surg 1994; 6(2):
Early and late outcome after reoperation for
116-119.
prosthetic valve dysfunction: Analysis of 549
2. Cohn LH. Myocardial protection for patients during a 26-year period. J Heart
reoperative cardiac surgery in acquired heart Valve Dis 1994; 3(1):81-87.
disease. Semin Thorac Cardiovasc Surg 1993; 17. Blackstone EH, Kirklin JW. Death and other
5(2):162-167. time-related events after valve replacement.
3. Buckberg GD, Allen BS. Myocardial protec- Circulation 1985; 72:753-767.
tion management during adult cardiac opera- 18. Larbalestier RI, Kinchla NM, Aranki SF et
tions. In: Baue A, Geha A, Hammond G et al. Acute Bacterial endocarditis. Optimizing
al. eds. Glenn’s Thoracic and Cardiovascular surgical results. Circulation 1992; 86(Suppl
Surgery. Stamford, CT: Appleton & Lange II):II-68-II-74.
1995:1653-1687. 19. Baumgartner WA, Miller DC, Reiz BA et al.
4. Lytle BW. Coronary reoperations. In: Surgical treatment of prosthetic valve en-
Edmunds LH Jr., ed. Cardiac Surgery in the docarditis. Ann Thorac Surg 1983; 35:
Adult. New York: McGraw-Hill Health Pro- 87-104.
fessions Division, 1997:573-596. 20. Oakley CM. Investigations before reoperation
5. Savage EB, Cohn LH. ‘No touch’ dissection, for acquired heart disease. In: Stark J, Pacifico
antegrade-retrograde blood cardioplegia, and AD, eds. Reoperations in Cardiac Surgery.
single aortic cross-clamp significantly reduce Berlin Heidelberg New York: Springer-Verlag,
operative mortality of reoperative CABG. 1989:17-37.
Circulation 1994; 90(Part 2):II-140-II-143. 21. Allen BS, Murcia-Evans D, Hartz RS. Inte-
6. Allen BS, Buckberg GD, Rozenkranz ER et grated cardioplegia allows complex valve re-
al. Topical cardiac hypothermia in patients pairs in all patients. Ann Thorac Surg 1996;
with coronary disease: An unnecessary adjunct 62:23-30.
to cardioplegic protection and cause of pul- 22. Gates R, Laks H, Drinkwater D et al. Gross
monary morbidity. J Thorac Cardiovasc Surg and microvascular distribution of retrograde
1992; 104:626-631. cardioplegia in explanted human hearts. Ann
7. Buckberg GD. Myocardial Protection: An Thorac Surg 1993; 56:410-417.
overview. Semin Thorac Cardiovasc Surg 23. Allen B, Winkelmann J, Hanafy H et al.
1993; 5(2):98-106. Retrograde cardioplegia does not perfuse the
8. Michielon G, Doty DB. Hypothermic circu- right ventricle. J Thorac Cardiovasc Surg
latory arrest for cardiac valve replacement 1995; 109:1116-1126.
reoperations. Ann Thorac Surg 1994; 57: 24. Buckberg GD, Beyersdorf F, Allen BS et al.
1281-1283. Integrated myocardial management: Back-
9. Senning A. Ventricular fibrillation during ground and initial application. J Card Surg
extracorporeal circulation. Acta Chir Scand 1995; 10:68-89.
1952; 171(Suppl):1-79. 25. Cox JL, Schuessler RB, D’agostino HJ et al.
10. Najafi H, Henson D, Dye WS. Left ventricu- The surgical treatment of atrial fibrillation.
lar hemorrhagic necrosis. Ann Thorac Surg III. Development of a definitive surgical pro-
1969; 7:550-561. cedure. J Thorac Cardiovasc Surg1991;
11. Uppal R, Wolfe WG, Lowe JE et al. Right 101(4):569-583.
thoracotomy for reoperative right coronary 26. Duran CG. Reoperations on the mitral and
artery bypass procedures. Ann Thorac Surg tricuspid valves. In: Stark J, Pacifico AD, eds.
1994; 57:123-125. Reoperations in Cardiac Surgery. Berlin
12. Ungerleider RM, Mills NL, Wechsler AS. Left Heidelberg New York: Springer-Verlag,
thoracotomy for reoperative coronary bypass 1989:325-349.
procedures. Ann Thorac Surg 1985; 40: 27. Cohn LH, Peigh PS, Sell J et al. Right tho-
11-15. racotomy, femoro-femoral bypass, and deep
13. Benetti FJ, Ballester C, Sani G et al. Video hypothermia for re-replacement of the mitral
assisted coronary bypass surgery. J Card Surg valve. Ann Thorac Surg 1989; 39:53-55.
1995; 10:620-625.
Protection Strategies in Reoperations 209

28. Crawford ES, Crawford JL, Safi HJ et al. 31. Svensson LG, Crawford ES, Hess KR et al.
Redo operations for recurrent aneurysmal dis- Deep hypothermia with circulatory arrest: De-
ease of the ascending aorta and transverse terminants of stroke and early mortality in
aortic arch. Ann Thorac Surg 1985; 40: 656 patients. J Thorac Cardiovasc Surg 1993;
439-455. 106(1):19-31.
29. Wareing TH, Davila-Roman VG, Barzilai B 32. Watanabe G, Misaki T, Kotoh K et al. Bi-
et al. Management of the severely atheroscle- lateral minimally invasive direct artery bypass
rotic ascending aorta during cardiac opera- grafting with the use of two arterial grafts. J
tions. J Thorac Cardiovasc Surg 1992; Thorac Cardiovasc Surg 1997; 113(5):
103(3):453-462. 949-951.
30. Crawford ES, Saleh SA. Transverse aortic arch 33. Sandhu AA, Spotnitz HM, Dickstein ML et
aneurysm: Improved results of treatment al. Cardiopulmonary bypass, myocardial man-
employing new modifications of aortic recon- agement, and support techniques. Retrograde
struction and hypothermic cerebral circulatory cardioplegia preserves myocardial function
arrest. Ann Surg 1981; 194:180-188. after induced coronary air embolism. J Thorac
Cardiovasc Surg 1997; 113(5):917-922.
CHAPTER 19

Protection Strategies
for Heart Transplantation
Juergen Martin, Armin Geiger and Friedhelm Beyersdorf

S
afe procurement and effective solution can not be washed out and periodic
preservation are fundamental features in replenishment of cardioplegia is not necessary.
heart transplantation. The preservation During storage the heart is surrounded by
technique might influence the rate of early ice-cold solution at a constant myocardial
graft failure as well as the incidence of graft temperature of 0-4˚C. This allows extended
vasculopathy.1,2 periods of storage without extracorporal
Data from the Registry of the International circulation.
Society for Heart and Lung Transplantation During heart transplantation 4 Phases of
demonstrate, that the early mortality rate after myocardial protection can be distinguished:9
heart transplantation remains 8-10% and 40% Phase 1: Preharvesting phase
of these deaths are caused by nonspecific graft Phase 2: Cardioplegic induction phase
failure.3 Phase 3: Organ storage
One important fact related to early graft Phase 4: Reperfusion phase
failure may be that brain death is associated
with hemodynamic deterioration, so that Preharvesting Phase
donor hearts often require inotropic support Brain death causes myocardial energy and
and are of questionable value.4-6 These inju- substrate depletion with a metabolic profile
ries are greater in the right ventricle and may similar to ischemically damaged hearts.10 Data
contribute to early right ventricular failure after confirm depletion of ATP, CP, glycogen as well
transplantation.7 as of tissue glutamate, a precursor of the Krebs
Furthermore, heart excision and preserva- cycle intermediate alpha-ketoglutarate.11 Fur-
tion techniques are thought to play a role in thermore, dysfunction of the hypothalamic-
myocardial damage.8 Research efforts are in pituitary axis results in acute hypothyroidism,
progress to develop preservation methods that adrenal insufficiency, and diabetes insipidus.
would decrease myocardial injury both dur- These factors may contribute to early nonspe-
ing the period of preservation and during the cific graft failure.
period of reperfusion. Due to the universal shortage of donor
There are some special topics of donor organs, optimal management of the brain dead
heart preservation in comparison with protec- patient is essential before organ harvest.
tion techniques in other open heart proce-
dures. One topic is that the donor heart is a
completely isolated organ without any collat-
eral blood flow. Therefore, the preservation

Ischemia-Reperfusion Injury in Cardiac Surgery, edited by Friedhelm Beyersdorf. ©2001 Eurekah.com.


Protection Strategies for Heart Transplantation 211

Pretreatment with Substances of adenosine can substitute for the 5 min of


that Limit Ischemic ischemia that are required to precondition the
and Reperfusion Damage isolated blood perfused rabbit heart against
necrosis. The effects of extracellular adenos-
ine are mediated predominantly via membrane
Allopurinol
receptors (A1 and A2 receptors) in the myocytes
Controversial results exist concerning the
and coronary vasculature. The mediated effects
effect of the xanthine oxidase inhibitor allopu-
of these receptors include: the stimulation of
rinol. Some experiments have shown a reduc-
glycolysis,22 delaying of ischemic contracture23
tion in irreversible myocardial injury follow-
and, reduction of neutrophil activation.24
ing pretreatment with allopurinol, but others
Support for preconditioning in humans
have failed to show an effect.12-14 A possible
comes from several sources: studies showing
explanation for this discrepancy could be that
increased tolerance to repetitive balloon infla-
the concentration of xanthine oxidase varies
tions during angioplasty, some studies suggest-
widely between species and has been reported
ing that preinfarction angina confers an early
as being undetectable in both the porcine and
beneficial effect and studies showing that
human hearts.15, 16
patients can develop sudden tolerance to
repetitive exercise- or pacing-induced ischemia.25
Calcium Channel Blockers
Alkhulaifi (1994) and co-workers per-
Damage of cardiac membranes after
formed examinations on patients during
ischemia and reperfusion may result in a tran-
aortocoronary bypass-surgery.26 Ischemic pre-
sient intracellular calcium overload, what may
conditioning was achieved by 2 periods of
be responsible for the prolonged postischemic
cross-clamping for 3 minutes. Following 10
dysfunction. Calcium channel blockers can
minutes of global ischemia they found better
attenuate stunning of the myocardium and
preservation of myocardial ATP-content when
enhance the recovery of wall motion during
preconditioning was performed.
reperfusion.17
Karck et al27 demonstrated in an isolated
Furthermore, calcium channel blockers
rat heart model that ischemic precondition-
inhibit superoxide production in human neu-
ing can improve contractile function after a
trophils.18
10-hr period of hypothermic ischemia. This
endogenous mechanism of cardioprotection
Ischemic Preconditioning was effective regardless of whether preserva-
and Adenosine Pretreatment tion was accomplished using different
Ischemic preconditioning describes the cardioplegic solutions or topical hypother-
phenomenon whereby a significant reduction mia alone. This may have clinical implica-
of cell necrosis, contractile malfunction or tions in myocardial preservation for heart
arrhythmias induced by ischemia and reper- transplantation.
fusion can be achieved if the injurious period Rat experiments in a Langendorff model
of ischemia is preceded by one or more brief indicated the benefit of adenosine pretreat-
(2-5 min) periods of ischemia and reperfusion. ment for prolonged cardiac storage.28 In con-
Studies in the rat19 and the rabbit20 have shown trast, investigations in an isolated ejecting rat
that ischemic preconditioning can improve heart preparation showed no evidence to sup-
functional recovery after a prolonged period port the involvement of adenosine to any
of global ischemia. major extent in preconditioning induced pro-
The mechanism underlying precondition- tection of postischemic contractile function.29
ing has not yet been elucidated but one sug- Acadesine (5-amino-4-imidazole carboxa-
gestion is that the protective effect is mediated mide reboside) and its derivatives protect
by adenosine that is released during the short against myocardial injury during ischemia and
episode of preconditioning ischemia. Liu et reperfusion. This effect is probably mediated
al21 have shown that an intracoronary infusion by augmented extracellular adenosine.30
212 Ischemia-Reperfusion Injury in Cardiac Surgery

Pretreatment with Thyroid potential function of hearts harvested form


Hormone hemodynamically impaired donors.
Myocardial deterioration after brain death Usually, cold crystalloid cardioplegic solu-
is related to metabolic and hormonal changes.31 tions are used for induction of cardiac arrest
Recent studies have shown that thyroid of donor hearts. But the initial use of profound
hormone and vasopressin administration in hypothermic solutions may have detrimental
the brain dead donor may be beneficial to effects.36-38
long-term graft survival.32 T3 replacement can In the isolated working rat heart model
reverse the increased anaerobic to aerobic cardiac arrest was induced with St. Thomas
metabolism and improve hemodynamics in cardioplegia before the hearts were stored for
brain dead animals.11 6 hours in cardioplegic solution. Thereafter
In contrast, Meyers and co-workers33 found cardiac output and creatine kinase leakage were
no significant change in cardiac function of measured. Induction was started with cardio-
brain dead pigs after T3 treatment. plegic solution at 7.5, 22, or 37˚C for 1 minute
and was followed by a secondary infusion (2
Vasopressin minutes) with cold (7.5˚C) cardioplegia. Pri-
Hemodynamic instability commonly mary infusion with warm solutions resulted
observed in brain dead patients has been in decreased coronary vascular resistance dur-
explained by diabetes insipidus secondary to ing infusion and greater postischemic cardiac
vasopressin depletion as the primary mecha- function.
nism. Hemodynamic support of the brain
dead patient has been based on vasopressin and Organ Storage
catecholamine therapy. Unfortunately, vaso- The period of cold organ storage is one that
pressin, a potent vasoconstrictor, may improve “prevents” further injury of hearts but “does
hemodynamics at the expense of decreasing not make a poor donor heart a good one.35
splanchnic and myocardial perfusion.34 The main principle of organ storage is deep
hypothermia because it
Cardioplegic Induction Phase 1. reduces metabolic rate
The aim of myocardial protection in this 2. avoids cumbersome continuous organ
phase should be to prevent that severely com- perfusion
promised hearts enter the storage phase with 3. prevents swelling and oxygen free radi-
substrate depletion. cal injury
Metabolic depletion can be reversed by It is generally accepted that a temperature
inducing cardiac arrest with a warm substrate- of 4˚C should be used for the cardioplegic
enriched blood cardioplegic solution during solution. This temperature is believed to be
harvesting.9,35 Brain dead dogs received a achieved when the harvested heart is placed
10 minute infusion of warm blood cardiople- on ice and transported in a cooler. Experimen-
gia containing glutamate and aspartate. Meta- tal assessments in dogs showed that after 4
bolic investigations of the myocardium hours of cooling with ice and saline solution
showed a: in a cooler temperature was below 0˚C
1. return of creatine phosphate levels to throughout the myocardium. Examination
normal with an electron microscope revealed serial
2. replenishment of glutamate changes, including moderate-to-severe cyto-
3. reduction in myocardial lactate content. plasmic and nuclear swelling and mitochon-
The authors conclude that warm substrate- drial calcium deposits. Cell membranes
enriched blood cardioplegic solution may remained intact, which suggests that the
increase the tolerance to subsequent ischemia damage was not irreversible.39 Nevertheless,
during organ storage, provide potential unacceptably low temperatures during organ
expansion of the donor pool, or improve the storage (< 4˚C) should be avoided.
Protection Strategies for Heart Transplantation 213

In contrast, investigations on isolated with L-arginine, a nitric oxide precursor can


Wistar rat hearts after 6 hours of preservation reduce vascular dysfunction after heart
with University of Wisconsin solution showed transplantation.45, 50
a better recovery of cardiac function, myocar-
dial adenine nucleotides content, and preven- Reperfusion Phase
tion of myocardial edema when subzero Reperfusion injury occurs when ische-
nonfreezing storage at –1˚C as compared with mically stored heart transplants are abruptly
4˚C was used.40 reoxygenated by the recipient’s blood. Dam-
An increase in early mortality with an age to the myocardium is generated through a
increase in donor organ ischemic times beyond variety of mechanisms including free radical
4 hours has been reported.41,42 generation, calcium overload, and abnormali-
To avoid edema and swelling, slightly ties of energetics.
hyperosmolar cardioplegic solutions are used Analysis of coronary sinus and arterial
for heart storage. The optimal osmolality is blood samples in 11 transplant recipients
280-290 mOsm. Moderate or severe hyperos- showed a lactate release and pyruvate uptake
molality can be detrimental to cardiac in the first minute of reperfusion. A massive
function.43 efflux of nucleotide catabolites was observed
To prevent edema formation and improve for more than 30 minutes of reperfusion.
postischemically coronary flow hyaluronidase There was nearly no oxygen uptake after onset
has been added to preservation solutions.44 of reperfusion. The oxygen saturation of
Hyaluronidase acts by specifically reducing the hemoglobin in coronary sinus blood decreased
hyaluronate content of tissues, which is the gradually over the first 45 minutes, indicating
principal glycosaminoglycan of the intersti- gradual restoration of myocardial oxygen
tium, has a high water-binding capacity and uptake.51
exerts a strong osmotic force. Application of Postischemic reperfusion damage can be
hyaluronidase decreases the interstitial volume minimized by a brief (i.e., 3-5 minutes) con-
of edematous tissues and prevents ischemically trolled reperfusion with normothermic, sub-
induced increase in vascular resistance, but strate-enriched blood cardioplegia (“hot shot”)
does not change the microvascular permeabil- during the initial phase of reoxygenation.52,53
ity. Thus, hyaluronidase is very effective to The factors which minimize reperfusion injury
diminish the no reflow phenomenon. include precise control of the conditions and
Myocardial contracture seems to play a composition of the reperfusate. This involves
major role in the reduced vasodilatory capac- keeping the heart vented, delivering reper-
ity of coronary vessels after an extended period fusate normothermically to optimize metabo-
of cold storage. This hypothesis is illustrated lism, providing a gentle reperfusion pressure
by the finding, that rat hearts treated with 2,3- (< 50 mmHg), keeping the heart arrested dur-
butanedione monoxime (BDM) showed a ing the initial phase of reperfusion to mini-
reduced degree of left ventricular stiffness and mize oxygen demands, lowering reperfusate
an increase in coronary flow after 10 hours of calcium, providing a buffer to reverse acido-
hypothermic storage.45 BDM is a reversible sis, replenishing substrates (i.e., amino acid
inhibitor of cross-bridge formation in the weak precursors of Krebs cycle intermediates which
binding state and actively cycling cross have been depleted during ischemia), provid-
bridges.46 ing hyperosmolarity to limit edema formation,
Ischemic and reperfusion injury is associ- and providing extra glucose as metabolic sub-
ated with vascular dysfunction. Vasodilation strate. Controlled reperfusion has been shown
in the heart is mediated through the release of excellent results after revascularization of
nitric oxide by the endothelium.47,48 After severely ischemically damaged hearts,54 but
begin of reperfusion nitric oxide release from only few clinical studies have assessed this
the endothelium is impaired.49 Therefore, the method after heart transplantation.55-57
supplementation of cardioplegic solutions
214 Ischemia-Reperfusion Injury in Cardiac Surgery

There is increasing evidence that damage Granulocytes contain the enzyme NADPH
from acute myocardial ischemia is not solely oxidase, can produce large quantities of
due to the interruption of nutrient blood flow superoxide and are an important origin of oxy-
nor to the failure to remove metabolic prod- gen-derived free radicals.67
ucts. Rather, reperfusion initiates a cascade of Experimental68 and clinical69 investigations
reactions, including oxygen-radical formation, showed an accumulation of polymorpho-
mechanical capillary obstruction by granulo- nuclear leukocytes in myocardial capillaries
cytes, and inflammatory events that markedly during ischemia and reperfusion. This phe-
increase the injury. Numerous mechanism nomenon has been termed leukocyte plugging
have been proposed, including interactions and is the main mechanism of the capillary
between reactive oxygen species, neutrophils, no-reflow phenomenon occurring during the
and the capillary endothelium. first 5 hours of reperfusion after an initial
Free radicals cause peroxidation of mem- period of reactive hyperemia. Accumulation
brane lipids and denaturation of proteins. of leukocytes results from the rheologic prop-
Measurements on isolated rat hearts after erties of the granulocyte such that marked
graded ischemia showed a peak of hydroxyl deformation is necessary for capillary transit,
radical generation 30-90 seconds after the and because of natural adherence, there is a
onset of reperfusion.58 small safety margin for granulocyte transport
Normally, protective mechanisms are through capillaries. Following 1 hour of
present in the cell to prevent damage by free ischemia and brief reperfusion in dogs, 24%
radicals. Superoxide dismutase catalyzes the of myocardial capillaries remained obstructed
dismutation of O2- to H2O2 and O2. The by granulocytes.68 The phenomenon of leu-
cytoplasmic enzymes glutathione peroxidase kocyte plugging can not be observed by rou-
and catalase provide the final detoxification tine histologic methods, probably because
steps with the reduction of H2O2 to H2O. single granulocytes obstruct individual capil-
Glutathione peroxidase seems to be a more laries in a stochastic fashion, which is quite
active enzyme than catalase in protecting myo- different from leukocyte infiltration.
cardial cells from H2O2 mediated damage.59 An additional effect of granulocyte activa-
There is increased evidence that oxygen free tion during myocardial ischemia and reper-
radical scavengers i.e., superoxide dismutase fusion is degranulation. The effect of enzymes
(SOD), catalase (CAT), and mercapto- contained within these granules in ischemic
propionyl-glycine (MPG) inactivate free radi- myocardium is not known. The serine pro-
cals and thus ameliorate reperfusion injury.60-62 teinase elastase has been implicated most
Sodium lactobionate has been shown to reduce consistently in polymorphonuclear leukocyte-
free hydroxyl radical generation. The com- mediated tissue damage. Elastase may hydro-
pound can complex large amounts of metal lyze a host of proteins in the extracellular ma-
ions. Especially iron ions participate in the trix (i.e., elastin, fibronectin, and collagen) as
Fenton reaction and Haber-Weiss cycle, the well as complement proteins and clotting fac-
most important source of hydroxyl radicals.63 tors. In addition, elastase may inhibit platelet
Lactobionate is contained in Henry Mondor function by proteolysis of platelet membrane
and Celsior cardioplegic solution. glycoproteins.70
The best time for administration of scav- A means to halt the inflammation might
engers is just before and just after the onset of allow reperfusion with granulocyte-depleted
reperfusion.61,64 blood and prevent granulocyte-mediated
Formerly, the source of the oxygen-radical injury. Furthermore, adenosine and perfluoro-
superoxide was thought to be xanthine oxi- chemicals have been shown to reduce neutro-
dase in the endothelial cell.65 Recent studies phil adherence and cytotoxicity to endothelial
indicated that granulocytes are the major cell cultures.71
source of superoxide radicals.66 Adenosine may counteract the potassium
chloride-induced vasoconstriction that occurs
Protection Strategies for Heart Transplantation 215

during hyperkalemic reperfusion and may thus cardioplegic solution and reperfused in a
improve coronary flow and myocardial Langendorff model recovered 84% of control
function.72 contractile function.77
Piglet hearts were arrested with crystalloid
Future Directions cardioplegia, excised, and stored for 12 hours
The established concept for heart allograft in saline solution at 0˚C. Initial reperfusion
preservation involves single-dose cardioplegia (10 min) revealed a left ventricular stroke work
and static hypothermia. But intensive research index of 3.8 ± 2.3, 14.6 ± 1.3, and 19.8 ± 1.6 x 103
activities exist to develop effective methods for erg/g when whole blood, unmodified perfluoro-
long term storage and to improve long term chemical, or aspartate/glutamate-enriched
results after transplantation. perfluorochemical cardioplegia was used.78
Kojima et al79 tested a hexanol cardioplegic
24-Hour Storage solution containing pyruvate in an isolated rat
The safe period for clinical heart preserva- heart model after 18-hours preservation. The
tion remains approximately 4 hours, whereas percent recovery of left ventricular developed
clinical liver or kidney preservation times rou- pressure and rate-pressure product were sig-
tinely exceed 12 hours, and exceptionally 24 nificantly better with the hexanol cardioplegic
hours.73 solution compared with St. Thomas and
Successful prolonged storage in clinical Stanford solution.
practice from 4-8 hours has been reported only Wicomb80 tested rabbit hearts in an in vitro
in a few single cases.74,75 Recently, Obadia et model after 24-hour storage. For perfusion and
al76 reported 14 heart transplantations per- storage a simplified UW solution (Cardiosol)
formed in exceptional circumstances after a with polyethylene glycol instead of hydroxy-
long preservation period (10-13 hours). The ethyl starch was used. The mean cardiac out-
hearts were perfused with St. Thomas car- put of the hearts reached 80% of controls.
dioplegia number 2 + 100 mg procaine and In an in vitro human right atrial muscle
stored in the same solution at 4˚C. Immedi- preparation recovery of function after a
ately before aortic unclamping cardiopulmo- 24 hour storage period at 4˚C or 12˚C was
nary bypass (CPB) was stopped and CPB flow assessed. The developed forces (DF) in the 4˚C
rate was increased by 500 ml/min every 30 group were 59, 77, and 61% of the control
seconds to achieve low reperfusion pressure of for Euro-Collins, University of Wisconsin,
the heart. A glucose-insulin-potassium (GIK) and Bretschneider solution. For those
infusion was started at the unclamping of the cooled to 12˚C, DF were 10, 30, and 96%,
aorta and maintained for the first hour after respectively.81
operation. The survival rate was 75% at 1 year
and 71% at 5 years. Intermittent Perfusion
The problems resulting from prolonged In an isolated rat heart model Zhu and
heart storage are mainly contracture and co-workers82 investigated poststorage function
reduced coronary flow which inhibit sufficient after 24 hour storage at 0˚C and intermitted
recovery of cardiac function in reasonable time perfusion with oxygenated crystalloid cardio-
to guarantee the survival of the recipient. Many plegic solution (CP-11EB) for 3 minutes at
attempts have been made in animal experi- 10 and 17 hours of storage. Poststorage aortic
ments to improve the outcome of prolonged- flow was 65%, coronary flow 44%, cardiac
stored hearts including continuous perfusion output 58% and work 53% of the unstored
systems, modifications of storage solutions, controls.
and development of new preservation
solutions. Continuous Perfusion
The concept of continuous normothermic
Rabbit hearts that were arrested, cooled, perfusion with blood was discussed by Lower
preserved for 24 hours in an extracellular
216 Ischemia-Reperfusion Injury in Cardiac Surgery

et al83 and was advocated by Robicsek and i.e., prostaglandins, calcium antagonists and
co-workers84 in 1969. But logistically problems radical scavengers.
and cumbersome methods have prevented it But this is not in conformity with the ethi-
from being accepted in clinical practice. cal premise formulated by the First Interna-
In an isolated rat heart model the effects of tional Workshop on NHBD that the therapy
glucose, insulin, and aspartate as components of a patient should not be compromised on
of oxygenated St. Thomas’ solution on recov- the grounds that he is a potential donor and
ery of cardiac function after 24-hour preser- that a therapy just for the purpose of organ
vation were assessed.85 When used for continu- procurement can also not be accepted.91
ous perfusion the best recovery of cardiac Using the in-vivo pig model we harvested
output was achieved at a temperature of 20˚C. hearts 30 minutes after circulatory arrest. The
Recovery at 4˚C was significantly lower than animals were not pre-treated with cardio-
at 20˚C. protective drugs. We performed a perfusion
The efficacy of oxygenated UWS con- with cold leukocyte-depleted blood cardiople-
taining endothelin-A receptor antagonist gia (BCP) containing the Na+-H+-exchange
FR139317 for 24-hour preservation was tested (NHE) inhibitor HOE 642. NHE inhibitors
in continuously perfused isolated rabbit have been shown excellent protective efficiency
hearts.86 Percent recovery rates of cardiac out- to reduce ischemia and reperfusion injury in
put and coronary flow were 93.3% and 94.3%, different in vitro and in vivo models.92,93 After
respectively. orthotopic transplantation a second controlled
In a swine model of heart transplantation reperfusion with BCP and HOE 642 was
simple hypothermic storage and continuous performed.
hypothermic perfusion with crystalloid solu- The contractility of these hearts expressed
tion were compared. For the hypothermic per- as maximal left and right ventricular stroke
fusion group there was significant improve- work index was not significantly different as
ment of myocardial function after orthotopic compared to the control group (transplanta-
transplantation.87 tion without normothermic ischemia). In con-
Ferrera et al88 compared isolated pig hearts trast, the contractility was reduced significantly
were preserved in cold St. Thomas solution when the reperfusions were performed with-
for 24 hours either by simple storage or con- out HOE 642.94
tinuous microperfusion. After reperfusion the
microperfusion group showed higher levels of Preservation Solutions
tissue adenosine triphosphate and higher mean Currently, the majority of transplant pro-
left ventricular developed pressure. grams use crystalloid-based cardioplegic solu-
tions for myocardial preservation. The
Non-Heart-Beating Donors hypothermic, hyperkalemic crystalloid car-
The use of donor hearts harvested 30 min- dioplegia techniques have enabled safe myo-
utes or later after circulatory arrest could cardial preservation for up to 4 hours.
expand the donor pool. A number of poten-
tial donors die, before the procurement of Bretschneider’s HTK Solution
organs can be performed. After cardiac arrest In 1979 Bretschneider introduced his His-
the organs of these “non-heart-beating donors” tidine-Tryptophane-Ketoglutarate (HTK)
(NHBD) are exposed to unprotected normo- solution into clinical practice (Table 19.1).
thermic ischemia. Cardioplegia for open heart surgery has been
Experiments in dogs89 and primates 90 the first aim of this preservation solution. Ger-
showed that hearts from NHBD could be man heart surgeons have used Bretschneider
successfully transplanted if the donors received solution in heart transplantation since 1985.95
cardioprotective drugs before cardiac arrest, Bretschneider solution contains the amino
acid histidine, a biologically compatible,
Protection Strategies for Heart Transplantation 217

administrable buffer system with a high buffer hydroxyethylstarch to prevent edema during
capacity. HTK solution is a flush solution like hypothermic storage. The benefit of an intra-
Euro-Collins or University of Wisconsin cellular potassium solution is an early and more
solution. To achieve optimal protective effects, complete arrest along with prevention of mem-
the vascular and extracellular space have to be brane flux during hypothermia. Glutathione
equilibrated with the fluid. This is nearly is added at the time of administration and acts
accomplished after 8-10 minutes of perfusion as an antioxidant. The solution also contains
with a hydrostatic pressure of 80 mmHg. allopurinol to prevent xanthine oxidase-me-
Therefore approximately 3000-4000 mL of diated production of free radicals. Adenosine
cardioplegic solution are required. is a metabolic precursor to the formation of
In a retrospective study in cooperation with ATP and high-energy phosphates.
Eurotransplant and five heart transplant cen- In a large number of articles originating
ters 591 heart transplant patients were from the University of Wisconsin, but also
included.42 Immediate postoperative graft fail- from many other groups, UW solution was
ure was observed in 4.2%. Within the first 30 claimed to be superior of all other preserva-
days 11.8% of organs failed. Acute graft fail- tion fluids.98, 99
ure and early mortality correlated with the A clinical trial comparing UWS and
length of ischemic time. In addition, a higher Stanford solution revealed better preservation
incidence of graft failure was observed when of ATP and creatine phosphate levels,
the perfusion volume was less than 1500 ml. decreased defibrillations, decreased intraopera-
The 1- and 5-year survival rates were 72 and tive pacing, and trend toward decreased
63%, respectively. The authors advocate to requirement for inotropic support in the UW
avoid ischemic time of more than 4 h. group.100
The efficacy of Bretschneider vs. Univer- Even for heart preservation the possibility
sity of Wisconsin solution (UWS) for long- of prolonged preservation with UW solution
term preservation was tested in a primate was reported.80,101,102 As shown by Astier and
allotransplantation model.96 Bretschneider’s Paul103 the glutathione component of the UW
HTK solution allowed storage of hearts for changes from the reduced into the oxidized
periods of up to 10 hours. UWS provided form with a half-life of about 4 days. UW
superior results with regard to clinical outcome solution containing oxidized instead of
and hemodynamic recovery of hearts after reduced glutathione is significantly less effec-
ischemic periods of up to 16 hours. tive for preservation.104,105
In contrast, examinations in an in vitro The so-called simplified UWS (Cardiosol)
human atrial muscle preparation showed a contains polyethylene glycol instead of
greatly improved recovery of contractility after hydroxyethyl starch.
24-hour storage in HTK-solution compared The high potassium concentration of UWS
with both Euro-Collins and UW solutions.81 is accused to be responsible for endothelial
Recovery of developed force was affected by injury of the coronary arteries. Investigations
temperature (0˚C vs. 12˚C) for Euro-Collins in neonatal pig hearts showed a loss of endo-
and UW solutions but not for Bretschneider’s thelium-dependent vasodilatation after UWS
solution. caused by the inability of the endothelium to
release nitric oxide. In contrast, blood car-
University of Wisconsin Solution dioplegia did not result in impaired endothe-
The University of Wisconsin organ pres- lial function.106
ervation solution (UWS, Table 19.3) was ini- To avoid the deleterious effect of hyper-
tially developed by Belzer and co-workers for kalemic cardioplegic solution on coronary
pancreas transplantation.97 It is an intracellu- endothelium a new extracellular UWS formu-
lar type solution, with a high potassium lation was created.107 This solution contains
concentration. Components include the 25 mEq/L of potassium and 129 mEq of
impermeables lactobionate, raffinose, and sodium. The solution was tested in an isolated
218 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 19.1. Composition of Bretschneider’s HTK solution

Histidine 180 mmol/L


Tryptophane 18 mmol/L
Mannit 2 mmol/L
K-Ketoglutarate 20 mmol/l
Magnesium chloride 9 mmol/L
Potassium chloride 9 mmol/L
Sodium chloride 15 mmol/L
Total osmolarity 20 mOsm
pH 7.1

Table 19.2. Composition of UWS109

Pentafraction 0.45 mmol/L


Lactobionic acid 100 mmol/L
Magnesium sulfate 5 mmol/L
Raffinose 30 mmol/L
Adenosine 5 mmol/L
Allopurinol 1 mmol/L
Glutathione 3 mmol/L
Dexamethasone 16mg/L
Potassium 140 mEq/L
Sodium 20 mEq/L
Total osmolarity 320 mOsm
pH 7.4

Table 19.3. Composition of Stanford cardioplegic solution109

HCO3 28mEq/L
Mannitol 25gm/L
Glucose 50gm/L
Potassium 30mEq/L
Sodium 28mEq/L
pH 8.0
Osmolarity 450mOsm

piglet heart model. After 24-hours storage Stanford Solution


there was no significant difference of stroke Stanford cardioplegic solution (Table 19.4)
work index, high-energy phosphate stores and is typical of the class of extracellular type solu-
myocardial water content between the two tions. The solution contains potassium as the
UWS groups. Experiments in neonatal duroc cardioplegic agent, bicarbonate as buffer, and
piglets suggest that the low-potassium UWS glucose and mannitol for their osmotic prop-
solution provides superior protection of the erties. Furthermore, mannitol acts as a free
endothelium by preserving the endothelium- radical scavenger. Most centers using this
dependent vasodilatory response to nitric oxide solution tend to limit the ischemic period to
release.108 4 hours.109
Protection Strategies for Heart Transplantation 219

Table 19.4. Composition of St. Thomas’ cardioplegic solution112

Sodium chloride 10mmol/L


Potassium chloride 16 mmol/L
Magnesium chloride 16 mmol/L
Calcium chloride 1.2 mmol/L
Sodium bicarbonate 10.0 mmol/L
pH 7.8
Osmolarity 285-300 mOsm

In a retrospective analysis 195 patients were In a prospective, randomized study com-


reviewed for the development of cardiac paring preservation with UWS versus St. Tho-
allograft vasculopathy with a mean follow-up mas’ Hospital solution (STS, Table 19.4) 39
of 24 months.75,110 These patients were treated heart transplant patients were enrolled.
in identical fashion, except for the type of car- Hemodynamic, electron microscopic, and bio-
dioplegia used, Stanford solution or UWS. The chemical evaluation did not reveal any signifi-
data supported the conclusion that UWS is cant differences in postoperative myocardial
associated with an enhanced incidence of performance. Only the number of intraopera-
vasculopathy (14% versus 22%, p < 0.03). It tive defibrillations and the rhythm stability
is suggested that the high potassium concen- after reperfusion indicated superiority of
tration of intracellular solutions cause myo- UWS.113
cyte and endothelial cell injury resulting in a
higher incidence of graft atherosclerosis. Celsior
Celsior, a new crystalloid cardioplegic
St. Thomas Solution No. 2 solution (Table 19.5), was developed by
In 1975 the St. Thomas’ Hospital cardio- Menasche.114 The development of Celsior has
plegic solution No. 1 was introduced into clini- been conducted according to the following
cal practice. 111 The formulation of this solu- guidelines:
tion was based on the desire to deviate as little 1. Prevention of calcium overload by the
as possible from the normal extracellular ionic low calcium concentration of the solu-
composition and to minimize the amount of tion (100mM), by magnesium at a con-
potassium that had traditionally been used to centration high enough (13mM) to
ensure rapid and complete arrest. This solu- antagonize calcium fluxes and by main-
tion had widespread clinical use throughout taining a slight degree of acidosis (pH
the world. As a result of numerous experimen- 7.30).
tal studies the St. Thomas Hospital cardio- 2. Prevention of myocardial edema by
plegic solution No. 2 was introduced in 1981. mannitol and lactobionate.
The main features of this solution, as com- 3. Prevention of severe intracellular acido-
pared to the earlier solution, are a reduction sis by buffering with histidine. The con-
in the sodium and potassium content, a centration of histidine in Celsior is simi-
reduction in calcium by 50%, the omission of lar to that of blood (30mM).
procaine, the incorporation of a small amount 4. Prevention of oxidative damage by
of bicarbonate, and the adjustment of pH to reduced glutathione.
7.8. In an isolated rat heart model St. Tho- Furthermore, Celsior contains glutamate
mas’ Hospital solution No. 2 provided sub- to restore the ischemia-induced loss of high
stantially superior protection compared with energy phosphates, in particular ATP. Hith-
No.1.112 erto, no controlled prospective clinical trials
with this solution have been published.
220 Ischemia-Reperfusion Injury in Cardiac Surgery

Table 19.5. Composition of Celsior cardioplegic solution82

Lactobionate 80 ∝mol/L
Mannitol 60 ∝mol/L
Reduced glutathione 3 ∝mol/L
Glutamate 20 ∝mol/L
Histidine 30 ∝mol/L
Potassium 15 ∝mol/L
Sodium 100 ∝mol/L
Magnesium 13 ∝mol/L
Calcium 0.26 ∝mol/L
Chloride 41.5 ∝mol/L
Total osmolarity 360 mOsm
pH 7.3

Blood Cardioplegia solution at 20-minute intervals from time of


Blood cardioplegia and warm reperfusion arrival in the recipient theater to the time of
(Table 19.6) have demonstrated enhanced implantation. In addition the hearts were
myocardial protection in standard open heart reperfused with warm blood cardioplegic
operations. But the use of blood cardioplegia solution containing 20 mmol/L aspartate (“hot
for the procurement of donor hearts in clini- shot”) just before aortic cross-clamp removal.
cal practice might bear problems because of No difference was found in allograft function,
the cumbersome technique. functional capacity, the development of trans-
Nataf and co-workers55 used blood car- plant-associated coronary disease or actuarial
dioplegia for the preservation of donor survival in the groups with an ischemic time
hearts. Harvesting of the donor graft began of less than 241 minutes, 241-300 minutes
with a perfusion of cold crystalloid solu- and more than 300 minutes.
tion (Plegisol). Perfusion of a first dose of
blood cardioplegia (8-10˚C) for 3 minutes was Conclusions
immediately started on the arrival of the graft The majority of available hypothermic,
in the operating room. Reinfusions were per- hyperkalemic crystalloid cardioplegia tech-
formed each 20 minutes. At the end of the niques in heart transplantation have enabled
aortic anastomosis myocardial warm reper- safe myocardial preservation for up to 4 hours.
fusion with glutamate-containing blood Research efforts are in progress to develop pres-
cardioplegia was started. Retrospective com- ervation methods that would decrease myo-
parison of two matched cohorts of 50 cardial injury both during the period of pres-
patients receiving either crystalloid or blood ervation and during the period of reperfusion.
cardioplegia revealed a significantly better Attention has to be focused on the best
recovery of cardiac function in the blood car- “myocardial preservation” as well as on the
dioplegia group. “best endothelial cell preservation” to avoid an
Briganti and co-workers56 reported success- initial injury may result in early graft failure,
ful long-term outcome after transplantation chronic rejection, or the development of car-
of hearts with prolonged ischemic time of diac allograft vasculopathy.
more than 300 minutes. Cardioplegia was One of the major challenges in transplan-
achieved with oxygenated St. Thomas’ Hos- tation remains the extension of the preserva-
pital No. 2 solution supplemented with aspar- tion period.
tate. Allografts received a reinfusion of aspar- Benefit of blood cardioplegic solutions for
tate-supplemented cold blood cardioplegic the preservation and reperfusion of hearts has
been shown in experimental and clinical
Protection Strategies for Heart Transplantation 221

Table 19.6. Composition of blood cardioplegia for reperfusion116

THAM (Tromethamine) 8-10mEq/L


Citrate Phosphate Dextrose Ca2+ 0.15-0.20 mmol/L
Glutamate 13 mmol/L
Aspartate 13 mmol/L
Glucose 40 mmol/L
Calcium 0.15-0.25 mmol/L
Potassium Chloride 8-10mEq/L
Total osmolarity 380-400 mOsm
pH 7.5-7.6

investigations but it’s use for the procurement 9. Buckberg GD. Invited letter concerning:
of donor hearts is limited by the cumbersome Phases of myocardial protection during heart
transplantation. J Thorac Cardiovasc Surg
technique. 1990; 100:461-2.
10. Burtin P, Mertes PM, Pinelli G et al. Myo-
References cardial ischemia during experimental brain
1. Drinkwater DC, Rudis E, Laks H et al. Uni- death. Transplant Proc 1993; 25:3107-8.
versity of Wisconsin Solution versus Stanford 11. Novitzky D, Cooper DKC, Morrell D et al.
Cardioplegic Solution and the development Change from aerobic to anaerobic metabo-
of cardiac allograft vasculopathy. J Heart lism after brain death and reversal following
Lung Transplant 1995; 14:891-6. triiodothyronine. Transplantation 1988;
45:32-6.
2. Day JD, Rayburn BK, Gaudin PB et al. Car- 12. Shatney CHO, MacCarter DJ, Lillehei RC.
diac allograft vasculopathy: The central patho- Effects of allopurinol, propanolol and methyl-
genic role of ischemia-induced endothelial cell prednisone on infarct size in experimental
injury. J Heart Lung Transplant 1995; myocardial infarction. Am J Cardiol 1976;
14:142-9. 37:572-80.
3. Hosenpud JD, Bennett LE, Berkeley M et al. 13. Arnold SL, DeWall RA, Kezdi P et al. The
The registry of the International Society of effect of allopurinol on the degree of early
Heart and Lung Transplantation, 14th offi- myocardial ischemia. Am Heart J 1980;
cial report 1996. J Heart Lung Transplant 99:616-624.
1997; 16:691-712. 14. Reimer KA, Jennings RB. Failure of the xan-
4. Darby JM, Stein K, Grenvik A et al. Ap- thine oxidase inhibitor allopurinol to limit
proach to management of the heart-beating infarct size after ischemia and reperfusion in
“brain dead” organ donor. JAMA 1989; dogs. Circulation 1985; 71:1069-75.
261:2222-8. 15. Eddy LJ, Stewart JR, Jones HP et al. Free
5. Mertes PM, Burtin P, Carteaux JP et al. radical-producing enzyme, xanthine oxidase,
Changes in hemodynamic performance and is undetectable in human hearts. Am J Physiol
oxygen consumption during brain death in 1987; 253:H709-11.
the pig. Transplant Proc 1994; 26:229-30. 16. Podzuweit T, Braun W, Muller A et al. Ar-
6. Sebening C, Hagl C, Szabo G et al. Cardio- rhythmias and infarction in the ischemic pig
circulatory effects of acutely increased intrac- heart are not mediated by xanthine oxidase-
ranial pressure and subsequent brain death. derived free oxygen radicals (Abstract). Cir-
Eur J Cardiothorac Surg 1995; 9:360-72. culation 1986; 74(Pt2):II346.
7. Bittner HB, Kendall SWH, Chen EP et al. 17. Heusch G. Myocardial stunning, a role for
The combined effects of brain death and car- calcium antagonists during ischaemia?
diac graft preservation on cardiopulmonary Cardiovasc Res 1992; 26:14-9.
hemodynamics and function before and after 18. Irita K, Fujita I, Takshige K et al. Calcium
subsequent heart transplantation. J Heart channel antagonists induced inhibition of su-
Lung Transplant 1996; 15:764-77. peroxide production in human neutrophils:
8. Emery RW, Cosk RC, Levinson MM et al. Mechanism independent of antagonizing cal-
The cardiac donor: A six-year experience. Ann cium influx. Biochem Pharmacol 1986;
Thorac Surg 1986; 41:356-62. 35:3465-71.
222 Ischemia-Reperfusion Injury in Cardiac Surgery

19. Cave AC, Downey JM, Hearse DJ. Adenos- 33. Meyers CH, D’Amico TA, Peterseim DS et
ine fails to substitute for preconditioning in al. Effects of triiodothyronine and vasopressin
the globally ischemic isolated rat heart (Ab- on cardiac function and myocardial blood
stract) J Mol Cell Cardiol 1991; 23(SIII):S76 flow after brain death. J Heart Lung Trans-
20. Omar B, Hanson A, Bose S et al. Reperfusion plant 1993; 12:68-80.
with pyruvate eliminates ischemic precondi- 34. Gaskill HV, Sirinek KR, Levine BA. Hemo-
tioning in the isolated rabbit heart: An ap- dynamic effects of vasopressin. Arch Surg
parent role for enhanced glycolysis. Coronary 1983; 118:434-7.
Heart Dis 1991; 2:799-804. 35. Tixier D, Matheis G, Buckberg GD et al.
21. Liu GS, Thornton J, Van Winkle DM, Pro- Donor hearts with impaired hemodynamics.
tection against infarction afforded by precon- Benefit of warm substrate-enriched blood
ditioning is mediated by A1 receptors in rab- cardioplegic solution for induction of car-
bit heart. Circulation 1991; 84:350-6. dioplegia during cardiac harvesting. J Thorac
22. Wyatt DA, Edmunds MC, Berne RM et al. Cardiovasc Surg 1991; 102:207-14.
Adenosine stimulates glycolytic flux in iso- 36. Magovern GJ Jr, Flaherty JT, Gott VL et al.
lated perfused rat hearts by A1 adenosine re- Failure of blood cardioplegia to protect myo-
ceptors. Am J Physiol 1989; 257:H1952-7. cardium at lower temperatures. Circulation
23. Lasley R, Rhee J, VanWylen D et al. Adenos- 1982; 66:I60-7.
ine A1 receptor mediated protection of the 37. Heitmiller RF, DeBoer LSV, Geffin GA et
globally ischemic isolated rat heart. J Mol Cell al. Myocardial recovery after hypothermic ar-
Cardiol 1990; 22:39-47. rest: A comparison of oxygenated crystalloid
24. Cronstein BN, Kramer SB, Weissman G et to blood cardioplegia. The role of calcium.
al. Adenosine: A physiological modulator of Circulation 1985;72:II241-53.
superoxide anion generation by human neu- 38. Takahashi A, Hearse DJ, Braimbridge MV et
trophils. J Exp Med 1983; 158:1160-77. al. Harvesting hearts from long-term preser-
25. Kloner RA, Yellon D. Does ischemic precon- vation. Detrimental effects of initial hypoth-
ditioning occur in patients? J Am Coll ermic infusion of cardioplegic solutions. J
Cardiol 1994; 24:1133-42. Thorac Cardiovasc Surg 1990; 100:371-8.
26. Alkhulaifi AM, Yellon DM, Pugsley WB. 39. Hendry PJ, Walley VM, Koshal A et al. Are
Preconditioning the human heart during temperatures attained by donor hearts dur-
aortocoronary bypass-surgery. Eur J ing transport too cold? J Thorac Cardiovasc
Cardiothorac Surg 1994; 8:270-6. Surg 1989; 98:517-22.
27. Karck M, Rahmanian P, Haverich A. Is- 40. Sakaguchi H, Kitamura S, Kawachi K et al.
chemic preconditioning enhances donor heart Preservation of myocardial function and me-
preservation. Transplantation 1996; 62:17-22. tabolism at subzero nonfreezing temperature
28. Fremes SE, Zhang J, Furukawa RD et al. storage of the heart. J Heart Lung Transplant
Adenosine pretreatment for prolonged cardiac 1996; 15:1101-7.
storage. An evaluation with St. Thomas’ Hos- 41. Young JB, Naftel DC, Bourge RC et al.
pital and University of Wisconsin solutions. Matching the heart donor and heart trans-
J Thorac Cardiovasc Surg 1995; 110:293-301. plant recipient. Clues for successful expansion
29. Cave AC, Clifford SC, Downey JM et al. of the donor pool: A multivariable, multi-in-
Improved functional recovery by ischemic stitutional report. J Heart Lung Transplant
preconditioning is not mediated by adenos- 1994; 13:353-65.
ine in the globally ischemic isolated rat heart. 42. Reichenspurner H, Russ C, Überfuhr P et al.
Cardiovasc Res 1993; 27:663-8. Myocardial preservation using HTK solution
30. Galinanes M, Bullough D, Mullane KM et for heart transplantation. A multicenter study.
al. Sustained protection by acadesine against Eur J Cardiothorac Surg 1993; 7:414-9.
ischemia- and reperfusion-induced injury: 43. Batty PR, Hicks GL, DeWeese JA et al. Op-
Studies in the transplanted rat heart. Circu- timal osmolality for cold storage of the car-
lation 1992; 86:589-97. diac explant. J Surg Res 1990; 48:601-5.
31. Finkelstein I, Toledo-Pregra L, Castellanos J. 44. Fischer JH, Jeschkeit S. Minimal amounts of
Physiologic and hormonal changes in experi- hyaluranidase in HTK or UW solution sub-
mentally induced brain dead dogs. Transplant stantially improve the recovery of preserved
Proc 1987; 19:4156-8. hearts. Transpl Int 1996; 9 (Suppl1):442-6.
32. Orlowski JP, Spees EK. Improved cardiac 45. Kevelaitis E, Mouas C, Menasche, P. Post-
transplant survival with thyroxin treatment of storage diastolic abnormalities of heart trans-
hemodynamic unstable donors: 95.2% graft plants: Is vascular dysfunction or myocardial
survival at 6 and 30 months. Transplant Proc contracture the culprit? J Heart Lung Trans-
1993; 25:1535 plant 1996; 15:461-9.
Protection Strategies for Heart Transplantation 223

46. Hajjar RJ, Ingwall JS, Gwathmey JK. Mecha- 60. Ambrosio G, Weisfeldt MYL, Jacobus W et
nism of action of 2,3-butanedione monoxime al. Evidence for a reversible oxygen radical-
on contracture during metabolic inhibition. mediated component of reperfusion injury:
Am J Physiol 1994; 267:H100-8. Reduction by recombinant human superox-
47. Benyo Z, Kiss G, Szabo C et al. Importance ide dismutase administered at the time of
of basal nitric oxide synthesis in regulation reflow. Circulation 1987; 75:282-91.
of myocardial blood flow. Cardiovasc Res 61. Bolli R, Jeroudi MO, Bharat SP et al. Marked
1991; 25:700-3. reduction of free radical generation and con-
48. Mankad PS, Chester AH, Yacoub MH. 5- tractile dysfunction by antioxidant therapy
Hydroxytryptamine mediates endothelial de- begun at the time of reperfusion. Evidence
pendent coronary vasodilation in the isolated that myocardial “stunning” is a manifestation
rat heart by the release of nitric oxide. of reperfusion injury. Circ Res 1989; 65:
Cardiovasc Res 1991; 25:244-8. 607-22.
49. Seccombe JF, Pearson PJ, Schaff HV. Oxy- 62. Sun SC, Appleyard R, Masetti P et al. Im-
gen radical-mediated vascular injury selectively proved recovery of heart transplants by com-
inhibits receptor-dependent release of nitric bined use of oxygen-derived free radical scav-
oxide from canine coronary arteries. J Thorac engers and energy enhancement. J Thorac
Cardiovasc Surg 1994; 107:505-9. Cardiovasc Surg 1992; 104:830-7
50. Weyrich AS, Ma XL, Lefer AM. The role of 63. Charloux C, Ishii K, Paul M et al. Reduced
L-arginine in ameliorating reperfusion injury production of the deleterious hydroxyl free
after myocardial ischemia in the cat. Circula- radical during the final reperfusion of isolated
tion 1992; 86:279-88. rabbit heart with the use of an improved so-
51. Smolenski RT, Seymour AM, Yacoub MH. dium lactobionate-based cardioplegic me-
Dynamics of energy metabolism in the trans- dium. J Heart Lung Transplant 1994; 13:
planted human heart during reperfusion. J 481-8.
Thorac Cardiovasc Surg 1994; 108:938-45. 64. Bando K, Teramoto S, Tago M et al. Oxy-
52. Buckberg GD. Myocardial protection: An genated perfluorocarbon, recombinant human
overview. Sem Thorac Cardiovasc Surg 1993; superoxide dismutase, and catalase ameliorate
5:98-106. free radical induced myocardial injury dur-
53. Buckberg GD. Update of current techniques ing heart preservation and transplantation. J
of myocardial protection. Ann Thorac Surg Thorac Cardiovasc Surg 1988; 96:930-8.
1995; 60:805-14. 65. McCord JM. Oxygen-derived free radicals in
54. Beyersdorf F, Mitrev Z, Sarai K et al. Chang- postischemic injury. N Engl J Med 1985;
ing patterns of patients undergoing emergency 312:159-63.
surgical revascularization for acute coronary 66. Engler R, Covell W. Granulocytes cause
occlusion. J Thorac Cardiovasc Surg 1993; reperfusion ventricular dysfunction after 15-
106:137-48. minute ischemia in the dog. Circulation Re-
55. Nataf P, Pavie A, Bracamontes L et al. Myo- search 1987; 61:20-28.
cardial protection by blood cardioplegia and 67. Babior BM. Oxygen dependent microbial kill-
warm reperfusion in heart transplantation. ing by phagocytes. N Engl J Med 1978;
Ann Thorac Surg 1992; 53:525-6. 659-68.
56. Briganti EM, Bergin PJ, Rosenfeldt FL et al. 68. Engler RL, Dahlgren MD, Morris D et al.
Successful long-term outcome with prolonged Role of leukocytes in response to acute myo-
ischemic time cardiac allografts. J Heart Lung cardial ischemia and reflow in dogs. Am J
Transplant 1995; 14:840-5. Physiol 1986; 252:H314-322.
57. Pradas G, Cuenca J, Juffe A. Continuous 69. Dhôte-Burger P, Vuillemenot A, Lecompte T
warm reperfusion during heart transplanta- et al. Is myocardial function influenced by
tion. J Thorac Cardiovasc Surg 1996; 111: neutrophil activation during ischemia and
784-90. reperfusion in coronary artery surgery? Trans-
58. Takemura G, Onodera T, Ashraf M. Quan- plant Proc 1995; 27:2786-8.
tification of hydroxyl radical and its lack of 70. Hansen PR. Role of neutrophils in myocar-
relevance to myocardial injury deriving early dial ischemia and reperfusion. Circulation
reperfusion after graded ischemia in rat hearts. 1995; 91:1872-85.
Circ Res 1992; 71:96-105. 71. Forman MB, Virmani R, Puett DW. Mecha-
59. Fantone JC, Ward PA. Role of oxygen-de- nisms and therapy of myocardial reperfusion
rived free radicals and metabolites in leuko- injury. Circulation 1990; 81(Suppl.IV):69-78.
cyte- dependent inflammatory reactions. Am 72. Habazettl H, Palmisano BW, Graf BH et al.
J Pathol 1982; 107:395-418. Improvement in functional recovery of the
isolated guinea pig heart after hyperkalemic
reperfusion with adenosine. J Thorac
Cardiovasc Surg 1996; 111:74-84.
224 Ischemia-Reperfusion Injury in Cardiac Surgery

73. Todo S, Nery J, Yanaga K et al. Extended 86. Okada K, Yamashita C, Okada M. Efficacy
preservation of human liver grafts with UW of oxygenated University of Wisconsin solu-
solution. JAMA 1989; 261:711-4. tion containing endothelin-A receptor antago-
74. Jeevanandam V, Auteri JS, Sanchez JA et al. nist in twenty-four-hour heart preservation.
Improved heart preservation with University J Heart Lung Transplant 1996; 15:475-84.
of Wisconsin solution-experimental and pre- 87. Qayumi AK, Jamieson WR, Rosado LJ et al.
liminary human experience. Circulation 1991; Preservation techniques for heart transplan-
84;324-8. tation: comparison of hypothermic storage
75. Drinkwater DC, Stein DG, Permut LC et al. and hypothermic perfusion. J Heart Lung
Clinical trial of University of Wisconsin So- Transplant 1991; 10:518-26.
lution for cardiac transplantation-preliminary 88. Ferrera R, Marcsek P, Larese A et al. Com-
results. J Thorac Cardiovasc Surg 1991; parison of continuous mircoperfusion and
102:798-9. cold storage for pig heart preservation. J
76. Obadia JF, Girard C, Ferrara R et al. Long Heart Lung Transplant 1993; 12:463-9.
conservation organs in heart transplantation: 89. Takagaki M, Hisamochi K, Morimoto T et
Postoperative results and long-term follow-up al. Successful transplantation of cadaver hearts
in fourteen patients. J Heart Lung Transplant harvested one hour after hypoxic cardiac ar-
1997; 16:256-9. rest. J Heart Lung Transplant 1996; 15:527-
77. Burt JM, Copeland JG. Myocardial function 31.
after preservation for 25 hours. J Thorac 90. Gundry SR, Fukushima N, Eke CC et al.
Cardiovasc Surg 1986; 92:238-246. Successful survival of primates receiving trans-
78. Martin SM, Laks H, Drinkwater DC et al. plantation with “dead”, nonbeating donor
Perfluorochemical reperfusion yields improved hearts. J Thorac Cardiovasc Surg 1995;
myocardial recovery after global ischemia. 109:1097-102.
Ann Thorac Surg 1993; 55:954-60. 91. Obermann K, Nagel E, Pichlmayr R. Ethical
79. Kojima S, Wu ST, Wikman-Coffelt J et al. considerations in procuring organs from non-
Eighteen-hour preservation of rat hearts with heart-beating donors after sudden cardiac
hexanol and pyruvate cardioplegia. J Am Coll death. Transplantation Proceedings 1995;
Cardiol 1993; 21:1238-44. 5:2924-5.
80. Wicomb WN, Hill DJ, Collins GM. Twenty- 92. Hendrikx M, Mubagwa K, Verdonck F et al.
four-hour ice storage of rabbit heart. J Heart New Na+-H+ exchange inhibitor HOE 694
Lung Transplant 1994; 13:891-4. improves postischemic function and high-en-
81. Hendry PJ, Labow RS, Keon WJ. Compari- ergy phosphate resynthesis and reduces Ca2+-
son of intracellular solutions for donor heart overload in isolated perfused rabbit heart.
preservation. J Thorac Cardiovasc Surg 1993; Circulation 1994; 89:2787-98.
105:667-73. 93. Scholz W, Albus U, Counillon L et al. Pro-
82. Zhu Q, Yang X, Claydon MA et al. Twenty- tective effects of HOE 642, a selective
four-hour intermittent perfusion storage of sodium-hydrogen exchange subtype 1 inhibi-
the isolated rat heart: The effect of perfusion tor, on cardiac ischaemia and reperfusion.
intervals on functional preservation. J Heart Cardiovasc Res 1995; 29:260-268.
Lung Transplant 1994; 13:882-90. 94. Martin J, Sarai K, Yoshitake M et al. Suc-
83. Lower RR, Stofer RC, Hurley EJ et al. Suc- cessful orthotopic pig heart transplantation
cessful homotransplantation for the canine from non-heart-beating donors. J Heart Lung
heart after anoxic preservation for seven Transplant 1999; 18:597-606.
hours. Am J Surg 1962;104:302-5. 95. Hölscher M, Groenewoud AF. Current sta-
84. Robicsek F, Tam W, Daugherty HK. Sur- tus of the HTK solution of Bretschneider in
vival of heart grafts. Arch Surg 1969; 99: organ preservation. Transplant Proc 1991;
750-2. 23:2334-7.
85. Shimada Y, Yamamoto F, Yamamoto H et 96. Human PA, Holl J, Vosloo S et al. Extended
al. Temperature-dependent cardioprotection cardiopulmonary preservation: University of
of exogenous substrates in long-term heart Wisconsin solution versus Bretschneider’s
preservation with continous perfusion: cardioplegic solution. Ann Thorac Surg 1993;
Twenty-four-hour preservation of isolated rat 55:1123-30.
heart with St. Thomas’ hospital solution con- 97. Belzer FO, Southard JH. Principles of solid-
taining glucose, insulin, and aspartate. J Heart organ preservation by cold storage. Transplan-
Lung Transplant 1996; 15:485-95. tation 1988; 45:673-6.
98. Breda MA, Drinkwater DC, Laks H et al.
Successful long-term preservation of the neo-
natal heart with a modified intracellular so-
lution. J Thorac Cardiovasc Surg 1992;
104:139-50.
Protection Strategies for Heart Transplantation 225

99. Jeevanandam V, Barr ML, Auteri JS et al. 108. Lee J, Drinkwater DC Jr, Laks H et al. Pres-
University of Wisconsin solution versus crys- ervation of endothelium-dependent vasodila-
talloid cardioplegia for human donor heart tion with low-potassium University of Wis-
preservation. A randomized blinded prospec- consin solution. J Thorac Cardiovasc Surg
tive clinical trial. J Thorac Cardiovasc Surg 1996; 112:103-10.
1992; 103:194-9. 109. Aziz S, Tada Y, Jaffery S et al. University of
100. Stein DG, Drinkwater DC, Laks H et al. Wisconsin solution provides superior myocar-
Cardiac preservation in patients undergoing dial preservation compared with Stanford
transplantation. A clinical trial comparing cardioplegic solution. J Heart Lung Trans-
University of Wisconsin solution and plant 1994; 1099-108.
Stanford solution. J Thorac Cardiovasc Surg 110. Drinkwater DC, Rudis E, Laks H et al. Uni-
1991; 102:657-65. versity of Wisconsin solution versus Stanford
101. Swanson DK, Pasaoglu I, Berkoff HA et al. cardioplegic solution and the development of
Improved heart preservation with UW pres- cardiac allograft vasculopathy. J Heart Lung
ervation solution. J Heart Lung Transplant Transplant 1995; 14:891-6.
1988; 7:456-61. 111. Braimbridge MV, Chayen J, Bitensky L et
102. Stein DG, Permut LC, Drinkwater DC et al. al. Cold cardioplegia or continuous coronary
Complete functional recovery after 24-hour perfusion? Report on preliminary clinical ex-
heart preservation with University of Wiscon- perience as assessed cytochemically. J Thorac
sin solution and modified reperfusion. Cir- Cardiovasc Surg 1977; 74:900-6.
culation 1991; 84(5Suppl):III 316-23. 112. Ledingham SJM, Braimbridge MV, Hearse
103. Astier A, Paul M. Instability of reduced glu- DJ. The St. Thomas’ Hospital cardioplegic
tathione in commercial Belzer cold storage solution. A comparison of the efficacy of two
solution. Lancet 1989; 2:556. formulations. J Thorac Cardiovasc Surg 1987;
104. Boudjema K, Vangulik TM, Lindell SL et al. 93:240-6.
Effect of oxidized and reduced glutathione in 113. Demertzis S, Wippermann J, Schaper J et al.
liver preservation. Transplantation 1990; University of Wisconsin versus St. Thomas’
50:948-51. Hospital solution for human donor heart
105. Fischer JH, Jeschkeit S. Effectivity of freshly preservation. Ann Thorac Surg 1993; 55:
prepared or refreshed solutions for heart pres- 1131-7.
ervation versus commercial Eurocollins, 114. Menasche P, Termignon JL, Pradier R et
Bretschneiders HTK, and University of Wis- al. Experimental evaluation of Celsior, a
consin solution. Transplantation 1995; 59: new heart preservation solution. Eur J
1259-62. Cardiothorac Surg 1994; 8:207-13.
106. Pearl JM, Laks H, Drikwater DC et al. Loss 115. Buckberg GD. Blood cardioplegia. Il Cuore
of endothelium-dependent vasodilatation and 1988; 3:271-81.
nitric oxide release after myocardial protec-
tion with University of Wisconsin solution.
J Thorac Cardiovasc Surg 1994; 107:257-64.
107. Drinkwater DC, Ziv ET, Laks H et al. Ex-
tracellular and standard University of Wiscon-
sin solutions provide equivalent preservation
of myocardial function. J Thorac Cardiovasc
Surg 1995; 110:738-45.
226 Ischemia-Reperfusion Injury in Cardiac Surgery

Index
A Cardiopulmonary bypass 2, 4-6, 8, 11, 14, 18,
19, 75, 76, 95, 117, 121, 122, 126,
Acetyl-CoA 82, 83, 94 129-131, 147, 160-162, 168, 171, 172,
Acute myocardial infarction 197-201 180, 194, 200, 201, 204-206, 208, 216
ADP 6 CD11/CD18 10
Alanine 82-84, 90, 91, 140, 143 Cold blood cardioplegia 161, 162, 184, 185,
Alpha-ketoglutarate 141, 211 192, 193
Amino acids 79-85, 87, 90-95, 136, 137, 140, Controlled reperfusion 200, 201
141, 151, 185, 187 Coronary operation 193, 194
Ammonia 91, 93
Antegrade cardioplegic infusion 206 E
Aortic dissection 204, 208
Aortic regurgitation 204, 207, 208 E-selectin 10
Aortic valve operation 193, 194 Endothelial dysfunction 6-9, 11, 17
Apoptosis 15, 21, 29, 45, 50, 51, 54-58 Endothelium 2-13, 15, 17, 21, 22, 54, 61-63,
Aspartate 80-87, 90, 91, 136, 140-142, 185, 65, 67, 95, 99, 160, 163, 214, 215, 218,
187, 192, 198, 202, 216, 220, 222, 224 219
ATP 6, 31, 34, 50, 55, 71-73, 79-85, 90, 91, Energy metabolism 151
93-95, 109-114, 116, 117, 125, 126, Energy reserve 145
128-130, 136, 141, 150, 177, 180, 181, Energy substrate 139
185, 211, 215, 223, 224
Autacoid 2, 4, 21 F
Axillary approach 205
Fas 41, 44
B Femoro-femoral access 205
Fumarate 83, 85, 87, 90-92
Bax 41, 46
Bcl-2 41, 44, 46 G
Blood cardioplegia 3, 10-12, 14-16, 18, 20,
91, 126, 141, 142, 159, 164-169, Glucose 52, 55, 108, 111-113, 117, 118,
171-177, 179-182, 184-188, 190-194, 135-140, 143, 146-148, 151, 178, 187,
196, 197, 199, 200, 223, 225, 226 214, 216, 217, 219, 222
Branched chain amino acid 80, 90, 93, 140 Glutamate 80-87, 90, 91, 93, 136-138,
140-151, 169, 185, 187, 192, 200, 211,
C 213, 216, 220-222
Glutamine 91, 140
C1-esterase inhibitor 46, 47 Glycogen 110-112, 127, 136-140, 159, 211
Cardiac surgery 2, 6, 8, 18
Cardiogenic shock 197-202 I
Cardioplegia 2, 3, 8, 10-12, 14-16, 18-21,
85-87, 96, 97, 99, 100, 117, 125, 126, Infectious endocarditis 204, 206
128, 130, 131, 136, 141, 142, 144, Inotropic drugs 135, 139, 140, 144-148, 150,
159-163, 166-172, 175-182, 184-188, 151
191-195, 200, 205, 207, 208, 213, 214, Insulin 137-140, 146, 147
216-218, 220-223 Insulin-like growth factor 46
Index 227

Integrated myocardial management 184, 187 O


Internal mammary artery 201
Intrapericardial tissue adhesion 204 Oxaloacetate 81, 82, 90, 91
Ion homeostasis 70, 95, 113, 118 Oxidation 3, 5, 15, 79-82, 84, 90, 95, 136,
Ischemia 2, 3, 6-11, 13-15, 17-21, 28, 29, 139, 140, 146, 150
32-38, 41, 42, 44-47, 49-56, 61-67, Oxygen consumption 159-162
70-75, 79, 81-85, 87, 90, 91, 93, 94, 95,
99, 100-104, 106-109, 111-113, P
115-117, 125-131, 135-142, 144-146,
148, 150, 151, 159, 163, 166, 168, 169, P-selectin 10, 17, 22
171, 176-178, 180, 181, 184-186, 192, Peroxynitrite 3, 5, 15, 19, 21, 22, 61, 65
194, 197-200, 202, 212-215, 217, 220 Programmed cell death 21, 37, 41, 42
Isoleucine 94, 140 Protein 8, 10, 15, 17, 19, 21, 35, 42, 44, 46,
49-57, 80-82, 87, 92, 94, 107, 111,
L 113-116, 125, 126, 128, 129, 178, 215
Pyruvate 82-84, 91, 136, 138-140, 150, 214,
L-arginine 3-7, 11-14, 18-22, 61-67, 92, 214 216
Lactate 91, 93, 104, 106, 110-112, 117, 127,
136, 137, 139, 140, 143, 161, 162, R
176-178, 180, 181, 213, 214
Left ventricular function 14, 18, 62, 63, 148, Remote myocardium 198, 200
149, 160, 162, 163, 180, 181, 197, 201, Reperfusion 2-4, 6-15, 17-22, 28-32, 34-38,
206 41-44, 46, 47, 49-57, 61-67, 70-75, 77,
Leucine 80, 82, 94, 140 79-82, 84-87, 90, 91, 93-95, 99, 100,
102, 106, 107, 109, 113, 115, 117,
M 125-127, 129, 130, 135, 136, 139-142,
145, 150, 160-163, 175-178, 184-188,
Malate 90-92, 140, 141 191, 193-195, 197, 199-201, 211-217,
Malate-aspartate shuttle 91, 140, 141 220-222
Membrane integrity 91, 95 Reperfusion injury 2-4, 9-11, 21, 22, 28-31,
Metabolic support 136, 144-148, 150, 151 34, 36-38, 53, 56, 61-67, 91, 93, 95, 99,
Mitral valve operation 186, 195 100, 135, 160, 163, 184, 185, 193, 214,
Myocardial ischemia 41, 42, 44-47, 53, 54, 215, 217
61, 65, 70-72, 79, 80, 93, 94, 99, 104, Retrograde cardioplegic infusion 205
126, 135, 136, 140, 142, 144-146, 150, Retrograde delivery 184, 188, 189, 191-194
151, 163, 169, 197, 199, 215
Myocardial metabolism 136, 137, 142, 146, S
147, 150, 151
Sarcoplasmic reticulum 160, 161
N Sternal re-entry 204-206, 208
Succinate 82-85, 87, 90-92
Necrosis 2, 8, 10, 14, 21, 28-32, 34-38, Superoxide radical 6, 19, 21, 61, 64, 65, 215
41-45, 49, 54, 63, 71, 73, 102, 125, 127, Supplementation 11, 12, 18, 83, 85, 91-93,
128, 131, 175, 176, 179, 198, 206, 212 113, 185, 214
Neutrophils 2, 3, 9-11, 15, 18, 21, 22, 29, 37,
53, 62, 63, 98, 129, 212, 215 T
Nitric oxide 2-5, 7, 10, 12, 13, 15, 18-21, 61,
65, 114, 141, 163, 214, 218, 219 Tepid cardioplegia 160-163
Nitric oxide synthase 3-5, 18, 20 Transamination 81, 82, 84, 91, 140
TUNEL-assay 42, 44
228 Ischemia-Reperfusion Injury in Cardiac Surgery

V W
Valine 94, 140 Warm induction 185-187, 192, 193
Vascular endothelium 2-4, 7-9, 11 Warm reperfusion 187, 193-195
Ventricular function 160, 162, 163

You might also like