a langendorff-perfused mouse heart model for … a langendorff-perfused mouse heart model for...

121
A Langendorff-Perfused Mouse Heart Model For Delayed Remote Limb Ischemic Preconditioning Studies By Sagar Rohailla A thesis submitted in conformity with the requirements for the degree of Masters of Science Institute of Medical Science University of Toronto © Copyright by Sagar Rohailla (2012)

Upload: vankhuong

Post on 18-May-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

A Langendorff-Perfused Mouse Heart Model For Delayed Remote Limb Ischemic Preconditioning Studies

By

Sagar Rohailla

A thesis submitted in conformity with the requirements for the degree of Masters of Science

Institute of Medical Science University of Toronto

© Copyright by Sagar Rohailla (2012)

ii

A Langendorff-Perfused Mouse Heart Model for

Delayed Remote Limb Ischemic Preconditioning Studies

Sagar Rohailla

Master of Science

Institute of Medical Science University of Toronto

2012

Abstract Remote ischemic preconditioning (rIPC) through transient limb ischemia induces potent

cardioprotection against ischemia reperfusion (IR) injury. I examined the delayed phase of

protection that appears 24 hours after the initial rIPC stimulus. The primary objective of this

study was to establish a mode of sedation and control treatment for delayed rIPC experiments. I

used an ex-vivo, Langendorff isolated-mouse heart preparation of IR injury to examine the

delayed effects of an intra-peritoneal (IP) injection, sodium-pentobarbital (SP), halothane and

nitrous oxide (N2O) anesthesia on post-ischemic cardiac function. Each anesthetic method

improved left-ventricular function after IR injury. SP and halothane anesthesia also reduced LV

infarct size. Delayed cardioprotection after IP injections was associated with an increase in

phosphorylated-Akt levels. The present study shows that IP injections and inhalational anesthesia

invoke cardioprotection and, therefore, indicates that these modes of sedation should not be used

as control treatments for studies examining the delayed rIPC phenotype.

iii

Acknowledgements

First and foremost I would like to thank my supervisor, Dr. Christopher Caldarone for his

support throughout this project. I am grateful for all your encouragement and guidance, and for

never hesitating to open your door for a chat. Your commitment to ensuring success among your

students and your ability to remind us of the bigger picture has always been reassuring. These are

qualities I hope to emulate in my future.

I would like to thank Dr. Andrew Redington for being a mentor and pillar over the last

year. Thank you for believing in me and for giving me the opportunity to develop as a scientist.

Working with you has had an immeasurable positive impact on me. I am enormously grateful for

the experience.

I would like to thank my committee members, Dr. Gregory Wilson and Dr. John Coles

for your insight and advice throughout the project. I hope we can work together again in the

future. To Dr. Edward Hickey, thank you for being a part of this experience and for connecting

me with the field of endotoxin preconditioning. Your PhD thesis was a friendly vision into the

world I hope to enter.

There are no adequate words to describe the support I received from Dr. Jing Li, Dr. Can

Wei and Dr. Xiao Jing Dai. Thank you Dr. Li and Dr. Wei for introducing me to the complexity

and beauty of the mouse Langendorff. Thank you for all of the hearts you mounted and cardiac

function data you helped me to collect. This project was possible because of your hard work and

generosity. I would also like to thank Alex Di Battista for all the support and numerous chats – it

has been extremely useful and fun bouncing ideas.

Lastly, thank you to Kimberly Elias, friends and my family. You all have been the best

academic counselors, companions and sources of support. I am truly thankful to have you in my

life.

iv

Table of Contents

Acknowledgements iii Table of Contents iv List of Tables vi List of Figures vii List of Appendices ix List of Abbreviations x Chapter 1: Review of Literature 1

1.1 Introduction: Unexpected Findings 1 1.2 Ischemia Reperfusion Injury 5 1.3 Inflammation and IR Injury 6 1.4 Remote Ischemic Preconditioning 8

1.4.1 Mechanism of rIPC 10 1.5 Reperfusion Injury Salvage Kinases (RISK) 12 1.6 Delayed Preconditioning 15

1.6.1 Mechanism of Delayed Preconditioning 16 1.7 Current models of 2W Preconditioning 17 1.8 Anesthetic Preconditioning 20

1.8.1 Effects of Anesthesia on Heart Function 21 1.8.2 Mechanism of APC 22 1.8.3 Isoflurane 24 1.8.4 Halothane 26 1.8.5 Sevoflurane, Enflurane and Desflurane 27 1.8.6 Nitrous Oxide 27 1.8.7 Intra-peritoneal Anesthesia: Ketamine and Barbiturates 28

1.9 Anesthetic and Ischemic Preconditioning: Clinical Utility 30 1.10 Langendorff Isolated Heart Model of IR Injury 31

Chapter 2: Research Aims and Hypotheses 33

2.1 Summary and Rationale 33 2.2 Research Aims/Objective 34 2.3 Hypotheses 34

Chapter 3: Methods 35

3.1 Ethics 35 3.2 Experimental Groups 35 3.3 Induction of rIPC Using Inguinal Tourniquet Model 38

v

3.4 A Langendorff Isolated Heart Model of Ischemia-Reperfusion Injury 39 3.5 Infarct Size Determination 43 3.6 Protein Concentration Determination 44 3.7 SDS-Page and Western Blot Analysis 45 3.8 Data and Statistical Analysis 47

Chapter 4: Results 48

4.1 The Delayed Effects of Intra-Peritoneal and Inhalational Anesthesia on Left-Ventricular Function after Global Ischemia 48 4.1.1 Baseline Function 48 4.1.2 Left Ventricular Developed Pressure 50 4.1.3 Left Ventricular End-Diastolic Pressure 55 4.1.4 Maximum Rate of Contraction 60 4.1.5 Maximum Rate of Relaxation 65

4.2 Delayed Preconditioning with Intra-Peritoneal and Inhalational Anesthesia Reduce Infarct Size after IR Injury 69

4.3 Delayed Preconditioning with Injectable and Gas Anesthesia increase phospho-Akt and phospho-p44/42 MAPK Expression 72

Chapter 5: Discussion 76

5.1 Intra-Peritoneal Injections Induce Delayed Preconditioning Against Global IR Injury 78

5.2 Halothane Anesthesia Induces Delayed Preconditioning Against Global IR Injury 82

5.3 Nitrous-Oxide Improves Post-Ischemic Cardiac Performance But Does Not Reduce Infarction Size 83

5.4 Cross-talk between signaling cascades 86

Chapter 6: Conclusions 88 Chapter 7: Future Directions 90

7.1. An In-Vivo Model of Delayed rIPC 91 7.2. Revisiting the role of TLR4 in delayed rIPC 92 7.3. The ‘Third’ Window And Exercise Preconditioning 92 7.4. Clinical Implications of A Mouse Model of Delayed rIPC 93

Chapter 8: References 95 Chapter 9: Appendices 109

vi

List of Tables

1. Baseline functional parameters in the Langendorff-isolated heart groups for IR injury

experiments 49

vii

List of Figures

1. Initial study examining the role of TLR4 in Delayed Preconditioning 4

2. Inflammatory response during IR injury: feed-forward cycle 9

3. Schematic representation of mechanisms involved in remote ischemic preconditioning

11

4. Cell signaling mechanisms involved in early and delayed ischemic preconditioning 14

5. The transition between the early and delayed phases of cardioprotection 18

6. A schematic of the study protocol 37

7. Remote ischemic preconditioning (rIPC) via transient ischemia of mouse hindlimb 38

8a. The mouse Langendorff-isolated heart model 40

8b. The mouse Langendorff-isolated heart model used in The Redington Lab 41

9a. The effects of intra-peritoneal anesthesia on post-ischemic left-ventricular developed

pressure (LVDP) 52

9b. The effects of inhalational anesthesia on post-ischemic left-ventricular developed pressure

(LVDP) 53

9c. Left-ventricular developed pressure (LVDP) at 60 min of reperfusion 54

10a. The effects of intra-peritoneal anesthesia on post-ischemic left-ventricular end-diastolic

pressure (LVEDP) 57

10b. The effects of inhalational anesthesia on post-ischemic left-ventricular end-diastolic pressure

(LVEDP) 58

10c. Post-ischemic left ventricular end-diastolic pressure (LVEDP) at 60 minutes of reperfusion

59

11a. The effects of intra-peritoneal anesthesia on post-ischemic rate of LV contraction (dP/dtmax)

62

viii

11b. The effects of inhalational anesthesia on post-ischemic rate of LV contraction (dP/dtmax)

63

11c. Post-ischemic rate of LV contraction (dP/dtmax) at 60 minutes of reperfusion 64

12a. The effects of intra-peritoneal anesthesia on post-ischemic rate of LV relaxation (dP/dtmin)

66

12b. The effects of inhalational anesthesia on post-ischemic rate of LV relaxation (dP/dtmin)

67

12c. Post-ischemic rate of LV relaxation (dP/dtmin) at 60 minutes of reperfusion 68

13. The effects of intra-peritoneal and inhalational anesthesia on left-ventricle infarct size after

IR injury 70

14. Representative cross-sections of mouse hearts from each treatment group

after IR injury 71

15. The effects of intra-peritoneal and inhalational anesthesia on phospho-Akt (Ser473) levels in

mouse heart before IR injury 74

16. The effects of intra-peritoneal and inhalational anesthesia on phospho-p44/42 MAPK

(Tyr202, Thr204) levels in mouse hearts before IR injury 75

ix

List of Appendices

1. Power-lab acquisition software: Cardiac output chart 110

2. Protein extraction-lysis-buffer 110

x

List of Abbreviations

1W First Window 2W Second Window 5-HD 5-Hydroxydecanoate Akt Serine Threonine Kinase- Protein Kinase B AMI Acute Myocardial Infarction AP-1 Activator Protein-1 APC Anesthetic Preconditioning AR Aldose-Reductase 1 ATP Adenosine Triphosphate Ca2+ Calcium Ion CaMKII Ca2+/calmodulin-dependent kinase II CAO Coronary Artery Occlussion COX-2 Cyclooxygenase-II CPB Cardiopulmonary Bypass DAMPs Danger-Associated Molecular Patterns dp/dtmax Maximum Rate of Contraction dp/dtmin Maximum Rate of Relaxation ECL Enhanced Chemiluminescence eNOS Endothelial Nitric-Oxide Synthase GPCR G-Protein Coupled Receptor GSK3β Glycogen Synthase Kinase 3β H+ Hydrogen Ion HMGB High Mobility Group Box 1 HO-I Heme-Oxygenase I HRP Horseradish Peroxidase Hsp27 Heat Shock Protein 27 kDa Hsp70 Heat Shock Protein 70 KDa IGF-1 Insulin Like Growth Factor-1 ICAM1 Intracellular Adhesion Molecule 1 IFN Interferon Il-6 Interleukin-6 Il-8 Interleukin-8 iNOS Inducible Nitric-Oxide Synthase IPC Ischemic Preconditioning IR Ischemia-Reperfusion K+ Potassium Ion K+-ATP Potassium Ion Adenosine Triphosphate Channel LAD Left Anterior Descending LPS Lipopolysaccharide Endotoxin LVDP Left Ventricular Developed Pressure LVEDP Left Ventricular End-Diastolic Pressure LVP Left Ventricular Pressure MAC Minimum Alveolar Equivalent MAO Mesenteric Artery Occlusion MAPK Mitogen Activated Protein Kinase

xi

MnSOD Manganese Superoxide Dismutase mPTP Mitochondrial Permeability Transition Pore Na+ Sodium Ion NF-κB Nuclear Factor-Kappa B nNOS Neuronal Nitric-Oxide Synthase NO Nitric Oxide N2O Nitrous Oxide ORB Orthodontic Rubber Band PAGE Polyacrylamide Gel-Electrophoresis PAMPs Pathogen-associated molecular patterns PC Preconditioning PI3K Phosphoinositide-3 Kinase PKA Protein Kinase A PKC Protein Kinase C PKG Protein Kinase G PLN Phospholamban Protein PRR Pattern recognition receptor PTK Protein Tyrosine Kinase rIPC Remote Ischemic Preconditioning RISK Reperfusion-Injury Salvage Kinases ROS Reactive Oxygen Species SDS Sodium Dodecyl Sulphate SEM Standard error of the mean SERCA2a Sarcoplasmic Reticulum Calcium ATPase 2a SNAP S-nitroso-N-acetylpenicillamine SNS Sympathetic Nervous System SP Sodium Penotobarbital STAT1 Signal Transducers and Activators of Transcription 1 TNF-α Tumor Necrosis Alpha VCAM1 Vascular Cell Adhesion Molecule 1

1

Chapter 1 Review of Literature

1.1 Introduction: Unexpected Findings

Our laboratory is interested in the biology of myocardial remote ischemic preconditioning

(rIPC), an innate form of organ protection whereby brief episodes of ischemia with intermittent

reperfusion to an organ (e.g., limb) can protect the heart against subsequent lethal ischemia-

reperfusion (IR) injury. We are conducting a range of studies examining the potential triggers

released from the remote organ to the activation of intracellular signaling pathways at the heart

mediating organ survival.

The initial objective of this study was to examine the role of innate immunity in the

development of cardioprotection after rIPC. A general feature of ischemic preconditioning (IPC)

involves suppressing inflammation during IR injury. Previous work in the lab has shown that

rIPC modifies gene expression in mouse myocardium and human neutrophils towards an anti-

inflammatory portfolio[1], [2]. We have also observed that repeated rIPC inhibits human

neutrophil activity by decreasing adhesion and phagocytosis[3-6]. These findings are similar to

those produced in other labs that have observed a preconditioning-induced systemic anti-

inflammatory response [7-12].

While it is well established that inflammation is a main contributor to the damaging

effects of IR injury, emerging evidence indicates that it may also be required to generate

cardioprotection. For example, mice deficient in tumor-necrosis alpha (TNF-α) or nuclear factor

kappa-B (NF-κB), key factors in the inflammatory response, do not develop ischemic tolerance

following a preconditioning stimulus[3], [7], [8], [13]. Further evidence for the role of an

inflammatory response in preconditioning is based on findings that exogenous administration of

sub-lethal doses of a variety of pro-inflammatory cytokines (TNF-α, Interleukin-6, 12 - IL-6,12)

2

or bacterial lipopolysaccharide (LPS) endotoxin induce cardioprotection through similar

mechanisms as IPC, particularly in the late or delayed phase of protection (24 hours after the

preconditioning stimulus)[14-18]. These studies found that the cytokine-induced cardioprotection

occurred in-part through activation of toll-like receptor 4 (TLR4), an integral component of

innate immunity[19-23]. Toll-like receptors are classified as pattern recognition receptors (PRR)

and are integral for orchestrating the initial steps of the innate immune response to exogenous

pathogens by binding to pathogen-associated molecular patterns (PAMP) present on microbial

species. However, TLRs are also important for initiating host immune responses to non-

microbial challenges such as hypoxia, ischemia, heat shock and sepsis. Given the importance of

TLRs in inflammation, a process also involved in generating preconditioning, I aimed to

investigate whether TLR4-signaling plays a role in IPC.

There are some studies that have already addressed this question. In a study examining

the effects of early phase (15 min after the preconditioning stimulus) IPC, cardioprotection was

intact in a TLR4-deficient strain[12], [24-26]. This finding aligns with previous studies on

endotoxin or cytokine preconditioning that TLR4-mediated protection occurs in the delayed

phase[27-30]. Pradillo et al. investigated the role of TLR4 in delayed preconditioning against

stroke injury. They discovered that TLR4-deficiency was associated with a decrease in the

magnitude of protection when compared to wild-type controls[20]. This was also associated with

reduced NF-κB activity and lowered expression of TNF-α. Further evidence for a role of TLR4

in preconditioning comes from findings generated by our lab in which it was observed that there

is a decrease in TLR4 gene expression in the early and delayed phases of protection [1], [2].

Based on these findings, I set out to test the hypothesis that cardioprotection arising from

delayed rIPC would be diminished in TLR4-deficient mice. I proposed that rIPC conferred

protection by inducing a mild-inflammatory response involving TLR4 signaling, and that this

3

process was required to initiate the pro-survival pathways mediating protection against lethal IR

injury.

As shown in figure 1, I designed an experiment to investigate whether TLR4-deficient

mice of the C3H/HeJ strain are capable of developing delayed rIPC. However, these experiments

were interrupted after I observed that wild-type mice (C3H/HeN), receiving the standard sedation

method of sodium-pentobarbital (SP) anesthesia via an intra-peritoneal injection as a control

treatment on day 1, also seemed to develop delayed cardioprotection. Using an isolated mouse

heart model, I discovered that this control group had reduced infarction sizes and preserved

cardiac function after global IR injury twenty-four hours after sedation with SP. Furthermore,

these mice that had been treated with SP anesthesia developed a kinase response typically seen

during the early phase of IPC.

The use of SP anesthesia has been established as an appropriate control treatment for

early phase rIPC studies in mice, as it has not been shown to induce cardioprotection when

administered 60-90 minutes prior to IR injury[31], and it was assumed that this would apply to

the delayed phase as well. It is required by animal care committees that mice be sedated with

anesthesia when applying transient ischemia to the limb to induce rIPC. However, as I uncovered

in my preliminary experiments, the mode and type of sedation is critical as there is accumulating

evidence suggesting that preconditioning can occur after exposure to anesthesia (see review by

Weber et al.) [32]. Experiments in this field need to control for these ‘additional’ potential

preconditioning stimuli in order to examine the cardioprotective phenotype specifically afforded

by rIPC. Thus, the primary objective of this study changed to developing a mode of sedation that

does not induce cardioprotection in order to identify and establish a control treatment for future

delayed rIPC experiments. I began examining alternative methods of anesthesia to identify a

4

method of sedation that would not confer delayed cardioprotection. The following dissertation is

a report of those experiments.

Figure 1: Initial study examining the role of TLR4 in Delayed rIPC. C3H/HeN and C3H/HeJ

(TLR4-/-) mice were allocated to receive sodium-pentobarbital anesthesia (SP) ± rIPC (4 cycles

of 5 min ischemia and 5 min reperfusion) on day 1. Mouse hearts were excised twenty-four hours

after treatment (day 2) and mounted on Langendorff preparation for ischemia-reperfusion injury

experiments. 2W – second window/delayed phase, rIPC – remote ischemic preconditioning

5

1.2 Ischemia-Reperfusion Injury

Coronary artery disease is a leading cause of morbidity and mortality throughout the

world[33]. Acute reductions in coronary bloodflow lead to the clinical syndromes of angina and

acute myocardial infarction (AMI). A significant number of these patients also undergo coronary

balloon angioplasty with stenting, or cardiac surgery, and many will experience episodes of peri-

procedural myocardial ischemia. Such events may severely impair myocardial function, delay

effective post-operative recovery and greatly increase the risk for future complications. The

primary aim of treatment for a coronary ischemic event is to quickly restore blood flow to

preserve muscle function and limit post-infarct sequelae [34]. However, paradoxically, restoring

coronary flow to occluded regions causes pronounced tissue damage in a process known as

reperfusion injury[35-37].

The discovery of reperfusion injury in 1960 by Jennings et al., and the advent of ischemic

preconditioning (IPC) over two decades later provided a better understanding of the mechanisms

involved in ischemia-reperfusion (IR) injury [35], [38]. These important early discoveries helped

to elucidate the independent contributions of ischemia and reperfusion to resulting organ injury,

and also provided an array of targets for pharmacological-based cardioprotective strategies[35],

[38], [39].

IR injury results in a battery of biochemical, structural, and metabolic changes in the

parenchymal and endothelial cells of the heart that impair contractile capacity and ultimately

may decrease blood flow to other essential organs, such as the brain. During ischemia, obstructed

coronary blood flow causes the affected cells to switch their metabolism towards anaerobic

pathways for energy production[35], [39], [40]. This causes a dramatic shift in ion concentrations

in and out of the cell. Lactate accumulates as a metabolic byproduct from pyruvate oxidation,

increasing hydrogen (H+) ion concentration within the cytoplasm. The ensuing acidosis causes

6

sodium (Na+) and calcium (Ca2+) ion levels to increase in an attempt to reestablish normal

physiological pH[35], [41], [42]. Active transport mechanisms, normally able to correct such ion

imbalances, are non-functional due to depleting adenosine-triphosphate (ATP) levels.

Upon reperfusion, extracellular pH is rapidly normalized, forcing the extrusion of H+ ions

from within the cell via the sarcolemmal Na+/H+ exchanger which drives a further increase in

Na+ and Ca2+ ions[36], [43]. Ca2+ overload is a crucial step in the pathogenesis of IR injury[37].

Ca2+ buildup causes hypercontracture of cardiomyocyte myofibrils, activates Ca2+-dependent

proteases involved in apoptosis, and triggers the formation of the mitochondrial permeability

transition pore (mPTP) – a nonselective channel between the inner mitochondrial membrane and

the sarcoplasm[35], [44].

The mPTP is a key component in the pathology of IR injury, as its formation initiates the

eventual death of cells within the infarct zone. Inhibiting pore formation has been a major focus

of many pharmacological interventions aimed at reducing the severity of IR injury[33].

Permeabilization causes the breakdown of oxidative phosphorylation and further exhausts ATP

levels[33]. Furthermore, the mitochondria serve as a reservoir for reactive oxygen species (ROS),

which upon release can overwhelm cellular anti-oxidant defenses, promote the formation of

mPTPs in surrounding cardiomyocytes, attract circulating leukocytes and activate pro-

inflammatory pathways within the affected cells – all of which promote cell death and local

organ injury[41], [45].

1.3 Inflammation and IR Injury

A pronounced inflammatory response is a major contributor to reperfusion injury. The

endothelium plays a large role in orchestrating this process by facilitating the infiltration of

inflammatory cells to the injury site[44]. In response to accumulating ROS, endothelial cells

7

increase the expression of cell-adhesion molecules (intracellular adhesion molecule-1, ICAM-1,

vascular cell adhesion molecule-1, VCAM-1) and release chemoattractants to recruit cells of the

innate immune system [45], [46]. Inflammation during IR injury is mediated initially by tissue

residing macrophages and later by infiltrating neutrophils[42], [44], [47]. Neutrophils migrate to

the region within the first 6 hours from the onset of reperfusion, and invade the myocardium over

the next 24 hours [47], [48]. Mobilization of inflammatory cells is accelerated by the marked

levels of cell necrosis that occurs during IR injury due to shifts in cell osmolality that induce

swelling and rupture [34], [49].

Necrosis liberates a plethora of intra-cellular molecules, often referred to as danger-

associated molecular patterns (DAMPs), that can serve as potential ligands for Toll-like receptors

(TLRs) present on innate immune cells, endothelium and cardiomyocytes[36], [42], [50-52].

DAMPs can include high-mobility group box-1 (HMGB1), heat shock proteins (Hsps), uric acid,

S100 proteins, hyaluronic acid, and oxidized lipoproteins [36], [51]. Cells that are undergoing

necrosis release their internal components into the extracellular space where they bind to and

activate TLRs present on macrophages and neutrophils[36], [53]. TLR signaling initiates a

cellular stress response involving nuclear localization of NF-κB and the transcription of genes

coding for pro-inflammatory cytokines (TNF-α, IL-6, IL-8) [46], [50], [53-55]. Activation of

TLRs on circulating neutrophils, causes the release of degradative enzymes, ROS, and other

cytokines that further exacerbate injury and promote continued DAMP release [36], [46], [53-

55]. This ultimately results in a feed-forward cycle of tissue injury (figure 2)

8

1.4 Remote Ischemic Preconditioning

Despite a greater understanding of the mechanisms underlying IR injury, interventions

aimed at improving clinical outcomes after acute myocardial infarction (AMI) or perioperative

IR injury have been suboptimal. This may largely be due to the likelihood that most interventions

target a specific component of IR injury, whereas the pathology of this disease involves a large

portfolio of destructive pathways. Furthermore, the heterogeneity of IR injury in a clinical setting

and the existence of co-morbidities in patients provide additional challenges in developing a

panacea treatment[56], [57].

However, over the last two decades it has become apparent from numerous studies that

exposing organs to brief periods of non-lethal ischemia, with intermittent reperfusion, termed

ischemic preconditioning (IPC), can protect against future lethal IR injury and limit irreversible

tissue damage. This seminal observation first made by Murry et al in 1986, in which canine

hearts that underwent local coronary IPC had nearly a 75% reduction in myocardial infarct size,

was later expanded to include endogenous protection derived from transient ischemia to distant

organs, known as remote IPC (rIPC) [37], [38], [58]. Initial experiments described rIPC of

myocardium through renal[59], [60], cerebral[61] or mesenteric artery occlusion[62-64] –

procedures that are limited in a clinical setting because of the need for invasive surgery. It was

later discovered in animal models that protection against a sustained coronary artery occlusion

(CAO) could be generated following transient ischemia of rat or rabbit skeletal muscle [65], [66].

In 2002, the lab group described a simpler, non-invasive method of inducing rIPC by applying 3-

4 cycles of transient ischemia and reperfusion to a limb using a standard blood pressure cuff in

patients or tourniquet in animals[67].

Figure 2: Inflam

matory R

esponse During IR

Injury: Feed-Forward C

ycle: 1. Ischemia causes the activation of N

uclear-Factor kappa B (N

F-κB) and

increases the release of reactive oxygen species (RO

S). 2. Mitochondrial perm

eability transition pore (mPTP) form

ation causes further release of RO

S initiating cell death by necrosis or apoptosis. 3. Liberated endogenous danger-associated m

olecular patterns (DA

MPs) act on toll-like receptors (TLR

s) present on infiltrating neutrophils. 4. TLR

activation causes neutrophils to release pro-inflamm

atory cytokines that upregulate cell-adhesion molecules on vessel endothelial

cells (intracellular cell-adhesion molecule, vascular cell adhesion m

olecule – ICA

M, V

CA

M, P-selectins) to facilitate neutrophil invasion into tissue. 5.

Neutrophils release R

OS and cytokines that act on TLR

s present on cardiomyocytes to re-initiate cycle[33], [34], [36], [42].

10

rIPC through limb ischemia has been shown to invoke a similar magnitude of protection

compared to local coronary IPC, and because of its ease of application, can realistically be

translated into clinical practice[68], [69].

1.4.1 Mechanism of rIPC

Since its discovery, there have been major advancements in our understanding of the

mechanisms underlying inter-organ protection. It is well established that rIPC activates a similar

intracellular signaling response as local IPC[49], [68], [70]. Transient ischemia of an organ

causes the release of endogenous substances from cells, such as adenosine, opioids, bradykinin,

and NO, which can enter the circulation and travel to the distant organ or act on nearby afferent

nerves[71] (figure 3). In either case, these preconditioning triggers can bind to their respective

receptors that are coupled to G proteins or directly activate intracellular mediators of

preconditioning [41], [49], [72], [73]. Activation of G-protein coupled receptors (GPCRs)

induces an intracellular signaling response involving a cascade of pro-survival kinases that

results in cardioprotection (figure 4). While the preconditioning phenotype encompasses

numerous proteins, a key step of the signal involves the activation of phosphatidylinositol-3-

kinase (PI3K) and its associated serine-threonine kinase (Akt) and the mitogen activated protein

kinase (MAPK) family, specifically the extracellular regulated kinases 1/2 (Erk 1/2) [74], [75].

While there is still some debate regarding the putative end effectors of preconditioning, it

is generally accepted that protection involves (1) limiting Ca2+ overload, (2) maintaining an open

state of sarcolemmal and mitochondrial K+-ATP sensitive channels, and (3) inhibiting the

formation of the mPTP[71]. As previously mentioned, Ca2+ overload initiates the process that

ultimately disables cell function and leads to death by necrosis or apoptosis.

11

Figure 3. Schematic representation of mechanisms involved in remote ischemic

preconditioning. Transient ischemia to the limb causes the release of endogenous substances

that can either enter blood stream, activated nearby afferent neurons or act on circulating

inflammatory cells. This leads to the activation of GPCRs on the myocardium to recruit intra-

cellular pathways that result in cardioprotection against infarction[69], [71].

12

Maintaining intracellular ion concentrations can ameliorate this destructive process. The

role of K+-ATP channels in limiting cell damage during IR injury is not completely understood,

yet they may be involved in reducing Ca2+ overload and stabilizing mitochondrial structure and

function, and thus maintaining ATP generation[43], [71], [76].

Additionally, IPC is known to inhibit circulating neutrophil function and causes a shift

towards the production of anti-inflammatory and anti-apoptotic mediators within the

myocardium[2], [3], [21], [22], [77]. A similar response also occurs with sub-lethal doses of pro-

inflammatory cytokines or endotoxin. Some studies suggest that cytokine preconditioning

involves suppression of the inflammatory response via negative feedback inhibitors of TLR

signaling[2], [3], [21], [22], [77]. This is thought to involve the PI3K-Akt signaling pathway

[17], [78], [79]. Following an increase in NF-κB activity, the p85 subunit of PI3K is recruited to

the cytoplasmic domain of TLR4 to modulate its activity (figure 4). This inhibits the TLR4 pro-

inflammatory pathway and initiates the pro-survival signaling pathways mediated through

Akt[17], [78]. Negative feedback on TLR4 may also be involved in rIPC-induced immune

suppression, but this remains to be studied.

1.5 Reperfusion-Injury Salvage Kinases (RISK)

In 2005, the Yellon group provided strong evidence that IPC involves the activation of a

set of kinases that collectively mediate cardioprotection against IR injury. These enzymes have

been termed the reperfusion-injury salvage kinases (RISK) and their activation has been

consistently associated with reduced cell death after IR injury[75], [80]. The RISK pathway is

also a convergence point for many forms of pharmacological preconditioning, as has been shown

in a number of preclinical studies evaluating the cardioprotective effects of insulin, glucagon-like

peptide 1, erythropoietin and adenosine (reviewed in Hausenloy & Yellon 2007)[81]. The Akt

13

and Erk1/2 kinases are important elements of the RISK pathway and can be considered markers

for the preconditioning phenotype, as they play a critical role in the cellular basis of IPC that

ultimately enhances cell survival.

Akt – Protein Kinase B

PI3K activation of Akt leads to the phosphorylation of several downstream targets

responsible for preconditioning the myocardium, some of which include endothelial nitric oxide

synthase (eNOS)[80], [82], [83], inactivation of glycogen-synthase kinase-3β (GSK-3β)[84] and

pro-apoptotic signaling proteins (caspases, Bad, Bax)[70], and the different isoforms of protein

kinase C (PKC)[85], [86] (figure 4). Akt is activated by the inositol lipid byproduct of PI3K, a

pathway that is well characterized in insulin-like growth factor-1 (IGF-1) signaling, promoting

cell survival and protein synthesis, while limiting apoptosis.

A number of studies indicate that Akt is activated after the preconditioning phase and is later

phosphorylated during reperfusion with antecedent IPC[75], [86], [87]. The Redington Lab

provided strong evidence for Akt activation after rIPC in a study examining the effects of four

cycles of unilateral hindlimb ischemia on cardiac function and infarction size in isolated mouse

hearts. It was found that hind limb ischemia led to nearly a two-fold increase in phosphorylated

–Akt levels in mouse hearts, which was also associated with improved cardiac recovery and

reduced infarction after IR injury. These cell signaling and cardiprotective effects of rIPC were

abrogated when co-adminstered with the PI3K inhibitor, wortmannin, indicating a critical role

for PI3K-Akt signaling [31].

14

Figure 4: Cell signaling mechanisms involved in early and delayed ischemic

preconditioning. 1. Early preconditioning. Initial triggers released into circulation from

remote tissue may include, GPCR agonists or endogenous DAMPs stimulating TLR4.

Intracellular mediators involve the PI3K-Akt pathway acting on downstream kinases, most

notably PKC. End-effectors include mitochondrial and sarcolemmal K+ATP

channels. 2. Delayed

preconditioning. Nuclear translocation of NF-κB and subsequent transcription of a battery of

genes leads to the development of delayed preconditioning[22], [32], [43], [59], [68], [88].

15

Extracellular Regulated Kinase (Erk1/2) – MAPK p44/42

The Erk 1/2 or p44/42 proteins are members of the MAPK protein family responsible for

regulating a number of proliferative pathways that are important for cell survival[81]. Similar to

Akt, p44/42 activation via GPCR signaling, or through PKC-espilon, occurs after

preconditioning and results in the inhibition of pro-apoptotic enzymes that normally orchestrate

cell death processes that occur during reperfusion[89], [90]. A number of studies have implicated

phosphorylated-p44/42 in IPC as a key element of the RISK pathway[75], [81], [89]. Further

studies would also confirm a role for MAPK in rIPC. Heidbreder et al. performed a study

comparing the role of p44/42 signaling in cardioprotection after both IPC and rIPC. They

observed an increase in phospho-p44/42 in rat hearts after preconditioning via transient coronary

artery occlusions (CAO) or mesenteric artery occlusion (MAO)[89]. Shimizu et al. observed a

similar increase in phospho-p44/42 levels after rIPC via transient limb ischemia in rabbits[85].

They found that rabbits subjected to four-cycles of hind limb ischemia with intermittent

reperfusion showed an increase in myocardial levels of phospho-p44/42, which was associated

with reduced infarction sizes in in-vivo and ex-vivo isolated hearts.

1.6 Delayed Preconditioning

A unique property of the conditioning response is that the target organ possesses the

ability to regain the preconditioned state long after the initial stimulus. In both local and remote

IPC, there is an initial phase of protection arising within minutes that can last for up to 2-4 hours,

termed a first window (1W) or early preconditioning response[77]. The early phase relies on the

activation of pro-survival signaling pathways mostly through post-translational modification of

existing proteins within cells[91]. Shortly after the discovery of rIPC, it was revealed that the

protective phenotype reappears 12-24 hours later, known as the second window (2W) or delayed

16

preconditioning response[92],[93]. The delayed phase is less robust but may have greater clinical

utility given its broader range and longer duration of protection (up to 3-4 days)[94]. Delayed

preconditioning has also been observed to limit post-ischemic myocardial stunning, a period in

which the heart’s contractile capacity is below pre-ischemic values[34]. Interestingly, recent

studies suggest that a third or chronic preconditioning effect may also exist with repeated

exposure to transient ischemia, operating through an entirely different signaling response to that

of both early and delayed preconditioning[95].

1.6.1 Mechanism of Delayed Preconditioning

Whether through non-pharmacological (ischemia, heat stress, exercise) or

pharmacological (adenosine/opioid receptor agonists, NO, ROS, LPS, TNF-α) stimuli, delayed

preconditioning develops through similar signal transduction pathways responsible for early

local and remote IPC[49]. The differences in the protective phenotype lie in the end-effectors

(figure 5). While early preconditioning involves protein modification, the late phase is largely

mediated by gene induction and de novo synthesis of cardioprotective proteins[91], [96]. The

prevailing hypothesis for delayed preconditioning is that the intracellular mediators of the early

response, most notably PKC-ε[97], protein tyrosine kinase (PTK)[98] and p44/42 MAPK, lead to

increased transcriptional activity of NF-κB, activator protein-1 (AP-1) and signal transducers and

activators of transcription 1 (STAT1) [9], [88], [99]. There is also some evidence to suggest that

Akt activation can also lead to an increase in NF-κB transcriptional activity[100]. These

activated transcription factors migrate to the nucleus and increase the expression of genes

responsible for delayed preconditioning.

In addition to its well-known detrimental role in inflammation, NF-κB modulates a host

of genes involved in the cell stress response to a variety of pathological stimuli[54]. It is

17

generally accepted that NF-κB signaling is obligatory for the development of delayed

preconditioning[13],[77]. In its quiescent state, NF-κB remains in the cytoplasm bound to its

inhibitor protein, I-Kappa B (IκB) that masks it nuclear localization sequence. When the cell is

under stress, such as following a preconditioning stimulus, IκB is degraded allowing NF-κB to

translocate to the nucleus, most often as a heterodimer consisting of the p50 and p65 subunits.

During IR injury, nuclear binding peaks at approximately 15 min and 3 hours after the onset of

reperfusion, likely from exposure to ROS or pro-inflammatory cytokines[46], [54]. However,

there is also an increase in NF-κB promoter binding activity at 30min following preconditioning,

which seems to occur through an increase in nitric-oxide (NO)-induced-PKC-ε activation[13].

NF-κB enters the nucleus and promotes the transcription of a variety of genes shown to be

important for delayed PC, which include heat shock proteins (Hsp70, Hsp27), manganese

superoxide dismutase (MnSOD), aldose reductase (AR), hemo-oxygenase-1 (HO-1)

cyclooxygenase-2 (COX-2) and inducible nitric-oxide synthase (iNOS). Similar to early

preconditioning, the protective phenotype converges on restricting pore formation and

maintaining an open state of mitochondrial and sarcolemmal K+-ATP channels (figure 5) [101],

[101], [102].

1.7 Current Models of Delayed Limb rIPC

Much of our initial understanding of the physiology of myocardial delayed

preconditioning is derived from local IPC studies. The Bolli group has conducted a number of

elegant experiments using a working model of delayed local IPC to characterize the mechanisms

behind the late phase of protection[93],[103]. Their work has provided evidence for the currently

established mediators of delayed preconditioning. In particular, they have demonstrated

18

Figure 5: The transition between the early and delayed phases of cardioprotection.

Transient ischemia causes the release of triggers from tissue that act on cell receptors to activate

intra-cellular pro-survival kinases. Activated kinases act on end-effectors of cardioprotection and

increase the activity of transcription factors promoting gene expression for the resurgent phase of

protection. Adapted from [88], [94], [104]

19

an essential role for cytokine signaling, validating the growing hypothesis that a mild

inflammatory response precedes ischemic tolerance.

The Redington group and many others have demonstrated in numerous studies that transient

limb ischemia provides potent early cardioprotection in animal and human models of IR injury

[4-6], [8-12], [105], [106]. While there have been major strides in elucidating the mechanism of

early rIPC via limb ischemia, evidence of its delayed and chronic preconditioning effects is

sparse. Li et al. provided some initial evidence of delayed rIPC in a mouse isolated heart model

of global ischemia. They found that six 5-minute cycles of hindlimb ischemia intercalated with

periods of reperfusion administered twenty-four hours prior to injury reduced infarct size by

34%. They also discovered that the delayed phenotype was absent in NF-κB or iNOS deficient

mice[7], [8], [13]. However, it is unclear what their control sedation treatment was for limb

ischemia – an important consideration as will be discussed in later sections.

In human studies, 10 days of repeated limb rIPC has been shown to reduce neutrophil

adhesion and phagocytosis activity from day one onwards[3], [15-18]. Furthermore,

Loukogeorgakis et al. found that limb rIPC generated a late phase of protection against

endothelial IR injury, which was blocked with administration of trimetaphan (an autonomic

ganglion blocker) at the time of limb ischemia and reperfusion, indicating the requirement for

intact neural signaling[14], [20-23]. However, some clinical trials have shown no observable

benefit from delayed limb rIPC. In a recent study by Pavione et al., it was shown that four cycles

of 5 min limb ischemia applied a day before children were placed on cardiopulmonary bypass

(CPB) for congenital heart disease surgery had no effect on reducing cardiac troponin I plasma

concentrations, a commonly used surrogate marker of myocardial damage[12], [19].

Part of the difficulty in examining the delayed phase of preconditioning comes from

emerging evidence of a protective effect arising from exposure to anesthesia used during

20

experiments [24-26], [28], [30]. Mice are sedated with intra-peritoneal anesthetics, such as

pentobarbital, in order to apply transient ischemia to the hind limb. This is also done to minimize

the stress of the stimulus. As such, control treatments for rIPC involve sedating mice with

anesthesia without applying unilateral hind limb ischemia. In the Redington lab, this protocol

has been effective for examining the first window or early phase of preconditioning after rIPC, as

pentobarbital does not give rise to early preconditioning in mice[31]. However, the preliminary

data of this study suggest that exposure to an intra-peritoneal injection of pentobarbital does give

rise to delayed cardioprotection. This and additional evidence describing a delayed

preconditioning effect from other forms of anesthesia has limited the effectiveness of studies

specifically examining the biology of delayed limb preconditioning in mice. This may be

because the threshold for eliciting benefits from additional preconditioning stimuli may be

greater after exposure to anesthesia. Furthermore, the recent clinical studies showing no benefits

from rIPC highlight the possibility that certain substances used during surgery may inhibit the

effects of antecedent rIPC[20], [27], [29].

1.8 Anesthetic Preconditioning

Cardioprotection from exposure to anesthesia, known as anesthetic preconditioning

(APC), may be of profound importance in clinical settings as an adjunct that can safely elicit

protection during high-risk cardiac surgery[107], [108]. The beneficial role of anesthesia in

minimizing myocardial injury during IR has been known for decades, described by several

groups in the late 1960s [109], [110]. It was later discovered that exposure to anesthesia could

mimic preconditioning by attenuating post-ischemic damage and improving cardiac functional

recovery [111].

21

The following sections will be provide an overview of the effects of anesthesia on heart

function, the signaling pathways involved in APC and a summary of the accumulated evidence

on inhalational and intra-peritoneal APC. This will provide some background information on

potential anesthetics that may used to develop a model of delayed rIPC.

1.8.1 Effects of Anesthesia on Heart Function

In today’s clinical settings, the currently used inhalational anesthetics include: nitrous

oxide (N2O), halothane, enflurane, isoflurane, sevoflurane and desflurane – all of which (with the

exception of N2O) share the common feature of possessing a halogenated atom on a carbon

backbone[112]. It is well known that inhalational anesthesia causes a dose-dependent depressive

effect on myocardial function[108], manifested through reduced myocardial oxygen demand,

metabolic activity and vasodilation of coronary arteries – all of which may protect the heart

during IR injury[113]. All of the modern volatile anesthetics reduce cardiac output and mean

arterial pressure (mostly via a decrease in systemic vascular resistance) to some extent. This

mainly occurs through changes in Ca2+-handling within the cardiomyocytes at both the

sarcolemmal and sarcoplasmic reticulum (SR) level [114]. Volatile anesthesia decreases calcium

entry through L-type Ca2+ channels, which reduces Ca2+-dependent-Ca2+ release from ryanodine

receptors located on the SR. As a result, intracellular Ca2+ levels fall which reduces cardiac

inotropy and output[108], [110].

Halothane is thought to have the greatest negative effect on cardiac output, but causes

minimal changes in vascular resistance[115]. Conversely, isoflurane has a minimal effect on

cardiac inotropy, but invokes a large decrease in systemic vascular resistance[113], [116].

Interestingly, it has been suggested that isoflurane can cause mild ischemic episodes within the

coronary vasculature by producing varied levels of vasodilation. This process has been termed,

22

“coronary steal, ” which describes how under isoflurane anesthesia, non-diseased vessels may

dilate and ‘steal’ blood flow from vessels incapable of maximal dilation[113]. This may explain

some of the preconditioning-like effects that develop after isoflurane exposure.

Ketamine and sodium-pentobarbital (SP) are commonly used intra-peritoneal anesthetics

in animal models of myocardial IR injury. Similar to volatile anesthesia, ketamine and SP are

also known to depress cardiac function through changes in SR Ca2+ release and uptake and by

reducing extracellular Ca2+ entry [117], [118]. Jiang et al. showed that both ketamine and SP

reduced heart rate, left-ventricle systolic pressure and the rate of contractility in isolated rat

hearts[118]. In the same study, it was observed that SP showed a pronounced respiratory

depressive effect compared to ketamine and chloral hydrate anesthesia. In-vivo, ketamine has

been shown to markedly decrease heart rate and coronary flow and may induce pronounced

hypotension[103].

1.8.2 Mechanism of APC

The signal transduction pathways involved in APC are strikingly similar to those

activated during IPC. APC acts through opioid and adenosine GPCRs, which amplify the initial

signal and lead to the activation of the protective intracellular signalling response[32], [119],

[120]. As it occurs in IPC, the protective phenotype involves activating PKC, maintaining

intracellular Ca2+ ion concentration and stabilizing mitochondrial function via K+-ATP

channels[59], [121]. ROS production from the mitochondria is also integral for APC [110]. The

transition between early and delayed APC is also bridged via NF-κB activation[122], [123],

which upregulates the production of many of the same mediators responsible for delayed IPC,

such as iNOS and NO, MnSOD and Hsps[32]. There even exists some resemblance between

both forms of preconditioning at the genomic level. Sergeev et al. found that both IPC and APC

23

modulate genes involved in cell defense, growth, metabolism and inflammation. This is also

consistent with the changes in gene expression after rIPC[1], [2]. Similarly to IPC, APC

upregulates the expression of Hsps, inhibits proteins from the Bcl-2 family involved in initiating

apoptosis, and also activates NF-κB and its associated downstream products[124].

There are several levels at which APC intervenes to protect the myocardium. As in other

forms of preconditioning, maintaining Ca2+ homeostasis and minimizing overload is of critical

importance to preserving mitochondrial and cell function[59]. The mitochondrial K+ATP channel

has consistently shown to be an important component of the preconditioning phenotype. There is

still some debate regarding the importance of the sarcolemmal potassium channel, although some

studies confirm its involvement in APC[125], [126]. Both in-vivo and in-vitro studies have

shown that administration of 5-hydroxydecanoate (5-HD - a mitochondrial K+ATP channel

antagonist) or HMR-1098 (sarcolemmal K+-ATP channel antagonist) abolishes the benefits from

APC[126]. The mechanism of how K+-ATP channel opening protects the myocardium against IR

injury is not completely understood. It appears that opening of mitochondrial K+ATP channels

depolarizes the mitochondrial membrane, the immediate effect of which limits Ca2+ buildup in

the organelle and in the greater sarcoplasm. Depolarization is believed to initially cause swelling

of the mitochondria and a transient decrease in ATP production, which in turn activates complex

compensatory mechanisms that ultimately optimize oxidative phosphorylation and ATP

generation [59].

A key component of the APC phenotype involves an increase in ROS activity. It is not

entirely clear when the ROS burst occurs during the development of APC, however it is known

that a surge is obligatory as evidenced by loss of cardioprotection with the use of free-radical

scavengers[127]. Transient depolarization of the mitochondrial membrane leads to ROS release

from the mitochondria, which amplify the signaling cascade even further by spreading

24

throughout myocardium through intercellular gap junctions. ROS activate PKC in surrounding

cardiomycytes and ultimately lead to the opening of more mitochondrial K+ATP channels and

further ROS bursts[110].

APC is also known to suppress the inflammatory response during IR injury[128].

Sevoflurane and isoflurane have been shown to precondition isolated rat hearts by inhibiting

neutrophil adherence to cells[129], [130]. This is partly explained by an anesthetic-induced

decrease in neutrophil expression of CD11b adhesion molecules[130]. Several other studies have

shown that pretreatment with isoflurane anesthesia reduces neutrophil superoxide production in

addition to decreasing their adherence to the coronary vasculature [131], [132].

1.8.3 Isoflurane

Exposure to isoflurane anesthesia prior to myocardial IR injury is perhaps the most well

established method of APC. As a more soluble halogenated anesthetic than halothane and

enflurane, isoflurane is commonly used in experimental animal studies, owing to its stability and

quick clearance[116]. In 1988, Warltier et al. produced one of the earliest observations of a

protective effect of anesthesia on the myocardium during IR injury. They discovered that

isoflurane and halothane, when given during ischemia, lead to rapid improvements in post-

ischemic myocardial contractile function in dogs[111]. This set the stage for investigations into

whether isoflurane could mimic IPC. In 1996, Kersten et al. found that 1 minimum alveolar

concentration (MAC – defined as the minimum concentration of anesthesia at 1 atm that prevents

movement in response to stimulation in 50% of subjects[116]) administered for 30 min, thirty

minutes prior to CAO in a canine model, led to an 80% recovery of cardiomyocyte cell

shortening compared to 16% in non-isoflurance treated dogs [133]. This effect was abolished

when isoflurane was co-administered with glibenclamide, a non-specific K+ATP channel

25

antagonist. Further studies have confirmed the preconditioning effects of isoflurane in both in-

vivo [133] and in-vitro isolated heart models of IR injury [107], [134], [135].

Isoflurane preconditioning occurs through similar pathways as local and remote IPC.

Early isoflurane preconditioning involves the activation of PKC-ε, with subsequent maintenance

of mitochondrial K+ATP channels [107], [121], [134]. Other studies have shown that isoflurane

may also activate the PI3K-Akt signaling pathway. Raphael et al. found that 30 min of isoflurane

administration prior to CAO, reduced infarct size in rabbits, and this effect abolished with the use

of the PI3K inhibitors, wortmanin or LY294002 [136]. Inhibition of PI3K was also associated

with a decrease in phospho-Akt levels, and an increase in the pro-apoptotic proteins, Bad, Bax

and caspase-3.

Isoflurane preconditioning is also known to cause a delayed protective effect[122], [137-

140]. Chen et al. found that delayed isoflurane preconditioning in rats occurs through local NO

release, which triggers the PKC-induced increase in NF-κB activity. They discovered that NF-κB

amplifies the initial NO signal through upregulation of iNOS and further NO synthesis, which

then spreads the signal throughout the myocardium [122]. Ultimately, protection manifests by

maintaining an open state of K+ATP channels. Tonkovic-Capin et al. provided evidence for an

essential role for both the sarcolemmal and mitochondrial K+-ATP channels in delayed isoflurane

preconditioning of isolated rabbit hearts. They observed that 1 MAC of isoflurane administered

24 hour prior to global ischemia reduced infarct size and improved left-ventricular develop

pressure recovery, an effect that disappears with the use of either 5-HD or HMR-1098 K+ATP

channel antagonists [138].

26

1.8.4 Halothane

Since its introduction in 1956, halothane has been viewed as an anesthetic with the

greatest cardiodepressant effects, producing a decrease in blood pressure, cardiac output and

heart rate[113], [115], [116], [141]. Halothane is effective at reducing myocardial injury when

applied at the onset of reperfusion [24], [141], [142]. Similar to other anesthetics, halothane

attenuates post-ischemic cardiac dysfunction by reducing Ca2+ cycling and overload[24], [26]. A

halothane-induced anti-inflammatory effect during reperfusion injury has also been described.

Kowalski et al. showed that halothane and isoflurane administration decreased post-ischemic

neutrophil adhesion in isolated guinea pig hearts that received 6x105 neutrophils in the coronary

perfusate prior to ischemia[27].

Despite considerable evidence describing the benefits of halothane during IR injury, its

role as a preconditioning stimulus is still uncertain. Preliminary findings indicate that the

protective effects of halothane anesthesia may be confined to mitigating reperfusion injury.

Roscoe et al. showed that halothane exerted no cardioprotective benefits on human atrial muscle

when given 30 min before anoxia, and may have even inhibited the effects of prior transient

anoxia preconditioning[107]. However, there is some evidence that halothane may exert

cardioprotective benefits[109], [111]. In a study by Piriou et al examining the effects of

halothane preconditioning on rabbit myocardium, it was shown that 30 minutes of halothane

exposure 15 minutes before IR injury reduced LV infarct size compared to untreated

controls[109]. In other systems of preconditioning, such as in the brain, halothane has been the

choice of anesthesia in mice undergoing transient middle cerebral artery occlusion, producing no

demonstrable protection in non-preconditioned controls[143], [144]. To our knowledge, there are

no previous studies describing a role for halothane anesthesia in the development of delayed

tolerance to IR injury.

27

1.8.5 Sevoflurane, Enflurane and Desflurane

Other halogenated anesthetics have been found to exert cardioprotective effects through

similar mechanisms as isoflurane, including the activation of PKC, mitochondrial K+ATP channels

and increased NO bioavailability [141], [145], [146]. Lu et al. showed that a critical step of

sevoflurane-APC involves ROS-dependent activation of NF-κB. In isolated rat hearts, they found

that 2.5% sevoflurane reduced infarct size, preserved cardiac function and reduced pro-apoptotic

protein activation [147]. The importance of ROS-induced Nf-κB activation has been confirmed

in several studies of sevoflurane preconditioning [148].

Additionally, there are several reports of a delayed preconditioning effect with

administration of sevoflurane, enflurane and desflurane. Chiari et al. found that 24 hours after

intravenous administration of emulsified enflurane and sevoflurane, infarct size after CAO was

reduced in APC rabbits by approximately 50% compared to lipid vehicle and saline treated

controls[149]. Furthermore, Lotz et al. found that 1MAC of desflurane administered 24 hours

before CAO, reduced infarction sizes in rabbits. They identified an essential role for peroxisome-

proliferator-activated receptor γ activation and increased levels of NO in desflurane

preconditioned hearts[150].

1.8.6 Nitrous Oxide

Nitrous oxide (N2O) is one of the oldest methods of anesthesia and is still in clinical use

today as a potent analgesic[28], [30], [113]. Nitrous oxide is often used with other halogenated

anesthetics and is known to potentiate their myocardial depressant effects[151]. The nitrous

oxide anti-nociceptive effects are known to be mediated by activation of κ-opioid receptors in the

periaqueductal gray region of the brain[152], [153]. Given the well known role of opioid

28

signaling in preconditioning, it is possible that nitrous oxide may induce cardioprotection by

binding to opioid receptors located on the myocardium[6], [33], [72].

Weber et al. conducted a study to examine whether nitrous oxide could precondition rat

hearts in a coronary artery occlusion (CAO) model of IR injury[154]. Rats received three 5 min

cycles of 60% N2O (with 20% O2 and 20% N2) interspersed with 5-min washout periods prior to

sustained CAO. This was compared this with the effects of APC via N2O+isoflurane delivered in

the same manner. They found that exposure to nitrous oxide provided no protection against IR

injury, as N2O-treated mice displayed similar infarct sizes as untreated controls. Not surprisingly,

isoflurane+N2O reduced infarction sizes by approximately 40%. As such, they concluded that

nitrous oxide might be the first inhalational anesthetic without preconditioning effects[154]. In

another study examining nitrous oxide preconditioning against hypoxia-reoxygenation injury, it

was found that N2O improved LVDP and reduced L-type Ca2+ channel currents in isolated rat

hearts when administered during hypoxia and had no effect when provided as a preconditioning

stimulus [155].

Therefore, it appears that like other inhalational anesthetics, nitrous oxide improves

myocardial function when administered during low oxygen challenges, but may not induce a

cardioprotective phenotype when given prior to lethal IR injury.

1.8.7 Intra-peritoneal Anesthesia: Ketamine and Barbiturates

Ketamine

There is some indication that intra-peritoneal anesthetics may induce or even inhibit the

effects of IPC. Ko et al. discovered that ketamine hydrochloride anesthesia decreases K+ATP

channel activity in a dose-dependent manner in rat ventricular myocytes [156]. Another study

extended this finding by showing that both the sarcolemmal and mitochondrial K+ATP channels

29

are inhibited with exposure to ketamine[157]. These findings suggested that ketamine might also

block the benefits of IPC. Later studies confirm this hypothesis, but showed that the ketamine-

blockade of IPC appears to be isomer specific. A number of studies discovered that treatment

with the optical isomer R(–) ketamine prior to local IPC blocked its infarct sparing effects in an

isolated heart or in-vivo animal model[158], [159]. With regards to delayed IPC, Müllenheim et

al. showed that a racemic mixture of the R and S enantiomers inhibited the cardioprotective

effects of late IPC via transient CAO. In each report, it was shown that S(+)Ketamine had no

effects on the preconditioning stimulus, further establishing that the inhibition is specific to the R

isomer.

Hanouz et al. sought to investigate whether the racemic mixture was in fact blocking

cardioprotection or inducing its own preconditioning-like phenotype[160]. They examined the

effects of racemic and S(+) ketamine on human atrial myocardium subjected to hypoxic injury.

Interestingly, they discovered that exposure to both the racemic and S isomer of ketamine

enhanced contractile recovery after hypoxia when compared to untreated controls[160]. It was

also discovered that these protective effects were abolished with co-administration of 5-HD,

HMR 1098 or α/β adrenergic receptor antagonists. These findings indicate that what was initially

believed to be a ketamine-induced blockade of IPC, may have actually been a preconditioning-

like effect overriding benefits from additional cardioprotective stimuli.

Pentobarbital

Pentobarbital is often the standard method of sedation used for isolated heart or in-vivo

APC experiments. SP has been shown to cause a large decrease in cardiac output, left-ventricle

systolic pressure and heart rate in rats in comparison to ketamine and chloral hydrate anesthesia

[35], [44], [118]. Barbiturates are also known to act as free radical scavengers and thus may limit

30

the damage from the ROS bursts occurring during IR injury. [110]. However, it is evident from a

number of studies that SP does not block the effect of early local and remote IPC in a number of

species [5], [8], [31], [103], [161-165]. The Bolli group has shown that SP anesthesia does not

induce cardioprotection in an early and late mouse model of local IPC[9], [103], [166-168]. Their

initial study in which they developed their model involved pilot work with various forms of

anesthesia and they found that barbiturate anesthetized mice exhibited heart rates closest to

normal physiological levels (avg. 650 beats/min)[103]. They reported that additional

experimental adjustments needed to be made to compensate for the cardiopulmonary depressive

effects of SP in mice, such as the constant monitoring of body temperature and proper tidal

ventilation [103]. Although the Bolli group has shown pentobarbital does not induce

preconditioning during local IPC, an evaluation into the effects of SP during late rIPC has not

been undertaken.

1.9 Anesthetic and Ischemic Preconditioning: Clinical Utility

APC represents a clinically useful and safer method of inducing cardioprotection when

compared to local coronary IPC, eliminating the requirement for invasive surgery that can

damage the coronary vasculature and increase the risk for future complications. The advent of

rIPC by transient limb ischemia overcomes this obstacle. Findings generated by our group and

others demonstrate the powerful clinical benefits of rIPC [69], [105], [106], [169]. In the last

decade, accumulating evidence has shown that making rIPC a part of the clinical management of

AMI can result in reduced patient mortality and morbidity [47], [48], [69], [106].

However, these important findings have met a fair share of conflicting results. Several

recent studies suggest that rIPC may provide no additional benefit against IR injury during

surgery. Rahman et al. conducted a large trial examining the effects of three-5-min cycles of

31

upper arm ischemia in patients undergoing coronary bypass surgery. They found that rIPC

provided no benefits in reducing troponin T levels or in improving cardiac function compared to

placebo sham-treated controls[170]. In a recent clinical trial, Kottentburg et al. found that the

effects of rIPC were negligible when applied with propofol anesthesia[29]. It should be noted

that neutral findings generated in these clinical studies may reflect differences in the settings in

which rIPC is applied. Our lab group is interested in examining the possibility that certain agents

used in patient care may block rIPC – a necessary investigation to further optimize the

preconditioning response.

Nonetheless, the usefulness of APC and the recent neutral trials does not eliminate the

need to better describe rIPC through limb ischemia, as it can be used outside of the clinical arena

where access to anesthesia is not possible. This is especially important when considering rIPC

may benefit cardiovascular function in a number of ways, other than minimizing IR injury. For

instance, our group has shown that rIPC can increase coronary blood flow and lower coronary

vascular resistance[171]. These and other findings highlight that intermittent ischemia may have

potential blood-pressure lowering effects, which could be of profound importance for exercise

therapy in patients with cardiovascular disease[172]. Our lab group has also shown that repeated

transient limb ischemia for 28 days can reduce post-infarct adverse remodeling[173], can induce

a sustained anti-inflammatory phenotype when repeated for several days[1-3] and can even

improve exercise performance in highly-trained athletes[174].

1.10 Langendorff Isolated-Heart Model of IR Injury

Anesthetic preconditioning, both in clinical and experimental settings, adds a challenge to

examining the biology of rIPC beyond the early phase of protection. Given that animals need to

be sedated with anesthesia in order to apply unilateral transient limb ischemia, it is imperative to

32

employ a mode of sedation without cardioprotective effects. This will also serve as part of a

control treatment for delayed rIPC. Based on preliminary findings that intra-peritoneal

pentobarbital, when administered twenty-four hours prior to IR injury, is associated with

cardioprotection in mice, I began to investigate the effects of various inhalational and injectable

anesthetics in order to develop a mouse model of delayed rIPC.

The isolated Langendorff mouse heart model has been an invaluable tool for examining

cardiac physiology and has played an important role in identifying the signaling pathways

involved in preconditioning. It is a highly reproducible preparation that can be operated at a

relatively low cost and provides a wealth of data describing the myocardial response to various

stimuli [175]. Advancing transgenic mouse species are making murine Langendorff models a

frequently used tool for studying basic cardiac physiology. The isolated heart preparation allows

measurement of a broad spectrum of physiological and biochemical parameters during a variety

of challenges, such as IR injury, hypoxia, drug-dose responses, hypo-/hyperthermia and

electrophysiological alterations[176], [177], the main advantage being that the acquired

responses are cardiac specific, devoid of any influence from systemic circulation, neuro-

hormonal or immunological influences.

The small size of the mouse heart makes the isolated-heart model technically challenging

in this species. As a result, initial studies employing this method showed a high degree of

baseline variability in their results. The Headrick group has conducted a number of valuable

studies aimed at characterizing the murine Langendorff model[175], [178]. They have provided

baseline assessments of various cardiac parameters that can be used as normal criteria for the

mouse isolated-heart preparation. Our lab has made use of their experimental approaches and

setups in developing our system.

33

Chapter 2 Research Aims and Hypotheses

2.1 Summary and Rationale

It is clear that further studies are required to improve our understanding of rIPC,

especially relating to its delayed and potential chronic effects. Investigating these additional

phases will provide a more general knowledge base of cardioprotection and help to identify

potential clinical settings in which the intervention can benefit individuals.

However, as with any potential clinical intervention, a sound physiological model must

first be developed to properly understand the mechanisms behind delayed preconditioning. This

is particularly relevant to studies of delayed preconditioning, as animal care committees currently

mandate anesthesia for the initial preconditioning stimulus. Thus an important component of this

model requires that it be able to separately examine the effects of anesthetic and ischemic PC. As

discussed earlier, there is a wealth of evidence showing that most modernly used volatile

anesthetics invoke potent early and delayed cardioprotection. However, it is still unknown

whether halothane and nitrous oxide cause delayed cardioprotection. Furthermore, while there is

a substantial body of literature in animal and human studies describing an early preconditioning

effect arising after transient limb ischemia, evidence of a late phase of protection is scarce.

Delayed preconditioning may have a strong impact in settings beyond the operating room, as it

can potentially be used to improve various parameters of cardiovascular function. It may also be

important in populations where maintaining a baseline preconditioned state can ameliorate the

negative side effects of certain cardiovascular diseases, such as diabetes, hypertension and

pathological cardiac hypertrophy.

34

Using the isolated heart model is an important and necessary first step in examining the

delayed preconditioning phenotype. Taking advantage of the cardiac specific responses will

expand our understanding of the late phase protection and may provide insight into future areas

of investigation required to optimize this powerful method of organ protection.

2.2 Research Aims/Objectives

Primary Objective

To identify and establish a method of sedation in mice for delayed rIPC experiments

using the Langendorff-model of IR injury

Specific Aims

1. To examine the delayed/second window (2W) effects of intra-peritoneal (IP) injection of

sodium-pentobarbital (SP) anesthesia or saline, on post-ischemic heart function, and to

compare the results with the early (first window-1W) control and rIPC groups

2. To investigate the delayed/2W effects of inhalational halothane and nitrous-oxide

anesthesia on post-ischemic heart function in order to assess their suitability as alternative

methods of sedation for delayed rIPC studies.

2.3 Hypotheses

i. It was hypothesized that intra-peritoneal injections of saline will induce delayed

cardioprotection against IR injury in isolated-hearts.

ii. It was hypothesized that halothane will induce delayed preconditioning against IR injury

whereas nitrous oxide will not confer cardioprotection.

35

Chapter 3 Methods

3.1 Ethics

All animal protocols were approved by the Animal Care and Use Committee of the

Hospital for Sick Children in Toronto and conformed to the Guide for the Care and Use of

Laboratory Animals published by the National Institutes of Health (NIH publication No. 85–23,

revised 1996).

3.2 Experimental Groups

Male C57BL/6 mice (9-11 weeks of age) were used for all treatment groups. Animals

were maintained on a 12h dark/light cycle and housed in single cages at room temperature and

were provided with food and water ad libitum. Mice were divided into intra-peritoneal and

inhalational anesthesia treatment categories to assess the delayed/second window (2W) effects of

these drugs with an established model of early/first window (1W) preconditioning studies used in

our lab. 6-8 mice were used in each group (figure 6).

First window model of rIPC

The established mode of preconditioning using the first window model of rIPC was

compared with the various delayed stimuli examined in this study to evaluate their potential

cardioprotective effects. In our model of early rIPC, mice were divided into first window control

(1WSP) and rIPC (1WSP+rIPC) groups. Mice in the 1WSP group received SP (60mg/kg of body

weight) via an intra-peritoneal (IP) injection and were kept under anesthesia for 40 minutes (to

match rIPC treatment duration). The 1WSP+rIPC group received SP and rIPC (four cycles of 5

36

min of hind-limb ischemia and 5 min of reperfusion). To examine the early phase of

preconditioning, hearts from the 1WSP and 1WSP+rIPC were isolated 15 minutes after each

treatment for mouse Langendorff IR injury or Western blot protein experiments (figure 3).

Intra-peritoneal anesthesia – the delayed effects of sodium-pentobarbital

To assess the delayed effects of SP alone and with additional rIPC, mice were divided

into 2WSP and 2WSP+rIPC groups. Mice in the 2WSP and 2WSP+rIPC groups received SP

anesthesia (60mg/kg of body weight; IP) on day1 ± rIPC (as per method in 1WSP+rIPC). We

also developed a delayed saline treated group (2W Saline) to test for the potential

preconditioning effects of an apparently ‘benign’ IP injection. Mice in this group received

0.12mL saline via IP injection as previously described[179]. To examine the delayed phase of

preconditioning, mice from these groups re-anesthetized with SP twenty-four hours later

(60mg/kg of body weight; IP) and hearts were isolated for global ischemia or for Western blot

protein experiments.

Inhalational Anesthesia – the delayed effects of halothane and N2O

To examine the delayed effects halothane and nitrous oxide inhalational anesthesia, mice

were divided into 2W Halothane and 2W N2O groups. The 2W Halothane group underwent

sedation with halothane anesthesia for 40 minutes at a 2% halothane/98% oxygen induction

concentration and was maintained at 1% halothane/99% oxygen for duration of the experiment.

These concentrations were determined from pilot experiments examining the amount of

halothane that was required to eliminate any response from hindlimb ischemia via a tourniquet.

Mice in the 2W N2O group were exposed to 40 minutes of N2O delivered at a 2:1 nitrous oxide

to oxygen ratio while placed in a 12 cm x 24 cm container. Twenty-four hours after sedation,

37

Figure 6: A schematic of the study protocol. Mice were divided into seven groups based on method of anesthesia±rIPC. Groups were also divided into intra-peritoneal and inhalational anesthesia for comparison with established 1W model of rIPC. For 1W groups, IR injury experiments occurred on the same day as preconditioning treatment. For 2W groups, IR injury experiments occurred on day 2, twenty-four hours after preconditioning treatment. Hearts were collected for western blot experiments at indicated points (WB). At the end of Langendorff IR injury, hearts were stained with TTC for LV infarct size analysis. 1W-first window/early, 2W-second window/delayed, rIPC-remote ischemic preconditioning, SP-sodium pentobarbital, N2O-nitrous oxide, TTC-triphenyltetrazolium chloride, WB-western blot protein analysis

38

mice were re-anesthetized with SP (60mg/kg of body weight via an IP injection) and hearts were

isolated for global ischemia or for Western blot protein experiments.

3.3 Induction of rIPC Using Inguinal Tourniquet Model

Mice were sedated with a method of anesthesia depending on the treatment group. rIPC

was induced by four cycles of 5 minutes of femoral artery occlusion intercalated with 5 minutes

of reperfusion. This was done using a tourniquet tied around the left hindlimb at the inguinal

level to apply unilateral ischemia to the femoral vasculature (figure 7). Ischemia was marked by

limb paleness and reperfusion by rapid hyperemia as previously described[31]. Hearts were then

isolated for Langendorff preparation or Western blot analysis 15 min (1W) or 24 hours (2W)

after rIPC.

Figure 7: Remote ischemic preconditioning (rIPC) via transient ischemia of mouse

hindlimb. A tourniquet is tied at the inguinal level to apply unilateral ischemia. Ischemia was

marked by distal limb paleness. Reperfusion was marked by rapid hyperemia.

5 min Ischemia

5 min Reperfusion

Tourniquet

Tourniquet

Tourniquet Tourniquet

39

3.4 A Langendorff Isolated Heart Model of Ischemia-Reperfusion Injury

IR injury was induced by 30 min of global ischemia followed by 60 min reperfusion in

isolated mouse hearts using a constant hydrostatic pressure, non-circulating, isovolumic

Langendorff preparation (Radnotti Technologies) as previously described in our lab [31], [178]

(figure 8a,b).

Preparation of Krebs-Heinseleit buffer

Hearts were perfused with modified Krebs-Heinseleit Buffer (KHB) at 80mmHg at 37°C

containing (mM): NaCl 120mM, NaHCO3 25mM, KCl 4.7mM, MgSO4 1.2 mM, KH2PO4 1.2

mM, CaCl2 2.5mM, EDTA 0.5mM and glucose 15mM. Buffer was prepared in a 2L Erlenmeyer

flask and subsequently filtered through a 0.2 µm Nalgene bottle top filter (Nalgene Cat#595-

4520, Rochester, NY). Buffer was adjusted to a pH of 7.4 by 6N HCl and oxygenated (95% O2,

5% CO2) in a water-jacketed reservoir via a sintered glass gas distributor (Radnotti

Technologies) for 60 minutes prior to experiment. Perfusion fluid was delivered to hearts

through water-jacketed tubes at 37°C.

Excision of Mouse Hearts and Aortic Cannulation

Mice were anesthetized with SP (60mg/kg of body weight) and received heparin to

prevent coagulation on apparatus (200 units – Sigma Inc.) via IP injections 5 min prior to heart

removal. Due to the respiratory depressant effects of SP[176], mice were then intubated and

ventilated at a respiratory rate of 100 breaths/min with a tidal volume of 1.5 mL per stroke. A

trans-abdominal incision was then made through the peritoneum to expose the diaphragm, which

is then cut to access the thoracic cavity and heart via a bilateral thoracotomy. Hearts were rapidly

excised and placed in a 100 mm dish containing oxygenated KHB at 37°C where it was trimmed

40

of excess connective, lung and thymus tissue. Hearts were then placed in a 4°C cannulate buffer

(NaCl 140mM, KCL 4.2mM, KH2PO4 1.2 mM, MgCl2 0.5mM, Hepes 10mM – pH 7.4) and the

aorta was cannulated with a 20-gauge metal cannula under magnification to the point just before

the aortic valves to allow for perfusate to enter through the coronary ostia. Sutures were tied

around aorta-cannula junction to secure aorta and to prevent leakage of KHB. Once the aorta was

cannulated, the three-way stopcock on the mounting position was turned to perfuse the coronary

circulation at 80mmHg with KHB.

Figure 8a. The mouse Langendorff-isolated heart model. Mouse coronary arteries are

perfused with modified Krebs-Heinseleit Buffer at a constant pressure of 80mmHg at 37°C, in

retrograde fashion through a cannula inserted into the aorta. An intra-ventricular balloon is

inserted into the left-ventricle through the mitral valve and connected to a pressure transducer

hooked up to bio-lab software to measure peak left-ventricle pressure (LVP), left-ventricular

end-diastolic pressure (LVEDP), left-ventricle developed pressure (LVDP), rate of contraction,

rate of relaxation and heart rate

41

Figure 8b. The mouse Langendorff-isolated heart model used in the Redington laboratory.

Langendorff setup can operate two mouse hearts using 2L of Krebs-Heinseleit buffer. Heart is

mounted on cannula inserted through aorta via a one way in-flow valve, which can readily be

closed to induce global ischemia.

42

Hearts were then mounted by attaching metal cannula to three-way stopcock on the Langendorff

apparatus. The enitre process from heart excision to mounting takes approximately 3-5 minutes.

Additional sutures were knotted around aorta to secure it to the cannula. To facilitate drainage of

perfusate from coronary circulation, a small incision was made at the base of pulmonary artery

using micro-scissors.

Insertion of An Intra-Ventricular Balloon

Once perfusion is initiated, hearts were submerged in a water-jacketed container at 37°C

throughout the duration of experiment. The auricular appendage of the left atrium was removed

to expose the atrium. A saran wrap balloon, with PE60 polyethylene tubing connected to a

microsyringe and pressure transducer (AD instruments (ADI)-ML844, Colorado Springs, CO),

was placed in the left ventricle through the mitral valve. The balloon volume was initially

deflated upon insertion and then adjusted once placed within the ventricle to give a preload left-

ventricular end-diastolic pressure (LVEDP) of 7-10mmHg (contains <20µL of distilled water).

Balloons were fastened to cannula using tape to maintain a straight entry into the ventricle. The

balloon volume was kept constant throughout the duration of the experiment.

Measurement of Cardiac Function and Induction of Global Ischemia

Hearts were stabilized on the apparatus for 20 min prior to global ischemia. Using Power-

lab acquisition software (ADI instruments, Colorado Springs, CO – see appendix 1) we measured

hemodynamic parameters of cardiac function throughout the experiment, which included peak

left ventricular pressure (LVP), maximum rate of ventricular contraction (+dP/dtmax) and

maximum rate of ventricular relaxation (-dP/dtmin), LVEDP and heart rate. Left-ventricular

developed pressure (LVDP) was determined as the difference between peak systolic and diastolic

43

pressures. Coronary flow rate was measured by collecting effluent produced in 1 min via the

coronary sinus at two time points: before global ischemia and at the end of 60 minutes of

reperfusion. Thirty minutes of global ischemia was induced by turning the three-way stopcock to

the closed position to cease perfusion with KHB and was set to the open position during

reperfusion (60min). At the end of the experiment, hearts were collected and immersed in 10%

KCl to induce diastole. Hearts were then weighed and frozen in liquid nitrogen and stored at

-80°C and later measured for infarct size.

Special Considerations

Given the technical challenges involved in excising and perfusing mouse hearts and the

various important details involved with the apparatus setup, it was important to use established

criteria for determining the suitability of hearts for the experiment[178]. At the end of the 20 min

stabilization, hearts were excluded if they acquire: (1) bradycardic heart rate < 300 bpm, (2) flow

rate > 5 mL/min, (3) LVP <80 mmHg (4) high level of arrhythmia.

3.5 Infarct Size Determination

After reperfusion, the frozen heart was transversely cut into six 1-mm thick slices using a

Mouse Heart Slicer Matrix (Zivic Instruments) which were stained with 1.25% 2,3,5-

triphenyltetrazolium chloride (TTC) in 200 mM Tris/HCL solution (pH 7.4) for 15 min in a 37°C

water bath. After staining, heart slices were fixed for 2-4 hours in 10% neutral buffered

formaldehyde. Both sides of each slice was then photographed at 1200 DPI resolution using a

computer scanner (CanoScan 4400F). Images were processed with Adobe Photoshop® CS2

software to measure infarct size and left-ventricle area using automated planimetry. Viable

myocardium stains red due to the reaction of tetrazolium salts with NADH and dehydrogenase

44

enzymes while infarcted tissue, that does not possess enzymes, appears pale[180]. Infarct sizes of

each slice were expressed as the percentage of the total left ventricle area. The reported infarct

size is the mean of infarct size measurements from both sides of all the individual slices.

3.6 Protein Extraction

For protein analysis experiments, hearts were harvested either 15 min (1W groups) or 24

hours (2W groups) after exposure to anesthesia ± rIPC without IR injury. Each heart was frozen

in liquid nitrogen and stored at –80°C. For determining protein concentration, hearts were cut

into small pieces (totaling 50mg) and shaken in 1 mL micro-tubes (Dia-Med Lab Supplies Inc.

Mississauga, ON) using two metals beads in order to homogenize tissue in lysis buffer (see

appendix). Homogenate was incubated on ice for 30 min to allow for tissue lysing. Homegenate

was then centrifuged at 10,000 rpm at 4°C for 30 min to obtain whole tissue supernatant and the

discarded pellet. Collected supernatant was then centrifuged at 10,000 rpm at 4°C for 10 min to

further separate supernatant and discarded pellet. Protein concentration was determined using a

bovine serum albumin (BSA) standard (25 µg/mL) (Sigma-Aldrich Co., St. Louis, MO). Protein

concentration was determined by preparing a solution consisting of 1-2 µL of resulting sample

supernatant, 200 µL of dye reagent protein assay concentrate (Bio-Rad Laboratories Catalog,

Hercules, CA) and 800 µL of dd-H2O. The solution was vortexed and allowed to react for a

minimum of 15min. Using a UV visible spectrophotometer (Ultrospec 3000, Fisher Scientific,

Markham, ON) a standard curve of BSA optical density (OD) measured at 595 nm for 0 µg, 2.5

µg, 5 µg, 10 µg, 15 µg and 20 µg was constructed. Sample protein concentrations were measured

at the same OD and the resulting concentration was determined from the developed standard

curve. Samples were aliquot to 6-9 tubes and stored at -80°C for further analysis.

45

3.7 SDS-PAGE and Western Blot Analysis

Resolving and Stacking Gel Preparation

Protein samples were separated by one-dimensional sodium dodecyl sulphate (SDS)-

polyacrylamide gel electrophoresis (PAGE) using 10% resolving and 5% stacking gel (see

appendix for gel make-up) on a Bio-Rad Mini-protean III gel electrophoresis system (Bio-Rad

Laboratories, Hercules, CA). Glass plates with 0.75 mm spacers were set up in a gel-casting

frame. The 10% resolving gel (see appendix) was prepared and added between glass plates and

allowed to polymerize for 45 min. Gel surface was overlaid with distilled water. 5% Stacking gel

was then prepared (see appendix 2) and added on top of resolving gel with a 10-well plastic-

comb and allowed to polymerize for 45 min.

Electrophoresis of Proteins

An equal amount of protein (30 µg) from whole tissue lysates was prepared with

homogenizing lysis buffer and 5 uL of 3X sample buffer w/ 100mM DTT reducing agent (Cell

Signaling Technologies; 87.5 mM Tris-HCl (pH 6.8 at 25ºC), 6% (w/v) SDS, 30% glycerol and

0.03% (w/v) bromophenol blue) to give a final loading volume of 15 uL. Samples were

centrifuged and boiled for 5 min at 100°C. 5µL of PageRulerTM Plus Prestained Protein Ladder

(Thermo Fisher Scientific, Rockford, IL) was placed in first well as a protein molecular weight

marker. Proteins were loaded into wells 2-10, and electrophoresed in a 1X Tris/Glycine/SDS

buffer solution (25mM Tris, 192 mM glycine, 0.1% SDS, stored at rm. temp, pH 8.3) with an

initial potential difference of 80V at the start (until gel-front migrated past stacking gel) and then

at 100mV for migration through resolving gel.

46

Transfer of Proteins to Nitrocellulose Membranes

Following electrophoresis, resolving gels equilibrated in transfer buffer (25 mM Tris, 192

mM glycine, 20% methanol-stored at 4°c) for 15 min. Nitrocellulose membranes (0.45 µm pore

size - Bio-Rad Laboratories) were placed dd-H2O for 15 min and then in transfer buffer for 5

min. Resolving gels were then placed in western-transfer sandwich consisting of brillo pad, two-

pieces of filter paper (Whatman®, Maidstone, England), nitrocellulose membrane, resolving gel,

two-pieces of filter paper and a final brillo pad. Western-transfer sandwich was then placed in

gel-transfer unit using Bio-Rad Mini-protean III transfer system with an ice-pack and constant

stirring. Proteins migrate from resolving gel on to nitrocellulose membrane across a potential

difference of 100V for 90min. The transfer of proteins was done in an ice-filled container to

minimize overheating of the apparatus.

Detection of Proteins with Anti-bodies

Following protein transfer, the nitrocellulose membranes were rinsed with three 5 min

cycles with 1X TBST buffer (10X stock – Tris 24.2g, NaCl 80.0g, pH 7.6 brought to 1L and

diluted 1/10 for 1X TBST + 1 mL Tween 20) at room temperature. The membrane was then

placed in blocking buffer solution (1 mL TBST buffer with 5% (w/v) skim milk powder) for 1

hr. Membranes were then incubated overnight with rabbit monoclonal antibodies against

phospho-Akt at the Ser473 residue (Catalog #4060 - Cell Signaling Tech.) at a 1:1000 dilution,

total-Akt (Catalog #4691 - Cell Signaling Tech.) at a 1:1000 dilution, phosphor-p44/42 MAPK at

the Thr202/Tyr204 residues (Catalog #4370 - Cell Signaling Tech.), total p44/42 (Catalog #4695

- Cell Signaling Tech. Danvers, MA) at a 1:1000 dilution and the mouse monoclonal antibody

GAPDH (Catalog #G8795 - Sigma-Aldrich Inc) at a 1:15,000 dilution. Membranes were then

washed with 1X TBST buffer for three 5 min cycles and subsequently incubated with a

47

horseradish-peroxidase (HRP)-conjugated secondary antibody (Santa Cruz Biotechnology) at an

appropriate dilution. Secondary antibody bound to a primary antibody was detected using

enhanced chemiluminescence (ECL) plus detection kit (Amersham Inc.). Membranes were then

scanned using STORM 840 analyzer and quantified using ImageQuant 5.0 densitometry software

(Molecular Dynamics).

3.8 Data and Statistical analysis

Data from Power Lab Acquisition software were analyzed using Lab Chart 7 and

processed to Microsoft Excel 2011 for Macintosh. Graphs and charts were made using Microsoft

Excel 2011 and GraphPad Prism 5.0 software. Statistical analysis was done using one-way

ANOVA and Student’s t-test function in GraphPad Prism 5.0 software. Hemodynamic data were

processed into 10-second segments from each isolated heart experiment, and values were

reported in 10 min interval summaries. LVP, +dP/dtmax, and -dP/dtmin were analyzed and

expressed as a percentage of baseline pre-ischemic values. All values are expressed as means ±

standard error of the mean (SEM). A statistically significant difference was among three or

more groups was determined using one-way ANOVA and post-hoc testing (Newman-Keuls Test)

through GraphPad Prism 5.0 software. A Student’s t-test was used to compare the differences in

means between two groups from western blot analysis. All treatment groups were compared to

1WSP control groups for effects. A difference associated with a P-value of ≤0.05 was considered

statistically significant.

48

Chapter 4 Results

4.1 The Delayed Effects of Intra-Peritoneal and Inhalational

Anesthesia on Left Ventricular Function after Global Ischemia

4.1.1 Baseline Function

In the present study we compared the delayed effects of intra-peritoneal SP and

inhalational halothane and N2O anesthesia with our established model of early rIPC on LV

function after global ischemia. The following hemodynamic parameters of isolated-hearts were

assessed throughout the duration of the IR injury experiments: Left ventricular developed

pressure (LVDP), left ventricular end-diastolic pressure (LVEDP), maximum rate of contraction

(dP/dtmax), maximum rate of relaxation (dP/dtmin) and heart rate. The baseline values from each

group were compared to ensure that there were no initial differences prior to induction of IR

injury. As shown in table 1, there were no statistically significant differences in baseline cardiac

function between all treatment groups at the end of the 20-minute stabilization period. Baseline

cardiac function was used to assess the recovery of hearts after IR injury, as the values of LVDP,

dP/dtmax and dP/dtmin acquired during reperfusion were expressed as a percentage of baseline

function. The LVEDP for each treatment was expressed as the absolute value throughout the

experiment. As described in the methods section, hearts were assessed for their suitability for IR

injury experiments based on an established set of criteria from the literature. Hearts were

excluded from the study if at the end of the stabilization, they exhibited a (1) bradycardic

HR<300 beats/min (2) CFR>5.0mL/min (3) LVP< 80mmHg and (4) high level of arrhythmia.

49

Group

Parameter 1WSP

(n=12) 1WSP+rIPC (n=10)

2WSP (n=10)

2WSP+rIPC (n=5)

2W Saline (n=6)

2W Halothane

(n=6)

2W N2O (n=6)

Body Weight (g) 26.60 ± 0.5 26.78 ± 0.2 24.46 ± 0.4 25.91 ± 1.0 25.20 ± 0.6 25.39 ± 0.5 25.75 ± 0.4

Heart Weight (mg)

145.4 ± 5.0 136.6 ± 5.0 143.2 ± 3.2 150.0±8.2 145.7 ± 5.2 139.1 ± 3.4 147.8 ± 5.1

Heart Rate (beats/min) 362.5 ± 14 346.7 ± 9.1 338.9 ± 13 341.8 ± 20 329.1 ± 8.4 321.5 ± 9.4 354.8 ± 7.9

Systolic LVP (mmHg)

93.48 ± 6.5 96.2 ± 4.8 94.16 ± 5.5 97.10 ± 4.4 89.18 ± 2.6 94.12 ± 3.6 90.54 ± 3.1

LVEDP (mmHg) 8.3 ± 0.8 7.6 ± 0.8 9.9 ± 0.6 10.3 ± 1.0 8.7±0.5 9.7 ± 0.6 8.4 ± 1.2

LVDP (mmHg) 85.19 ± 6.7 81.74 ± 6.2 84.32 ± 5.6 86.76 ± 4.2 80.31 ± 2.7 84.43 ± 3.6 82.13 ± 3.2

dP/dtmax (mmHg/sec) 3467.4 ± 387 3450.5 ± 247 3935.6 ± 460 4099.7 ± 665 3704.6 ± 464 3758.3 ± 471 4476.3 ±476

dP/dtmin (mmHg/sec) 2451.9 ± 237 2524.0 ± 207 2506.7 ± 205 2619.6 ± 288 2209.1 ± 137 2395.8 ± 174 2765.8 ± 139

Coronary Flow Rate (mL/min)

2.3 ± 0.3 2.0 ± 0.5 2.0±0.3 1.7 ± 0.1 1.5 ± 0.1 1.7 ± 0.1 2.0 ± 0.1

Table 1: Baseline functional parameters in the Langendorff-isolated heart groups for IR

injury experiments. All cardiac function parameters represent values obtained at the end of the

20-minute stabilization period. 6-8 mice were used in each treatment group. Data are expressed

as mean values ± SEM. 1W-first window/early, 2W-second window/delayed, rIPC-remote

ischemic preconditioning, SP-sodium pentobarbital, N2O-nitrous oxide. LVP-left-ventricle

pressure, LVEDP-left ventricular end-diastolic pressure, LVDP-left ventricular developed

pressure, dP/dtmax – maximum rate of contraction, dP/dtmin – maximum rate of relaxation

50

4.1.2 Left Ventricular Developed Pressure

Left ventricular developed pressure (LVDP) provides an indication of contractile capacity

and can be used to assess the functional recovery of the ventricle after IR injury. LVDP is

calculated from the pressure difference (in mmHg) between peak left ventricular systolic and

diastolic pressures. As shown in table 1, there were no significant differences in LVDP among

the treatment groups. LVDP was approximately 75-85mmHg at the end of stabilization and

decreased to 0mmHg during ischemia in all groups (figure 9a,b). All values obtained during

ischemia and subsequent reperfusion are expressed as a percentage of the pre-ischemic baseline

value obtained at the end of stabilization.

As we have shown in previous studies, early preconditioning with transient hindlimb

ischemia enhances the recovery of LVDP after IR injury. After 30 minutes of ischemia and 60

minutes of reperfusion, the 1WSP+rIPC group showed an 81.9 ± 4.7% recovery of LVDP

compared to only 57.3 ± 4.9% in 1WSP controls (P<0.05) (figure 9a,b).

A primary objective of this study was to examine the delayed effects of SP anesthesia and

rIPC on LV function. We did this by sedating mice with SP and applying rIPC (in the same

fashion as 1WSP+rIPC) twenty-four hours prior to IR injury. We found that both delayed SP and

SP+rIPC significantly improved (P<0.05) LVDP after IR injury compared to 1WSP control mice

(2WSP: 82.6 ±2.4%, 2WSP+rIPC 77.9 ± 5.3%). There were no significant differences in LVDP

between the 2WSP and 2WSP+rIPC groups (figure 9a,c).

To investigate whether there were any effects from the method of delivery of SP intra-

peritoneal anesthesia, we also assessed whether an IP injection could affect cardiac function in

the 2W of protection. Interestingly, we found that an IP injection of saline fluid, twenty-four

hours prior to IR injury, also led to a significant improvement in LVDP compared to 1WSP

controls after 60 minutes of reperfusion (2W Saline, 76.8 ± 3.8%) (figure 9a,c). Given that the

51

2WSP and 2W Saline groups showed similar recovery in developed pressure suggests that the

method of drug delivery could be inducing delayed cardioprotection.

Another objective of this study was to study the potential delayed preconditioning effects

of halothane and nitrous oxide inhalational anesthesia. Similar to the intra-peritoneal groups, we

found that prior administration of both gases for 40 minutes a day before IR injury significantly

improved (P<0.05) LVDP recovery compared to 1WSP controls (2W Halothane: 78.8 ± 2.0%,

2W N2O: 73.3 ± 3.6%) (figure 9b,c).

There were no significant differences in LVDP between the delayed intra-peritoneal and

inhalational anesthetic groups and the early rIPC group after 60 minutes or reperfusion (figure

9c). These findings indicate that a variety of stimuli can improve post-ischemic left ventricular

function by recovering developed pressure to near pre-ischemic function.

52

Figure 9a: The effects of intra-peritoneal anesthesia on post-ischemic left-ventricular

developed pressure (LVDP). All values are expressed as a percentage of the baseline LVDP

obtained at the end of the 20-minute stabilization period. IR injury was induced by 30 minutes of

ischemia and 60 minutes of reperfusion. 1W groups represent treatment with anesthetic±rIPC 15

minutes prior to IR injury. 2W groups represent treatment with anesthetic or saline 24 hours prior

to IR injury. An * denotes a statistically significant difference between treatment group and

1WSP controls at 60 minutes reperfusion (P<0.05). Data are expressed as mean values ± SEM.

(n=6-8 per treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-remote

ischemic preconditioning, SP-sodium pentobarbital.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

100

1WSP1WSP+rIPC

2WSP

2WSP+rIPC2W Saline

Ischemia Time (sec)

LVD

P (%

Pre

-Isch

emia

)

*

Time (min)

53

Figure 9b: The effects of inhalational anesthesia on post-ischemic left-ventricular

developed pressure (LVDP). All values are expressed as a percentage of the baseline LVDP

obtained at the end of the 20-minute stabilization period. IR injury was induced by 30 minutes of

ischemia and 60 minutes of reperfusion. 1W groups represent treatment with anesthetic±rIPC 15

minutes prior to IR injury. 2W groups represent treatment with anesthetic 24 hours prior to IR

injury. An * denotes a statistically significant difference between treatment group and 1WSP

controls at 60 minutes reperfusion (P<0.05). Data are expressed as mean values ± SEM. (n=6-8

per treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-remote

ischemic preconditioning, N2O – nitrous oxide.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

100

1WSP1WSP+rIPC

2W Halothane2W N2O

Ischemia Time (sec)

LVD

P (%

Pre

-Isch

emia

)

*

Time (min)

54

Figure 9c: The effects of intra-peritoneal and inhalational anesthesia on post-ischemic left-

ventricular developed pressure (LVDP) at 60 minutes of reperfusion. All values are

expressed as a percentage of the baseline LVDP obtained at the end of the 20-minute

stabilization period. 1W groups represent treatment with anesthetic±rIPC 15 minutes prior to IR

injury. 2W groups represent treatment with anesthetic or saline 24 hours prior to IR injury. An *

denotes a statistically significant difference between treatment group and 1WSP controls

(P<0.05). Data are expressed as mean values ± SEM. (n=6-8 per treatment group). 1W-first

window/early, 2W-second window/delayed, rIPC-remote ischemic preconditioning, SP-sodium

pentobarbital, N2O-nitrous oxide.

1WSP

1WSP+rIPC

2WSP

2WSP+rIPC

2W Saline

2W Halothane

2W N2O0

20

40

60

80

100

* (P<0.05)LV

DP (%

Pre

-Isch

emia)

55

4.1.3 Left Ventricular End-Diastolic Pressure

At the beginning of each experiment, the intra-ventricular balloon volume was adjusted to

set isolated-hearts at a LV preload equivalent to a diastolic pressure of 5-10mmHg (table 1).

Balloon volume was maintained throughout the experiment and thus further alterations in

LVEDP signify the effects of IR injury on the LV. IR injury reduces ventricular compliance and

the increased stiffness is manifest by higher end-diastolic pressures. The progressive increase in

LVEDP after IR injury likely occurs because of changes in cardiomyocyte Ca2+-handling and the

depletion of high-energy phosphates[181], [182].

Consistent with the reported benefits of rIPC on post-ischemic cardiac function, transient

limb ischemia induced early cardioprotection by attenuating the elevation of LVEDP after IR

injury (figure 10a,b). Within only 10 minutes of reperfusion, there was a significant difference

(P<0.001) in LVEDP between 1WSP (67.1 ± 5.4 mmHg) and 1WSP+rIPC (38.5 ± 5.1 mmHg)

groups (figure 10a). Increasing reperfusion time brings a steady decrease in LVEDP as hearts

begin to recover contractile capacity, although end-diastolic pressures remain above pre-ischemic

values. In our early model of rIPC, the 1WSP+rIPC group continued to show a significantly

greater (P<0.001) decline in LVEDP until the end of the 60 minutes reperfusion compared to

1WSP controls (1WSP: 43.1 ± 4.1 mmHg, 1WSP+rIPC: 21.8 ± 2.7 mmHg)(figure 10c).

We also compared the delayed effects of SP (2WSP) on LVEDP with our early

preconditioning groups (1WSP, 1WSP+rIPC). We found that the mice pretreated with SP

anesthesia 24 hours before the experiment also displayed a significantly lower (P<0.001)

LVEDP after 60 min of reperfusion (2WSP: 17.6 ± 1.7 mmHg) compared to 1WSP controls

(figure 10a,c). The 2WSP+rIPC group also displayed a significantly lower increase in LVEDP

than 1WSP controls after reperfusion (2WSP+rIPC: 17.3 ± 3.5 mmHg). There were no

differences in end-diastolic pressure between the 2WSP and 2WSP+rIPC groups, suggesting that

56

rIPC provided no additional benefits in the delayed phase. Interestingly, we also observed that an

IP injection of saline significantly attenuated the rise of LVEDP after 60 minutes of reperfusion

compared to 1WSP controls (2W Saline: 28.0 ± 6.6 mmHg) (figure 10a,c).

Inhalational anesthesia produced a similar effect on the post-ischemic increase of

LVEDP. We found that pre-treatment with halothane (2W Halothane) and nitrous-oxide (2W

N2O) anesthesia significantly reduced (P<0.05) LVEDP at the end of reperfusion compared to

1WSP controls (1WSP: 43.1 ± 4.1 mmHg, 2W Halothane: 19.4 ± 1.7 mmH, 2W N2O: 26.7 ± 2.2

mmHg) (figure 10b,c).

There were no significant differences in LVEDP between the delayed intra-peritoneal and

inhalational anesthetic groups and the early rIPC group after 60 minutes of reperfusion (figure

9c). These findings indicate that, similar to the improvements in the LVDP, both early and

delayed forms of preconditioning can attenuate post-ischemic diastolic dysfunction.

57

Figure 10a: The effects of intra-peritoneal anesthesia on post-ischemic left-ventricular end-

diastolic pressure (LVEDP). All values are expressed as the absolute value of LVEDP in

mmHg. IR injury was induced by 30 minutes of ischemia and 60 minutes of reperfusion. 1W

groups represent treatment with anesthetic±rIPC 15 minutes prior to IR injury. 2W groups

represent treatment with anesthetic or saline 24 hours prior to IR injury. An * denotes a

statistically significant difference between treatment group and 1WSP controls at 60 minutes of

reperfusion (P<0.05). Data are expressed as mean values ± SEM. (n=6-8 per treatment group).

1W-first window/early, 2W-second window/delayed, rIPC-remote ischemic preconditioning, SP-

sodium pentobarbital.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

1001WSP1WSP+rIPC

2WSP

2WSP+rIPC2W Saline

Ischemia Time (sec)

LVED

P (m

mH

g)

*

Time (min)

58

Figure 10b: The effects of inhalational anesthesia on post-ischemic left-ventricular end-

diastolic pressure (LVEDP). All values are expressed as the absolute value of LVEDP in

mmHg. IR injury was induced by 30 minutes of ischemia and 60 minutes of reperfusion. 1W

groups represent treatment with anesthetic±rIPC 15 minutes prior to IR injury. 2W groups

represent treatment with anesthetic 24 hours prior to IR injury. An * denotes a statistically

significant difference between treatment group and 1WSP controls at 60 minutes of reperfusion

(P<0.05). Data are expressed as mean values ± SEM. (n=6-8 per treatment group). 1W-first

window/early, 2W-second window/delayed, rIPC-remote ischemic preconditioning, N2O-nitrous

oxide.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

100

1WSP1WSP+rIPC

2W Halothane2W N2O

Ischemia Time (sec)

LVED

P (m

mH

g)

*

Time (min)

59

Figure 10c: The effects of intra-peritoneal and inhalational anesthesia on post-ischemic left-

ventricular end-diastolic pressure (LVEDP) at 60 min of reperfusion. All values are

expressed as the absolute value of LVEDP in mmHg. 1W groups represent treatment with

anesthetic±rIPC 15 minutes prior to IR injury. 2W groups represent treatment with anesthetic or

saline 24 hours prior to IR injury. An * denotes a statistically significant difference between

treatment group and 1WSP controls (P<0.05). Data are expressed as mean values ± SEM. (n=6-8

per treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-remote

ischemic preconditioning, SP-sodium pentobarbital, N2O-nitrous oxide.

1WSP

1WSP+rIP

C2W

SP

2WSP+rIP

C

2W Sali

ne

2W H

alotha

ne

2W N

2O0

10

20

30

40

50

60LV

EDP

(mm

Hg)

* (P<0.05)

60

4.1.4 Maximum Rate of Contraction

The maximum rate of contraction (dP/dtmax) was measured by the changes in left

ventricular pressure over time (expressed as a positive rate of gain in pressure measured in

mmHg/sec). Post-ischemic values of dP/dtmax can be used to evaluate LV systolic function.

dP/dtmax is known to be affected by heart rate and is dependent on LV preload, as it describes the

rate at which the LV develops pressure before opening of the aortic valve [183]. As with the

other measures of cardiac function, there were no significant differences in dP/dtmax between the

treatment groups at the end of the stabilization period (table 1). Post-ischemic measurements of

dP/dtmax were normalized and expressed as a percentage of their pre-ischemic values.

Similar to LVDP, non-preconditioned, 1WSP treated mice, recovered only 50-60% of

their pre-ischemic rate of LV contraction after IR injury. The early preconditioning effects of

rIPC significantly augmented the recovery in dP/dtmax after 60 minutes of reperfusion when

compared to 1WSP controls (1WSP: 54.3 ± 4.5% of pre-ischemic value, 1WSP+rIPC: 83.4 ±

4.6% - P<0.05) (figure 11a-c). Intra-peritoneal SP anesthesia and saline injection also

significantly improved dP/dtmax in the delayed phase of protection compared to 1WSP controls

(2WSP: 85.6 ± 2.7%, 2W Saline: 81.4 ± 4.5%) (figure 11a,c). There were no additional benefits

from rIPC in the 2WSP+rIPC compared to SP alone (2WSP), although recovery in this group

was still significantly better compared to 1WSP treated mice.

Twenty-four hours pretreatment with halothane anesthesia also had a delayed effect on

dP/dtmax as hearts in this group had improved recovery compared to 1WSP controls (2W

halothane: 82.9 ± 6.2% - P<0.05). Interestingly, while the 2W N2O group also exhibited a

significant recovery in dP/dtmax (2W N2O: 67.4 ± 2.1%) compared to 1WSP mice, the magnitude

of recovery at the end of reperfusion was intermediate between 1WSP and all the other treatment

groups (figure 11b,c).

61

There were no significant differences in dP/dtmax between the delayed intra-peritoneal and

inhalational anesthetic groups and the early rIPC group after 60 minutes of reperfusion (figure

11c). These findings support the observed effects of the various early and delayed stimuli on

other parameters of cardiac function (LVDP, LVEDP) after IR injury, indicating a

cardioprotective benefit on not only the extent, but also the rate of systolic pressure development.

62

Figure 11a: The effects of intra-peritoneal anesthesia on post-ischemic maximal rate of

contraction (dP/dtmax). All values are expressed as a percentage of the baseline dP/dtmax

obtained at the end of the 20-minute stabilization period. IR injury was induced by 30 minutes of

ischemia and 60 minutes of reperfusion. 1W groups represent treatment with anesthetic±rIPC 15

minutes prior to IR injury. 2W groups represent treatment with anesthetic or saline 24 hours prior

to IR injury. An * denotes a statistically significant difference between treatment group and

1WSP controls at 60 minutes of reperfusion (P<0.05). Data are expressed as mean values ±

SEM. (n=6-8 per treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-

remote ischemic preconditioning, SP-sodium pentobarbital.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

100

1WSP1WSP+rIPC

2WSP

2WSP+rIPC2W Saline

Ischemia Time (sec)

Dp/

Dt M

ax (%

Pre

-Isch

emia

)

*

Time (min)

63

Figure 11b: The effects of inhalational anesthesia on post-ischemic maximal rate of

contraction (dP/dtmax). All values are expressed as a percentage of the baseline dP/dtmax

obtained at the end of the 20-minute stabilization period. IR injury was induced by 30 minutes of

ischemia and 60 minutes of reperfusion. 1W groups represent treatment with anesthetic±rIPC 15

minutes prior to IR injury. 2W groups represent treatment with anesthetic 24 hours prior to IR

injury. An * denotes a statistically significant difference between treatment group and 1WSP

controls at 60 minutes of reperfusion (P<0.05). Data are expressed as mean values ± SEM. (n=6-

8 per treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-remote

ischemic preconditioning, N2O-nitrous oxide.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

100

1WSP1WSP+rIPC

2W Halothane2W N2O

Ischemia Time (sec)

Dp/

Dt M

ax (%

Pre

-Isch

emia

)

*

Time (min)

64

Figure 11c: The effects of intra-peritoneal and inhalational anesthesia on post-ischemic

maximal rate of contraction (dP/dtmax) at 60 minutes of reperfusion. All values are expressed

as a percentage of the baseline dP/dtmax obtained at the end of the 20-minute stabilization period.

1W groups represent treatment with anesthetic±rIPC 15 minutes prior to IR injury. 2W groups

represent treatment with anesthetic or saline 24 hours prior to IR injury. An * denotes a

statistically significant difference between treatment group and 1WSP controls (P<0.05). Data

are expressed as mean values ± SEM. (n=6-8 per treatment group). 1W-first window/early, 2W-

second window/delayed, rIPC-remote ischemic preconditioning, SP-sodium pentobarbital, N2O-

nitrous oxide.

1WSP

1WSP+rIPC

2WSP

2WSP+rIPC

2W Saline

2W Halothane

2W N2O0

20

40

60

80

100

Dp/D

t Max

(% P

re-Is

chem

ia)

* (P<0.05)

65

4.1.5 Maximum Rate of Relaxation

Similar to the rate of contraction, the maximal rate of relaxation (dP/dtmin) provides a

measure of cardiac diastolic function by assessing the maximal rate at which the ventricle relaxes

and decreases pressure during diastole (expressed as a maximal negative rate of change in

pressure measured in mmHg/sec). There were no significant differences in dP/dtmin in hearts

among the different treatment groups at the end of the stabilization period (table 1). Post-

ischemic measurements of dP/dtmin were normalized and expressed as a percentage of their pre-

ischemic values.

After 60 minutes of reperfusion, the rate of relaxation recovered to similar levels in the

1WSP+rIPC (76.7 ± 4.7% of pre ischemic value) and 2W intra-peritoneal anesthesia groups

(2WSP: 81.5 ± 3.0%, 2WSP+rIPC: 78.7 ± 4.7%, 2W Saline 75.9 ± 5.8% - with all groups

displaying significantly higher (P<0.05) levels of recovery compared to 1WSP hearts (53.0 ±

4.7%)(figure 12a,c). Additionally, halothane inhalational anesthesia also significantly improved

dP/dtmin compared to 1WSP controls (2W Halothane 84.1 ± 9.9%). Similar to the recovery of the

rate of contraction, 2W N2O mice exhibited an intermediate level of recovery in dP/dtmin (2W

N2O: 61.0 ± 2.4%) after 60 minutes of reperfusion, however it was not significantly different

from 1WSP controls (figure 12b,c).

There were no significant differences in dP/dtmin between the delayed intra-peritoneal and

halothane anesthesia groups and the early rIPC group after 60 minutes of reperfusion (figure

12c). These findings add to the accumulating evidence from the other measures of cardiac

function assessed in this study that early rIPC and delayed intra-peritoneal and inhalational

anesthesia can similarly improve the contractile capacity of hearts after IR injury.

66

Figure 12a: The effects of intra-peritoneal anesthesia on post-ischemic rate of relaxation

(dP/dtmin). All values are expressed as a percentage of the baseline dP/dtmin obtained at the end

of the 20-minute stabilization period. IR injury was induced by 30 minutes of ischemia and 60

minutes of reperfusion. 1W groups represent treatment with anesthetic±rIPC 15 minutes prior to

IR injury. 2W groups represent treatment with anesthetic or saline 24 hours prior to IR injury. An

* denotes a statistically significant difference between treatment group and 1WSP controls at 60

minutes of reperfusion (P<0.05). Data are expressed as mean values ± SEM. (n=6-8 per

treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-remote ischemic

preconditioning, SP-sodium pentobarbital.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

100

1WSP1WSP+rIPC

2WSP

2WSP+rIPC2W Saline

Ischemia Time (sec)

Dp/

Dt M

in (%

pre

-isch

emia

)

*

Time (min)

67

Figure 12b: The effects of inhalational anesthesia on post-ischemic rate of relaxation

(dP/dtmin). All values are expressed as a percentage of the baseline dP/dtmin obtained at the end

of the 20-minute stabilization period. IR injury was induced by 30 minutes of ischemia and 60

minutes of reperfusion. 1W groups represent treatment with anesthetic±rIPC 15 minutes prior to

IR injury. 2W groups represent treatment with anesthetic 24 hours prior to IR injury. An *

denotes a statistically significant difference between treatment group and 1WSP controls at 60

minutes of reperfusion (P<0.05). Data are expressed as mean values ± SEM. (n=6-8 per

treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-remote ischemic

preconditioning, N2O-nitrous oxide.

0 10 20 30 40 50 60 70 80 90 100 110 1200

10

20

30

40

50

60

70

80

90

100

1WSP1WSP+rIPC

2W Halothane2W N2O

Ischemia Time (sec)

Dp/

Dt M

in (%

pre

-isch

emia

)

*

Time (min)

68

Figure 12c: The effects of intra-peritoneal and inhalational anesthesia on post-ischemic rate

of relaxation (dP/dtmin) at 60 minutes of reperfusion. All values are expressed as a percentage

of the baseline dP/dtmin obtained at the end of the 20-minute stabilization period. 1W groups

represent treatment with anesthetic±rIPC 15 minutes prior to IR injury. 2W groups represent

treatment with anesthetic or saline 24 hours prior to IR injury. An * denotes a statistically

significant difference between treatment group and 1WSP controls (P<0.05). Data are expressed

as mean values ± SEM. (n=6-8 per treatment group). 1W-first window/early, 2W-second

window/delayed, rIPC-remote ischemic preconditioning, SP-sodium pentobarbital, N2O-nitrous

oxide.

1WSP

1WSP+rIPC

2WSP

2WSP+rIPC

2W Saline

2W Halothane

2W N2O0

20

40

60

80

100

Dp/D

t Min

(% p

re-is

chem

ia)* (P<0.05)

69

4.2 Delayed Preconditioning with Intra-Peritoneal and Inhalational Anesthesia Reduce Infarct Size after IR Injury

At the end of reperfusion, hearts were cut into six 1mm slices and stained with TTC to

examine LV infarct size. Infarction sizes are expressed as a percentage of the LV area.

Paralleling the cardiac function data on early preconditioning via hindlimb ischemia, we found

that 1WSP+rIPC hearts had significantly reduced (P<0.05) LV infarct sizes compared with the

1WSP control group (1WSP: 43.6 ± 5.9%, 1WSP+rIPC: 19.6 ± 3.5%) (figure 13a) . Furthermore,

in addition to its delayed benefits on post-ischemic heart performance, SP anesthesia is capable

of reducing LV infarct size in the 2W of protection. The 2WSP group had significantly smaller

LV infarct sizes compared to 1WSP controls (2WSP: 28.0 ±1.9%). Hearts from the 2WSP+rIPC

and 2W Saline groups exhibited significantly reduced infarction size compared to 1WSP treated

controls (2WSP+rIPC: 30.4 ± 3.7%, 2W Saline: 30.7 ± 1.8%). With regards to inhalational

anesthesia, we found that halothane also significantly reduced (P<0.05) infarct size in the 2W of

protection compared to 1WSP controls (2W Halothane: 28.8 ± 2.1%) (figure 13a).

Interestingly, we observed that the 2W treatment groups did not reduce infarct size to the

same extent as 1WSP+rIPC, which aligns with consensus that cardioprotection from delayed

preconditioning is not as robust as the early phase[49]. Interestingly, despite the delayed benefits

of 2W N2O treatment on cardiac function, 40 minutes of nitrous oxide exposure did not reduce

infarct size after IR injury (2W N2O: 46.8 ± 2.5%). Therefore, these findings suggest that

treatment with intra-peritoneal injections or halothane anesthesia produces delayed protection

against cardiac dysfunction and cell damage after IR injury. Furthermore, while 2W N2O

preserved cardiac function, it did not protect against infarction. See figure 13 and 14 for infarct

size data and representative stained cross-sections.

70

A

B

Figure 13: The effects of intra-perionteal and inhalational anesthesia on left-ventricle infarct size

after IR injury. (A) Mean infarct sizes from each group. (B) Individual infarct sizes from each mouse. IR

injury was induced by 30 minutes of ischemia and 60 minutes of reperfusion. An * denotes a statistically

significant difference between treatment group and 1WSP controls (P<0.05). Data are expressed as mean

values ± SEM. (n=6-8 per treatment group). 1W-first window/early, 2W-second window/delayed, rIPC-

remote ischemic preconditioning, SP-sodium pentobarbital, N2O-nitrous oxide.

1WSP

1WSP+rIPC

2WSP

2WSP+rIPC

2WSaline

2WHalothane

2WN2O

0

10

20

30

40

50

60

70

80

90

Infa

rct S

ize

(% o

f LV

)

1WSP

1WSP+rI

PC2W

SP

2WSP+rI

PC

2WSali

ne

2WHalo

thane

2WN2O

0

10

20

30

40

50

60* (P<0.05)

Infa

rct S

ize

(% o

f LV)

71

Figure 14: Representative cross-sections of m

ouse hearts from each treatm

ent group after IR injury. A

t the end of reperfusion,

hearts were frozen at -80°C

. Hearts w

ere then cut into six 1 mm

slices and stained with triphenyltetrazolium

chloride (TTC) to

visualize the infarcted area. Viable m

yocardium stains red, w

hereas infarcted regions stain white.

72

4.3 Delayed Preconditioning with Injectable and Gas Anesthesia Increase phospho-Akt and phospho-p44/42 MAPK expression

I compared the effects of early and delayed preconditioning using rIPC, intra-peritoneal

and gas anesthesia on phospho-Akt (P-Akt) and phospho-p44/42 MAPK (P-p44/42) signaling

pathways in mouse hearts. Phosphorylation of Akt and p44/42 Erk proteins has been shown to be

involved in the cardioprotective phenotype induced by both IPC and APC [32], [74], [85].

Previous findings in our lab have shown that 1WSP+rIPC induces a significant (P<0.05) increase

in P-Akt (1.6 ± 0.1 fold increase) and P-Erk levels (1.6 ± 0.1 fold increase) compared to 1WSP

mouse hearts (figure 15, 16). In the current study, it was discovered that SP produces a

significant increase (P<0.05) in phospho-Akt expression in the 2W of protection (2WSP group)

compared to the 1WSP controls (3.2 ± 0.4 fold increase) (figure 15). A similar finding was seen

with delayed SP+rIPC treatment, with P-Akt levels significantly greater compared to 1WSP

hearts (3.1 ± 0.4 fold increase). To confirm whether SP was in fact causing delayed

cardioprotection, we tested the effects of an IP injection of 0.12 mL of saline solution. We found

that an IP injection of saline also caused a significant increase (P<0.05) in P-Akt levels

compared to 1WSP controls (2.7 ± 0.4 fold increase). Furthermore, we observed a delayed effect

of halothane and N2O anesthesia on P-Akt levels. Both gases augmented the expression of P-Akt,

however the increases were not significantly different from 1WSP P-Akt levels (2W halothane:

1.4 ± 0.2 fold, 2W N2O: 1.6 ± 0.3 fold increases compared to 1WSP) (figure 15).

We also examined the effects of 2WSP and 2W Saline on P-p44/42 MAPK expression in

hearts. While we observed an increase in P-p44/42 MAPK levels in 2WSP and 2WSP+rIPC

hearts, the expression levels were not significantly different from 1WSP hearts (2WSP: 1.4 ± 0.1

fold increase P=0.0640, 2WSP+rIPC: 1.5 ± 0.2 fold increase P=0.0788) (figure 16).

Additionally, there was no change in P-p44/42 MAPK levels in 2W Saline, or 2W N2O treated

73

hearts (2W Saline: 0.9 ± 0.2 fold, 2W N2O 1.0 ± 0.2 fold compared to 1WSP). However,

halothane anesthesia resulted in a significant increase in P-p44/42 MAPK levels in the 2W of

protection (1.3 ± 0.04 fold increase compared to 1WSP) (figure 16).

Consistent with the data on post-ischemic improvements in cardiac function and the

reductions in infarct size, these findings suggest that early and delayed forms of preconditioning

against IR injury are associated with the activation of pro-survival signaling pathways. The

differential expression of P-Akt and P-p44/42 in the various treatment groups suggests that the

preconditioning stimuli can recruit multiple pathways associated with cardioprotection.

74

Figure 15: The effects of intra-peritoneal and inhalational anesthesia on phospho-A

kt (Ser473) levels in mouse heart before IR

injury. D

ata are expressed as fold increases in p-Akt levels vs. 1W

SP controls. 1W groups represent treatm

ent with anesthetic±rIPC

15 m

inutes prior to IR injury. 1W

data shown on the left are from

previous work in the laboratory. 2W

groups represent treatment w

ith anesthetic 24 hours prior to IR

injury. SDS-PA

GE blots from

representative experiments are show

n below as scanned from

Image

Quant 5.0 softw

are. An * denotes a statistically significant difference betw

een treatment group and 1W

SP controls (P<0.05). Data are

expressed as mean values ± SEM

. (n=6-8 per treatment group). IR

- ischemia-reperfusion, 1W

-first window

/early, 2W-second

window

/delayed, rIPC-rem

ote ischemic preconditioning, SP-sodium

pentobarbital, N2 O

-nitrous oxide.

0.0#

1.0#

2.0#

3.0#

4.0#

P-A

kt (Ser473) 60kD

a

Total Akt 60kD

a

GA

PD

H 37kD

a

*

P=0.0678

1WSP

2W

SP

2WSP

+rIPC

1W

SP 2W

Saline

1WSP

2W

H

alothane 1W

SP 2W

N2 O

0.0#

1.0#

2.0#

3.0#

4.0#

phospho,Akt#Expression#(Ser473)#(Fold#vs.#Control)#

1WSP

1W

SP+rIP

C

*

*

75

Figure 16: The effects of intra-peritoneal and inhalational anesthesia on phospho-p44/42 M

APK

(Tyr202,T

hr204) levels in m

ouse hearts before IR injury. D

ata are expressed a fold increases in p-p44/42 MA

PK levels vs. 1W

SP controls. 1W groups

represent treatment w

ith anesthetic±rIPC 15 m

inutes prior to IR injury. 1W

data shown on the left are from

previous work in the lab.

2W groups represent treatm

ent with anesthetic 24 hours prior to IR

injury. SDS-PA

GE blots from

representative experiments are

shown below

as scanned from Im

age Quant 5.0 softw

are. An * denotes a statistically significant difference betw

een treatment group

and 1WSP controls (P<0.05). D

ata are expressed as mean values ± SEM

. (n=6-8 per treatment group). IR

- ischemia-reperfusion, 1W

-first w

indow/early, 2W

-second window

/delayed, rIPC-rem

ote ischemic preconditioning, SP-sodium

pentobarbital, N2 O

-nitrous oxide.

0.0#

0.5#

1.0#

1.5#

2.0#

0.0#

0.5#

1.0#

1.5#

2.0#

phospho,p44/42#MAPK#(Thr#202/Tyr204)#(Fold#vs.#Control)#

1WSP

2W

SP

2WSP

+rIPC

1W

SP 2W

Saline

1WSP

2W

H

alothane 1W

SP

2W N

2 O

1WSP

1W

SP+rIP

C

P-p44/42 M

AP

K (Thr202, Tyr204)

Total p44/42 MA

PK

GA

PD

H 37kD

a

P=0.0640

P=0.0788 *

*

76

Chapter 5 Discussion

Our understanding of the delayed phase of protection afforded by rIPC (defined as

protection arising 24-hours after initial stimulus) is scant in comparison with other forms of

cardioprotection. This is largely because there is currently no existing in-vivo or in-vitro model

for use in mouse studies. Therefore, the primary objective of the current study was to to identify

and establish a method of sedation for delayed rIPC experiments using the mouse Langendorff-

model of IR injury. Additional objectives that arose from our initial observations included (1)

investigating the delayed effects of SP anesthesia and IP injections on post-ischemic recovery of

mouse hearts and lastly to (2) examine the delayed effects of inhalational halothane and nitrous

oxide anesthesia in order to assess their suitability for use in future delayed rIPC studies.

The findings produced in this study were compared with our currently established mouse

isolated-heart model of early phase rIPC and its associated protection against global IR injury.

The present study offers several important findings that contribute to our understanding of

delayed rIPC and provide a base for future studies aiming to use the Langendorff technique.

The first important finding of this study is the observation that SP anesthesia, when given

via an IP injection, induces delayed preconditioning against global IR injury in mice. This was

shown by a decrease in post-ischemic cardiac dysfunction (i.e. improved LVDP, LVEDP,

dP/dtmax, dP/dtmin) and a reduction in LV infarct size. This unexpected observation arose during

my preliminary experiments, when I observed that there were no differences in cardiac recovery

after between 2WSP and 2WSP+rIPC treated mice, suggesting that SP may give rise to

cardioprotection. SP is the anesthetic of choice used as a first window control treatment for limb

ischemia and is required to apply transient ischemia. However, I have shown that its delayed

77

cardioprotective effect precludes its use as a control treatment and sedative for 2W

preconditioning studies.

A second important observation of this study was that an injection of saline fluid into the

mouse peritoneum also gave rise to delayed preconditioning, as hearts showed enhanced

functional recovery and reduced infarction after IR injury. While another lab group has produced

this finding in another study [179], a novel aspect finding from my experiments was the

observation of an accompanying cell signal signature archetypal of preconditioning. We found

that protection from IP injections was associated with an increase in phospho-Akt levels. This

implies that the irritation or stress involved with an IP injection may be the stimulus that results

in delayed preconditioning, as opposed to a direct effect from SP anesthesia.

The third aspect of this study was that halothane anesthesia induced delayed

preconditioning. I found that twenty-four hour prior treatment of 1-2% halothane resulted in

improved cardiac recovery and reductions in infarct size after global IR injury. Halothane

preconditioning was also associated with an increase in phospho-p44/42 MAPK levels.

Lastly, we discovered an unusual pattern of delayed cardioprotection 24 hours after 40

minutes exposure to nitrous oxide. Mice in this group displayed enhanced post-ischemic cardiac

functional recovery but did not show a preconditioning kinase signature, or a reduction in LV

infarct size. Instead, this group displayed similar levels of cell injury to that seen in 1WSP

control mice. This suggests that other mechanisms not related to the traditional preconditioning

phenotype, such as improvements in post-ischemic cardiomyocyte Ca2+ handling, may also be

involved in protecting cardiac function during IR injury.

78

5.1 Intra-Peritoneal Injections Induce Delayed Preconditioning against Global IR Injury

To my knowledge, there are no previous studies identifying a delayed preconditioning

effect from intra-peritoneal pentobarbital anesthesia. However, as I have shown, preconditioning

with pentobarbital may in fact be related to the method of injection as opposed to a direct effect

from the drug itself. As previously mentioned, pentobarbital has been used in a number of

studies examining the 1W of protection from rIPC, including our own, with no observable early

preconditioning effect[31]. Barbiturates are often the anesthetic of choice because of their

minimal cardiovascular depressive effects. However, Jiang et al. reported that pentobarbital leads

to a significant decrease in HR, LV peak systolic pressure, dP/dtmax in isolated hearts compared

to other intra-peritoneal anesthetics such as ketamine and chloral hydrate[118]. However, it was

never tested whether these depressive effects induce delayed preconditioning.

Li et al. examined the effects of delayed rIPC via bilateral hindlimb ischemia on infarct

size in isolated-mouse hearts[7]. They observed no delayed protection in sham mice sedated with

SP anesthesia via an IP injection, contradicting the results produced in the present study. In fact,

they were able to show that bilaterial hindlimb ischemia induces delayed preconditioning as

evidenced by a reduction in infarct size (35% in sham mice vs. 24% in delayed rIPC mice,

compared to 28-30% in the 2W IP injection groups in the present study). It should be noted that

the sham treated mice in their study exhibited a mean LV infarct size that is lower then what has

been reported in previous studies from our lab and others using a similar severity of IR injury, in

which mean infarct sizes are often >45%[9], [31]. This suggests that the control mice used in the

study by Li et al. may have also possessed some level of cardioprotection.

The same group published a study nearly a year later in which they reported a potential

preconditioning effect in mouse hearts after an IP injection. Labruto et al. found that

79

administration of Ringer’s solution via an IP injection, if given twenty-four hours prior to IR

injury, induces delayed cardioprotection by reducing infarction sizes and improving cardiac

performance[179]. They also showed no preconditioning effect after needle pricking, suggesting

that fluid within peritoneum is the required stimulus for inducing protection. We confirmed their

finding in the present study as we observed that mice that received a similar volume of saline via

IP injection had improved post-ischemic cardiac function, as evidenced by better recovery of

LVDP, dP/dtmax, dP/dtmin and attenuated increase of LVEDP. Mice in this group also showed

significant reductions in infarct size compared to 1WSP controls. Building on the Labruto et al.

study, we produced a novel finding in which we observed a three-fold increase in phospho-Akt

levels in mouse hearts that had received a saline injection twenty-fours prior to heart harvesting.

This cell signaling response may account for some of the observed cardioprotective benefits.

Further experiments using a kinase defective Akt mouse strain or an inhibitor of the upstream

PI3K (e.g. Wortmannin), would strongly indicate the role of Akt in IP injection-induced delayed

preconditioning.

Another essential kinase of the RISK pathway is p44/42 MAPK. While mice in the 2WSP

and 2WSP did exhibit a rise in phospho-p44/42 expression, the level of increase did not reach

statistical significance (P=0.0640 – 2WSP, P=0.0788 – 2WSP+rIPC). Furthermore, there were no

changes in phospho-p44/42 expression twenty-four hours after saline IP injection. This suggests

that the delayed preconditioning effects of an IP injection operate primarily via the Akt signaling

pathway. This finding also supports the notion that preconditioning can operate through multiple

pro-survival pathways available to the cell to yield the same cardioprotective effect.

A mechanism explaining how fluid in the peritoneum results in delayed preconditioning

at the myocardium has not been explored. In the same study, Labruto et al. examined the role of

NF-κB in the mouse heart and found that there was no increase in its nuclear-binding activity,

80

two hours after fluid injection. However, this does not eliminate a role for NF-κB as two hours

may have been too long after peak binding activity. Several studies have shown that NF-κB

migrates to the nucleus to increase gene expression of a variety of pro-inflammatory and cell

survival proteins as early as 15-min to 1 hour following the initial preconditioning stimulus by

rIPC and APC[7], [54]. We did not examine NF-κB in the hearts of mice that received saline

injections, leaving it open to future studies.

As described in the literature review section, one mechanism to explain the observed

cardioprotective effects from IP injections relates to the idea that a mild-inflammatory response

precedes the development of protection against IR injury. An IP injection may cause some

damage to the cells or blood vessels within the peritoneum, which may liberate ligands (e.g

DAMPs) that can activate the innate immune system. In a review article on the role of TLR4 in

ischemic tolerance, Kariko et al. describes how a sub-lethal stimulus, such as endotoxin, transient

ischemia or heat shock, generates a mild-inflammatory response, which is subsequently

suppressed with an increase in TLR4 negative-feedback regulators[22]. For example, necrosis or

injury to cells of the peritoneum might release endogenous ligands of TLR4, such as Hsps or

fibrin, which act on TLR4 in both inflammatory and non-immune related cells (cardiomyocytes,

endothelial cells). This could lead to the release of pro-inflammatory cytokines, but may also

increase the activity of negative regulators of the TLR signaling pathway such as PI3K – an

important component of the preconditioning phenotype. In the present study I showed that an IP

injection of saline causes an increase in phospho-Akt levels in the heart, which is a downstream

target of PI3K. Therefore, saline injection may have generated a mild-inflammatory response

within the peritoneum that resulted in the systemic suppression of TLR4 signaling by increasing

the activity of regulators that are also responsible for cardioprotection.

81

An added complexity to the issue of IP injections and their role in preconditioning is

related to the absence of any cardioprotective effects in an in-vivo model of IPC and IR injury.

The Bolli group conducted a study in which they developed an in-vivo mouse model of IPC via

transient ischemia to coronary arteries, in which mice were sedated with SP anesthesia via IP

injection[103]. They have used this model in a number of studies examining delayed

cardioprotection after IPC. In their initial study, they observed no early or delayed

preconditioning effect with SP treatment[103]. They also addressed the issue of delayed

preconditioning being caused by invasive surgical procedures by developing a sham group which

involved leaving the chest open for 60 minutes 24 hours prior to lethal IR injury. They found that

the infarct size was indistinguishable between this group and untreated controls.

There are several explanations to account for the contradictory results produced in our

study. Firstly, while it has been shown that rIPC and IPC generate a similar magnitude of

protection with regards to reductions in infarct size, there may be some differences in their

efficacy of inducing cardioprotection. Ahmed et al. compared the effects of local coronary IPC

with hindlimb rIPC on biochemical and histological changes in rats after IR injury[184]. They

found that local IPC was more effective at reducing post-ischemic levels of creatine kinase-MB

and lactate. There was also greater preservation of ATP levels and fewer episodes of ventricular

arrhythmias with IPC. While this group did not compare the effects of IPC and rIPC on infarct

size reductions or post-ischemic cardiac functional recovery, it may be that these additional

benefits from local IPC make it a more capable method of preconditioning above the increased

threshold set by an IP injection of SP anesthesia.

Secondly, the contrast between our study and the one conducted by the Bolli group could

be explained by the differences in the experimental protocol. In addition to the nature of the

preconditioning stimulus (local coronary IPC vs. limb rIPC), the differences in method of

82

ischemia (in-vivo, regional ischemia via CAO vs. ex-vivo global ischemia using Langendorff

preparation) and the absence of systemic influences (neural, hormonal and immune involvement

vs. isolated hearts) may account for the conflicting results produced in both studies.

5.2 Halothane Anesthesia Induces Delayed Preconditioning Against Global IR Injury The primary objective of this study was to develop a mouse model of delayed rIPC for

Langendorff isolated-heart studies of IR injury. My initial findings that an IP injection invokes

delayed preconditioning forced me to examine alternative methods of sedation to induce

transient hind limb ischemia in mice. Therefore, I used halothane and nitrous oxide anesthesia,

which have not been previously shown to induce ischemic tolerance in the delayed phase of

protection.

I examined the delayed effects of a 40-minute exposure to 1-2% halothane anesthesia on

post-ischemic cardiac function in isolated hearts. I found that halothane induced delayed

preconditioning by reducing LV infarct size and enhancing post-ischemic cardiac performance

that was comparable to the delayed benefits afforded with IP SP or saline administration. I also

showed that pre-treatment with halothane increases phospho-p44/42 MAPK levels in the delayed

phase of protection. The Redington lab group and others have shown the involvement of the

MAPK-protein family in the cardioprotective phenotype induced by rIPC and APC[85], [185].

The present finding suggests that halothane induces myocardial preconditioning by recruiting

similar signal transduction pathways. Halothane did not increase the levels of phospho-Akt,

indicating that similar to IP injections, there are multiple ways of generating cardioprotection.

Several studies have shown that administration of halothane anesthesia during IR injury

protects against post-ischemic cardiac dysfunction. This may be related to its potent negative

83

inotropic effects on cardiac contractile function and dose-related decreases in mean arterial blood

pressure. The mechanism of halothane protection during IR injury may involve a reduction in

Ca2+ overload during reperfusion by inhibiting the function of sarcolemmal L-type Ca2+ channels

[26]. Diminished Ca2+ overload may also occur through shortening of the action potential with

the opening of sarcolemmal K+ATP

channels. This ultimately results in the preservation of

myocardial ATP and creatine phosphate levels[186].

Previous studies have reported an early preconditioning effect from halothane exposure

with a decrease in infarct size, an effect which was abolished with adenosine receptor blockade,

using 8-phenyltheophylline, and PKC inhibition with chelerythrine [111]. It is well established

that PKC is upstream of NF-κB transcription factor activation and that this step is critical for the

development of delayed APC against IR injury[13], [32], [122], [123]. PKC activation is also

upstream of the p44/42 MAPK proteins, which we showed in the present study to be increased

with halothane exposure. Therefore, delayed cardioprotection with halothane may involve early

PKC activation of NF-κB, which subsequently leads to the transcription of genes responsible for

delayed preconditioning (Hsp, COX-2, iNOS etc). While detection of these markers was beyond

the scope of the present study, future work investigating delayed cardioprotection by halothane

can analyze whether this form of APC operates via similar mechanisms as other volatile

anesthetics.

5.3 Nitrous Oxide Improves Post-Ischemic Cardiac Performance but Does Not Reduce Infarction Size

I also examined the delayed effects of nitrous oxide on post-ischemic cardiac function to

determine its suitability as a method of anesthesia for 2W rIPC experiments. I found that twenty-

four hour prior exposure to 40 minutes of nitrous oxide at a 2:1 ratio with oxygen did not reduce

84

LV infarct size. Mice in this group showed comparable levels of cell injury to 1WSP treated

controls with an infarct size of approximately 45% of the total LV area. To my knowledge, this is

the first study to demonstrate that nitrous oxide does not have a delayed infarct-sparing effect

after IR injury.

Weber et al. reported that nitrous oxide does not induce an early preconditioning effect on

the heart. They showed that three cycles of 5 min of nitrous-oxide administration before

prolonged CAO does not reduce infarction sizes in rats. There was also no increase in the

phosphorylation or translocation of PKC-epsilon and PTK. Both proteins are known to play a

central role in mediating APC against IR injury and are also critical for the development of the

delayed phase of preconditioning[88], [93]. In the present study, there was no increase in p-

p44/42 MAPK or p-Akt levels with prior N2O treatment, confirming the findings of Weber et al.

that N2O does not recruit the traditional cell signaling pathways of IPC and APC.

Paradoxically, I observed that prior treatment with N2O enhanced post-ischemic cardiac

performance. LVEDP and LVDP showed similar improvements to the other forms of

preconditioning examined in this study. Rates of contraction and relaxation also approached pre-

ischemic values, although the magnitude of recovery for these parameters was intermediary

between other the preconditioning and 1WSP groups. These observations suggest that certain

preconditioning stimuli can benefit post-ischemic hearts without parallel improvements in both

cell survival and cardiac function. Several other studies have shown that IPC can cause

reductions in infarction size without improvements in cardiac function[187], [188]. However, the

present study shows that the opposite may also occur in that a conditioning stimulus can result in

recovery of cardiac function despite damage to heart tissue. This suggests that perhaps the

remaining viable tissue compensates in some fashion to maintain contractile function. While it

85

was out of the scope of this study to examine this phenomenon, a potential mechanism may

involve improvements in post-ischemic Ca2+ handling within the cardiomyocytes.

During systole, Ca2+ flows down its electrochemical gradient from the sarcoplasmic

reticulum (SR) through the coupling of sarcolemmal L-type voltage-gated and SR ryanodine

receptor Ca2+ channels. Relaxation occurs through active re-uptake of cytosolic Ca2+ into the SR

via the SR Ca2+-ATPase (SERCA2a)[189]. SERCA2a activity is regulated by the SR trans-

membrane protein phospholamban (PLN), which in its dephosphorylated state inhibits channel

activity and thus Ca2+ reuptake[190]. PLB is regulated by protein kinase A (PKA), which is

stimulated through sympathetic nervous system (SNS) activation. SNS activity raises

norepinephrine/epinephrine levels, which act on beta-adrenergic receptors (β-AR) on the heart to

increase PKA levels. PKA-inhibition of PLN leads to an increase in contractility and heart

rate[190].

The mechanism underlying how prior N2O administration improves post-ischemic Ca2+

handling, and thus contractile function, may involve N2O induced-inhibition of PLN. This may

occur in two ways. Firstly, while it is well established that nitrous oxide exerts dose-dependent

myocardial depression, there is some evidence suggesting that it may cause simultaneous bursts

of SNS activity[191], [192]. Previous studies have shown that 40-60% N2O can cause

intermittent increases in SNS outflow within 15-30 min of administration[192]. This is also

associated with an increase in plasma norepinephrine levels and systemic vascular resistance

[191]. As previously mentioned, increases in norepinephrine inhibit PLN via increased β-AR-

activation of PKA. Secondly, as part of its anti-nociceptive function, N2O causes an increase in

cell protein kinase G (PKG) activity via a neuronal nitric oxide (nNOS)-cyclic guanylyl cyclase

(cGMP)-signaling pathway[193]. Previous studies have shown that the cGMP-PKG pathway

may also play a role in protecting cardiomyocytes against IR injury through phosphorylation of

86

PLN. Gorbe et al. found that treatment of neonatal cardiomyocytes with a cGMP-analog or NO-

donor (S-nitroso-N-acetylpenicillamine – SNAP) decreased cell death after simulated IR injury,

and this was associated with increased the levels of phosphorylated PLN[194].

Therefore, the improvements in post-ischemic cardiac performance observed after

twenty-four hour prior treatment with N2O may operate via maintaining efficient Ca2+-cycling

through a PKA/PKG induced inhibition of PLN activity. The evidence presented here for a N2O-

induced inhibition of PLN via the PKA/PKG pathway comes from studies examining immediate

or early effects. While the present study did not evaluate the effects of N2O on PLN activity, it

may be that nitrous oxide alters the function of this regulatory protein in both the early and

delayed phases of protection. The N2O-induced kinase activation may lead to delayed changes in

gene expression of PLN or other Ca2+-cycling regulatory proteins that limit cardiac dysfunction

after IR injury. In the present study, we found that N2O led to partial recovery of the rates of

contraction and relaxation that were still significantly greater than 1WSP controls. Improvements

in Ca2+-cycling may account for these benefits, although this remains to be studied.

5.4 Cross-Talk Between Signaling Cascades

A recurring observation in the present study was that cardioprotection after IP injection or

exposure to inhalational anesthesia was associated with a statistically significant increase in

either phospho-Akt or phospho-p44/42, but not both. This indicates that the different stimuli

investigated in this study can activate either component of the RISK pathway to yield

cardioprotection. Nitrous oxide did not result in an increase in either kinase-signaling cascade,

which may account for the absence of infarct-sparing benefits.

Although only one component of the RISK pathway may have been activated after each

stimulus, it is likely that both kinase cascades are still important for mediating myocardial

87

protection against IR injury. The Yellon group has provided substantial evidence for the essential

role of the Akt and p44/42 kinases in mediating IPC-induced protection. They have shown that

both Akt and p44/42 are phosphorylated during the preconditioning phase and later reactivated at

the time of reperfusion[75]. In an earlier study, the same group described how the these kinase

pathways exhibit cross-talk during reperfusion, as inhibition of one kinase upregulates the

activity of the other pathway, acting in a compensatory fashion to ensure the cardioprotective

signal is relayed to downstream targets[195]. Interestingly, inhibition of either pathway abrogates

the cardioprotective effects of IPC on infarct size reduction, indicating the necessity for

activation of both pathways. Thus, while the present study did not assess the activation of Akt

and p44/42 during or after reperfusion, it is likely that both kinases are recruited at the onset of

reperfusion to mediate protection.

88

Chapter 6 Conclusion

In summary, the present study shows that sedation with SP anesthesia delivered via IP

injection and inhalational anesthesia results in delayed cardioprotection against global IR injury

in isolated mouse hearts, as evidenced by improved post-ischemic cardiac performance and

reduced infarction sizes. Treatment with nitrous oxide did not produce a delayed infarct sparing

effect but was associated with improved cardiac function after IR injury.

This study also showed that an IP injection of saline induced delayed cardioprotection,

indicating that this form of drug delivery should be avoided in future studies examining the 2W

of protection after rIPC in mice.

The finding that IP injections cause delayed preconditioning validates the primary

hypothesis of this study and clarifies my initial observation of a 2W cardioprotective effect from

SP anesthesia. IP injections reduced post-ischemic cardiac dysfunction and infarction sizes,

which was associated with an increase in phospho-Akt signaling. While this study did not

investigate how IP injections induce cardioprotection, one mechanism may involve injury to the

cells of the peritoneum, causing DAMP release and a mild inflammatory response. This stimulus

may trigger a shift from TLR4 pro-inflammatory signaling to an increase in negative feedback

inhibitors that are also known to mediate cardioprotection.

Based on these initial findings I aimed to survey alternative methods of anesthesia for use

in experiments on delayed protection after transient limb ischemia. I examined the effects of

halothane and nitrous oxide anesthesia, which hitherto had not been shown to cause 2W

cardioprotection. Halothane anesthesia led to delayed preconditioning with significant reductions

in infarction size and post-ischemic dysfunction after IR injury. This was accompanied by a cell

signal response involving p44/42 MAPK, which like Akt, is a member of the RISK pathway.

89

This aligned with my secondary hypothesis confirming halothane as a stimulus for delayed

preconditioning.

Finally, I found that treatment with nitrous oxide did not protect against infarction, but

did enhance post-ischemic cardiac recovery. Interestingly, unlike the other forms of anesthesia

explored in this study, there was no increase in phospho-Akt and phospho-p44/42 expression

twenty-four hours after treatment with nitrous oxide. While these findings do not align with the

established notion of preconditioning, it does indicate that other signaling mechanisms may

induce cardioprotection, possibly through changes in the function or expression of SR-Ca2+

regulatory proteins.

The primary aim of this study was to develop a mode of sedation that does not induce

cardioprotection in mice in order to establish a control treatment for future delayed rIPC

experiments. The present study highlights the importance of designing a study protocol that

controls for experimental stimuli that may induce or even block preconditioning via transient

ischemia. The type of anesthesia, method of delivery and conditions during sedation are all key

factors requiring careful consideration for myocardial preconditioning experiments. Nonetheless,

this study adds to the already well-known concept that preconditioning can be achieved from a

variety of stimuli.

90

Chapter 7 Future Directions

The findings from this study indicate that further work is required to develop a mouse

model of delayed rIPC for isolated-heart experiments examining myocardial protection against

IR injury.

However, the present investigation introduces several novel areas of study that will help

characterize the delayed preconditioning phenotype. Firstly, it is clear that a model of delayed

rIPC needs to include a control treatment that does not give rise to second window

preconditioning. This is not a simple task in the mouse because as I have shown, preconditioning

may occur from exposure to various stimuli. As such, future studies using a mouse model need

to investigate alternative methods of sedation or perhaps develop a means of inducing transient

ischemia in non-sedated mice.

There may be several methods to induce ischemia in a conscious mouse. Crawford et al.

have developed a method of applying hind limb ischemia using orthodontic rubber bands (ORB)

tied at proximal thigh level of mice[196]. I have begun testing the use of this method for delayed

rIPC experiments. Additionally, it may be possible to apply transient ischemia to the hind limb

while placing mice in a restraining tube with minimal stress or pain to the animal. This method is

commonly used for saphenous vein blood collection or tail-cuff blood pressure measurements in

mice[197].

Beyond developing a novel method of inducing transient ischemia, there are several other

aspects of the current study that require further investigation. The phenomenon that nitrous oxide

does not protect against infarction but does enhance post-ischemic cardiac function requires

further investigation. The role for improved Ca2+ handling through changes in the function or

expression of SR regulatory proteins has not been previously implicated in delayed

91

preconditioning. As such, future studies should examine the role for improved Ca2+ cycling,

particularly as it pertains to phospholamban regulation, as a potential novel means of protecting

the heart against post-ischemic cardiac dysfunction.

The present study was designed to survey a variety of methods of sedation to establish a

control treatment for delayed rIPC studies. As such, I did not explore the mechanisms behind the

various forms of preconditioning investigated in this study and have therefore provided several

avenues for further exploration.

7.1 An In-Vivo Model of Delayed rIPC

Future studies can also benefit from simultaneous investigations using an in-vivo model

of delayed rIPC and IR injury. The Langendorff-isolated heart technique is an invaluable tool to

examine the preconditioning phenotype as it provides cardiac-specific responses to IR injury.

However, an in-vivo model of IR injury with the added influence of the systemic response may

offer a more holistic, albeit more complex, depiction of cardioprotection. It is important to

investigate how rIPC affects the systemic inflammatory response during myocardial IR injury

and to examine the involvement of other organ systems (endocrine, peripheral/central nervous

system) in the resulting cardioprotective phenotype. Researchers can still examine cardiac

function in an in-vivo model of rIPC/IR injury through mouse echocardiography[198]. An in-

vivo model of delayed rIPC would also help to reconcile the conflicting results produced by the

current study and the findings generated by the Bolli et al. group. While there are a variety of

differences in experimental methodology between studies, it is important to confirm their finding

that sodium pentobarbital does not induce delayed cardiprotection against a lethal CAO.

92

7.2 Revisiting the role of TLR4 in delayed rIPC

It is becoming increasingly evident that the inflammatory response may be an important

component in the development of delayed cardioprotection. Therefore, future work in this area

can begin with addressing the initial hypothesis of this study that TLR4 is involved in the broad

physiological responses and signaling mechanisms of rIPC. Several studies have indicated that

preconditioning with sub-lethal doses of TLR4 ligands induces delayed cardioprotection against

IR injury. As discussed in the literature review section, these ligands activate a similar kinase

response mediating cardiprotection after IPC, which function to suppress TLR4 signaling and

therefore may inhibit a systemic inflammatory response. This is of profound importance as

reducing innate immune function may harmful in certain immune compromised disease states,

which may preclude the use of rIPC in these settings.

An investigation into the role of TLR4 can begin with examining the mechanism of how

IP injections cause delayed preconditioning. It is possible that the irritation or stress of fluid in

the peritoneum may induce an inflammatory response that serves as an adequate stimulus for the

developing subsequent second window protection against IR injury. Future studies can examine

whether circulating cytokines (TNF-α, IL-6) or TLR4-NF-κB signaling plays a role in this mode

of protection.

7.3 The ‘Third’ Window And Exercise Preconditioning The intriguing finding that a third or chronic window of preconditioning may exist has

spurred a great deal of interest in the additional phases of cardioprotection beyond the initial,

ephemeral first window. This interest has also been fueled by recent reports that chronic aerobic

exercise and repetitive rIPC confer cardioprotection through similar signaling mechanisms. As

93

reviewed in [199], [200], exercise-induced cardioprotection occurs through an increase in gene

expression of heat shock proteins, anti-oxidant enzymes and nitric-oxide synthase and through

adaptations in the coronary vasculature to facilitate perfusion. An important mechanism of the

cardioprotective phenotype after chronic exercise involves an increase in cell-stress induced

autophagy, the process by which cells remove damaged intra-cellular components[200].

Interestingly, Depre et al. showed that repetitive coronary artery occlusions or stenosis in a

porcine model reduces infarction through similar changes in gene expression and increases in

intracellular autophagy[95].

The Redington lab is interested in identifying whether there exists a reciprocal effect

between chronic exercise and repetitive rIPC, insofar that both modalities may modify the

resulting cardioprotective phenotype conferred after either form of preconditioning. The

existence of a third window of preconditioning has yet to be confirmed by other lab groups.

However, given the potential use of repeated RIPC stimuli for more chronic ischemic syndromes

(e.g. chronic stable angina, enhancement of exercise performance in heart failure) requires its

effects to be examined formally prior to widespread translation to the clinical arena. The present

study is an important first step for investigating the biology of chronic preconditioning in mice,

as these experiments will require a developed method of inducing transient ischemia in conscious

mice or employ the use of a non-cardioprotective sedative.

7.4 Clinical Implications of A Mouse Model of Delayed rIPC

Finally, the broader implications of developing a mouse model of delayed rIPC are the

ability for researchers to use this tool to examine the physiology of the second window of

cardioprotection and to better understand its therapeutic role in cardiovascular disease settings. It

is not surprising that the mouse, with the array of transgenic species available, has become a

94

popular animal model for examining the cellular and molecular basis of rIPC. This is especially

relevant given that delayed preconditioning is based on a genetic reprogramming that exploits the

evolutionary conserved stress-response available to cells – a property that also allows for a

longer duration and broader range of protection. These features have stimulated a great deal of

interest in developing novel strategies to mimic the delayed preconditioning phenotype, which

may involve the use of pharmacological agents or the transfer of genes to maintain a sustained

level of cardioprotection[88]. Such therapeutic approaches can have a strong impact both in and

out of the operating room and may be important for individuals at risk for cardiovascular disease.

This will require researchers to develop a stronger understanding of how the different

cardioprotective strategies interact with the pathological processes underlying various disease

conditions.

95

Chapter 8 References

[1] I. E. Konstantinov, S. Arab, J. Li, J. G. Coles, C. Boscarino, A. Mori, E. Cukerman, F. Dawood, M. M. H. Cheung, M. Shimizu, P. P. Liu, and A. N. Redington, “The remote ischemic preconditioning stimulus modifies gene expression in mouse myocardium.,” The Journal of Thoracic and Cardiovascular Surgery, vol. 130, no. 5, pp. 1326–1332, Nov. 2005.

[2] I. E. Konstantinov, S. Arab, R. K. Kharbanda, J. Li, M. M. H. Cheung, V. Cherepanov, G. P. Downey, P. P. Liu, E. Cukerman, J. G. Coles, and A. N. Redington, “The remote ischemic preconditioning stimulus modifies inflammatory gene expression in humans.,” Physiol. Genomics, vol. 19, no. 1, pp. 143–150, Sep. 2004.

[3] M. Shimizu, P. Saxena, I. E. Konstantinov, V. Cherepanov, M. M. H. Cheung, P. Wearden, H. Zhangdong, M. Schmidt, G. P. Downey, and A. N. Redington, “Remote ischemic preconditioning decreases adhesion and selectively modifies functional responses of human neutrophils.,” J. Surg. Res., vol. 158, no. 1, pp. 155–161, Jan. 2010.

[4] G. Heusch, J. Musiolik, E. Kottenberg, J. Peters, H. Jakob, and M. Thielmann, “STAT5 Activation and Cardioprotection by Remote Ischemic Preconditioning in Humans.,” Circ. Res., Nov. 2011.

[5] K. L. Redington, T. Disenhouse, S. C. Strantzas, R. Gladstone, C. Wei, M. B. Tropak, X. Dai, C. Manlhiot, J. Li, and A. N. Redington, “Remote cardioprotection by direct peripheral nerve stimulation and topical capsaicin is mediated by circulating humoral factors.,” Basic Res. Cardiol., vol. 107, no. 2, pp. 1–10, Mar. 2012.

[6] S.-Z. Zhang, N.-F. Wang, J. Xu, Q. Gao, G.-H. Lin, I. C. Bruce, and Q. Xia, “Kappa-opioid receptors mediate cardioprotection by remote preconditioning.,” Anesthesiology, vol. 105, no. 3, pp. 550–556, Sep. 2006.

[7] G. Li, F. Labruto, A. Sirsjö, F. Chen, J. Vaage, and G. Valen, “Myocardial protection by remote preconditioning: the role of nuclear factor kappa-B p105 and inducible nitric oxide synthase.,” Eur J Cardiothorac Surg, vol. 26, no. 5, pp. 968–973, Nov. 2004.

[8] B. Dawn, Y. Guo, A. Rezazadeh, O.-L. Wang, A. B. Stein, G. Hunt, J. Varma, Y.-T. Xuan, W.-J. Wu, W. Tan, X. Zhu, and R. Bolli, “Tumor necrosis factor-alpha does not modulate ischemia/reperfusion injury in naïve myocardium but is essential for the development of late preconditioning.,” Journal of Molecular and Cellular Cardiology, vol. 37, no. 1, pp. 51–61, Jul. 2004.

[9] M. P. Flaherty, Y. Guo, S. Tiwari, A. Rezazadeh, G. Hunt, S. K. Sanganalmath, X.-L. Tang, R. Bolli, and B. Dawn, “The role of TNF-alpha receptors p55 and p75 in acute myocardial ischemia/reperfusion injury and late preconditioning.,” Journal of Molecular and Cellular Cardiology, vol. 45, no. 6, pp. 735–741, Dec. 2008.

[10] S. L. Stevens, P. Y. Leung, K. B. Vartanian, B. Gopalan, T. Yang, R. P. Simon, and M. P. Stenzel-Poore, “Multiple preconditioning paradigms converge on interferon regulatory factor-dependent signaling to promote tolerance to ischemic brain injury.,” J. Neurosci., vol. 31, no. 23, pp. 8456–8463, Jun. 2011.

[11] N. Yamashita, S. Hoshida, K. Otsu, N. Taniguchi, T. Kuzuya, and M. Hori, “The involvement of cytokines in the second window of ischaemic preconditioning,” British Journal of Pharmacology, vol. 131, no. 3, pp. 415–422, 2000.

96

[12] J.-W. Dong, J. G. Vallejo, H.-P. Tzeng, J. A. Thomas, and D. L. Mann, “Innate immunity mediates myocardial preconditioning through Toll-like receptor 2 and TIRAP-dependent signaling pathways.,” Am. J. Physiol. Heart Circ. Physiol., vol. 298, no. 3, pp. H1079–87, Mar. 2010.

[13] Y. T. Xuan, X. L. Tang, S. Banerjee, H. Takano, R. C. Li, H. Han, Y. Qiu, J. J. Li, and R. Bolli, “Nuclear factor-kappaB plays an essential role in the late phase of ischemic preconditioning in conscious rabbits.,” Circ. Res., vol. 84, no. 9, pp. 1095–1109, May 1999.

[14] S. P. Loukogeorgakis, A. T. Panagiotidou, M. W. Broadhead, A. Donald, J. E. Deanfield, and R. J. MacAllister, “Remote ischemic preconditioning provides early and late protection against endothelial ischemia-reperfusion injury in humans: role of the autonomic nervous system.,” Journal of the American College of Cardiology, vol. 46, no. 3, pp. 450–456, Aug. 2005.

[15] Y. Wang, B. Yin, S. Liu, and S. Xue, “Cardioprotective Effect by Tumor Necrosis Factor-α and Interleukin-6 through Late Preconditioning in Unstable Angina Patients,” Archives of Medical Research, vol. 38, no. 1, pp. 80–85, Jan. 2007.

[16] N. Smart, M. H. Mojet, D. S. Latchman, M. S. Marber, M. R. Duchen, and R. J. Heads, “IL-6 induces PI 3-kinase and nitric oxide-dependent protection and preserves mitochondrial function in cardiomyocytes.,” Cardiovasc. Res., vol. 69, no. 1, pp. 164–177, Jan. 2006.

[17] T. Ha, F. Hua, X. Liu, J. Ma, J. R. McMullen, T. Shioi, S. Izumo, J. Kelley, X. Gao, W. Browder, D. L. Williams, R. L. Kao, and C. Li, “Lipopolysaccharide-induced myocardial protection against ischaemia/reperfusion injury is mediated through a PI3K/Akt-dependent mechanism.,” Cardiovasc. Res., vol. 78, no. 3, pp. 546–553, Jun. 2008.

[18] T. Ha, Y. Hu, L. Liu, C. Lu, J. R. McMullen, J. Kelley, R. L. Kao, D. L. Williams, X. Gao, and C. Li, “TLR2 ligands induce cardioprotection against ischaemia/reperfusion injury through a PI3K/Akt-dependent mechanism.,” Cardiovasc. Res., vol. 87, no. 4, pp. 694–703, Sep. 2010.

[19] M. A. Pavione, F. Carmona, M. de Castro, and A. P. C. P. Carlotti, “Late remote ischemic preconditioning in children undergoing cardiopulmonary bypass: A randomized controlled trial.,” The Journal of Thoracic and Cardiovascular Surgery, Jan. 2012.

[20] J. M. Pradillo, D. Fernández-López, I. García-Yébenes, M. Sobrado, O. Hurtado, M. A. Moro, and I. Lizasoain, “Toll-like receptor 4 is involved in neuroprotection afforded by ischemic preconditioning.,” J. Neurochem., vol. 109, no. 1, pp. 287–294, Apr. 2009.

[21] G. Valen, “Extracardiac approaches to protecting the heart,” Eur J Cardiothorac Surg, vol. 35, no. 4, pp. 651–657, 2009.

[22] K. Karikó, D. Weissman, and F. A. Welsh, “Inhibition of toll-like receptor and cytokine signaling--a unifying theme in ischemic tolerance.,” J. Cereb. Blood Flow Metab., vol. 24, no. 11, pp. 1288–1304, Nov. 2004.

[23] S. Frantz, G. Ertl, and J. Bauersachs, “Mechanisms of disease: Toll-like receptors in cardiovascular disease.,” Nat Clin Pract Cardiovasc Med, vol. 4, no. 8, pp. 444–454, Aug. 2007.

[24] W. Schlack and B. Preckel, “Effects of halothane, enflurane, isoflurane, sevoflurane and desflurane on myocardial reperfusion injury in the isolated rat heart.,” British journal of …, 1998.

97

[25] M. Zaugg, E. Lucchinetti, M. Uecker, T. Pasch, and M. Schaub, “Anaesthetics and cardiac preconditioning. Part I. Signalling and cytoprotective mechanisms,” British Journal of Anaesthesia, vol. 91, no. 4, p. 551, 2003.

[26] A. Lochner, I. S. Harper, R. Salie, S. Genade, and A. R. Coetzee, “Halothane protects the isolated rat myocardium against excessive total intracellular calcium and structural damage during ischemia and reperfusion.,” Anesth. Analg., vol. 79, no. 2, pp. 226–233, Aug. 1994.

[27] C. Kowalski, S. Zahler, B. F. Becker, A. Flaucher, P. F. Conzen, E. Gerlach, and K. Peter, “Halothane, isoflurane, and sevoflurane reduce postischemic adhesion of neutrophils in the coronary system.,” Anesthesiology, vol. 86, no. 1, pp. 188–195, Jan. 1997.

[28] W. Chao, Y. Shen, X. Zhu, H. Zhao, M. Novikov, U. Schmidt, and A. Rosenzweig, “Lipopolysaccharide improves cardiomyocyte survival and function after serum deprivation.,” J. Biol. Chem., vol. 280, no. 23, pp. 21997–22005, Jun. 2005.

[29] E. Kottenberg, M. Thielmann, L. Bergmann, T. Heine, H. Jakob, G. Heusch, and J. Peters, “Protection by remote ischemic preconditioning during coronary artery bypass graft surgery with isoflurane but not propofol - a clinical trial.,” Acta Anaesthesiol Scand, vol. 56, no. 1, pp. 30–38, Jan. 2012.

[30] W. Chao, “Toll-like receptor signaling: a critical modulator of cell survival and ischemic injury in the heart.,” Am. J. Physiol. Heart Circ. Physiol., vol. 296, no. 1, pp. H1–12, Jan. 2009.

[31] J. Li, W. Xuan, R. Yan, M. B. Tropak, E. Jean St Michel, W. Liang, R. Gladstone, P. H. Backx, R. K. Kharbanda, and A. N. Redington, “Remote preconditioning provides potent cardioprotection via PI3K/Akt activation and is associated with nuclear accumulation of β-catenin.,” Clin. Sci., vol. 120, no. 10, pp. 451–462, May 2011.

[32] N. C. Weber and W. Schlack, “Inhalational anaesthetics and cardioprotection.,” Handb Exp Pharmacol, no. 182, pp. 187–207, 2008.

[33] D. M. Yellon and D. J. Hausenloy, “Myocardial reperfusion injury,” New England Journal of Medicine, vol. 357, no. 11, pp. 1121–1135, 2007.

[34] A. Frank, M. Bonney, S. Bonney, L. Weitzel, M. Koeppen, and T. Eckle, “Myocardial Ischemia Reperfusion Injury: From Basic Science to Clinical Bedside.,” Semin Cardiothorac Vasc Anesth, Feb. 2012.

[35] A. T. Turer and J. A. Hill, “Pathogenesis of myocardial ischemia-reperfusion injury and rationale for therapy.,” Am. J. Cardiol., vol. 106, no. 3, pp. 360–368, Aug. 2010.

[36] F. Arslan, D. P. de Kleijn, and G. Pasterkamp, “Innate immune signaling in cardiac ischemia,” Nature Publishing Group, vol. 8, no. 5, pp. 292–300, Mar. 2011.

[37] D. J. Hausenloy, E. Boston-Griffiths, and D. M. Yellon, “Cardioprotection during cardiac surgery.,” Cardiovasc. Res., Mar. 2012.

[38] C. E. Murry, R. B. Jennings, and K. A. Reimer, “Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium.,” Circulation, vol. 74, no. 5, pp. 1124–1136, Nov. 1986.

[39] J. Kaszaki, A. Wolfárd, L. Szalay, and M. Boros, “Pathophysiology of Ischemia-Reperfusion Injury,” Transplantation Proceedings, vol. 38, no. 3, pp. 826–828, Apr. 2006.

[40] A. Skyschally, R. Schulz, and G. Heusch, “Pathophysiology of myocardial infarction,” Herz, vol. 33, no. 2, pp. 88–100, 2008.

98

[41] C. Penna, D. Mancardi, R. Rastaldo, and P. Pagliaro, “Cardioprotection: a radical view Free radicals in pre and postconditioning.,” Biochim. Biophys. Acta, vol. 1787, no. 7, pp. 781–793, Jul. 2009.

[42] F. Arslan, B. Keogh, P. McGuirk, and A. E. Parker, “TLR2 and TLR4 in ischemia reperfusion injury.,” Mediators Inflamm., vol. 2010, p. 704202, 2010.

[43] E. Murphy and C. Steenbergen, “Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury.,” Physiol. Rev., vol. 88, no. 2, pp. 581–609, Apr. 2008.

[44] D. Carden, “Pathophysiology of ischaemia–reperfusion injury - Carden - 2000 - The Journal of Pathology - Wiley Online Library,” The Journal of pathology, 2000.

[45] L. Toledo-Pereyra and F. Lopez-Neblina, “Reactive oxygen species and molecular biology... [Ann Transplant. 2004] - PubMed - NCBI,” … : quarterly of the Polish …, 2004.

[46] S. Frantz, R. A. Kelly, and T. Bourcier, “Role of TLR-2 in the activation of nuclear factor kappaB by oxidative stress in cardiac myocytes.,” J. Biol. Chem., vol. 276, no. 7, pp. 5197–5203, Feb. 2001.

[47] J. Jordan and Z. Zhao, “The role of neutrophils in myocardial ischemia–reperfusion injury,” Cardiovascular …, 1999.

[48] L. Ao, N. Zou, J. C. Cleveland, D. A. Fullerton, and X. Meng, “Myocardial TLR4 is a determinant of neutrophil infiltration after global myocardial ischemia: mediating KC and MCP-1 expression induced by extracellular HSC70.,” Am. J. Physiol. Heart Circ. Physiol., vol. 297, no. 1, pp. H21–8, Jul. 2009.

[49] D. M. Yellon and J. M. Downey, “Preconditioning the myocardium: from cellular physiology to clinical cardiology.,” Physiol. Rev., vol. 83, no. 4, pp. 1113–1151, Oct. 2003.

[50] S. A. Tavener and P. Kubes, “Cellular and molecular mechanisms underlying LPS-associated myocyte impairment.,” Am. J. Physiol. Heart Circ. Physiol., vol. 290, no. 2, pp. H800–6, Feb. 2006.

[51] M. E. Bianchi, “DAMPs, PAMPs and alarmins: all we need to know about danger.,” J. Leukoc. Biol., vol. 81, no. 1, pp. 1–5, Jan. 2007.

[52] F. Arslan, M. B. Smeets, L. A. J. O'Neill, B. Keogh, P. McGuirk, L. Timmers, C. Tersteeg, I. E. Hoefer, P. A. Doevendans, G. Pasterkamp, and D. P. V. de Kleijn, “Myocardial ischemia/reperfusion injury is mediated by leukocytic toll-like receptor-2 and reduced by systemic administration of a novel anti-toll-like receptor-2 antibody.,” Circulation, vol. 121, no. 1, pp. 80–90, Jan. 2010.

[53] A. Linde, D. Mosier, F. Blecha, and T. Melgarejo, “Innate immunity and inflammation--New frontiers in comparative cardiovascular pathology.,” Cardiovasc. Res., vol. 73, no. 1, pp. 26–36, Jan. 2007.

[54] G. Valen and Z. Yan, “Nuclear factor kappa-B and the heart,” Journal of the American College of Cardiology, vol. 38, no. 2, p. 307, 2001.

[55] J. H. Boyd, S. Mathur, Y. Wang, R. M. Bateman, and K. R. Walley, “Toll-like receptor stimulation in cardiomyoctes decreases contractility and initiates an NF-kappaB dependent inflammatory response.,” Cardiovasc. Res., vol. 72, no. 3, pp. 384–393, Dec. 2006.

[56] P. Balakumar and N. K. Sharma, “Healing the diabetic heart: Does myocardial preconditioning work?,” Cellular Signalling, pp. 1–7, Sep. 2011.

[57] P. Ferdinandy, R. Schulz, and G. F. Baxter, “Interaction of cardiovascular risk factors with myocardial ischemia/reperfusion injury, preconditioning, and postconditioning.,” Pharmacol. Rev., vol. 59, no. 4, pp. 418–458, Dec. 2007.

99

[58] K. Przyklenk, B. Bauer, M. Ovize, R. A. Kloner, and P. Whittaker, “Regional ischemic ‘preconditioning’ protects remote virgin myocardium from subsequent sustained coronary occlusion.,” Circulation, vol. 87, no. 3, pp. 893–899, Mar. 1993.

[59] K. Tanaka, L. M. Ludwig, J. R. Kersten, P. S. Pagel, and D. C. Warltier, “Mechanisms of Cardioprotection by Volatile Anesthetics,” Anesthesiology, vol. 100, no. 3, p. 707, Mar. 2004.

[60] T. Pell, G. Baxter, and D. Yellon, “Renal ischemia preconditions myocardium: role of adenosine receptors and ATP-sensitive potassium channels,” American Journal of …, 1998.

[61] S. Tokuno, K. Hinokiyama, K. Tokuno, C. Löwbeer, L.-O. Hansson, and G. Valen, “Spontaneous ischemic events in the brain and heart adapt the hearts of severely atherosclerotic mice to ischemia.,” Arterioscler. Thromb. Vasc. Biol., vol. 22, no. 6, pp. 995–1001, Jun. 2002.

[62] B. Gho, R. Schoemaker, and M. van den Doel, “Myocardial Protection by Brief Ischemia in Noncardiac Tissue,” Circulation, 1996.

[63] R. Huda, D. H. Chung, and M. Mathru, “Ischemic preconditioning at a distance: altered gene expression in mouse heart and other organs following brief occlusion of the mesenteric artery.,” Heart Lung Circ, vol. 14, no. 1, pp. 36–43, Mar. 2005.

[64] H. H. Patel, J. Moore, A. K. Hsu, and G. J. Gross, “Cardioprotection at a distance: mesenteric artery occlusion protects the myocardium via an opioid sensitive mechanism.,” Journal of Molecular and Cellular Cardiology, vol. 34, no. 10, pp. 1317–1323, Oct. 2002.

[65] Y. Birnbaum and S. Hale, “Ischemic Preconditioning at a Distance : Reduction of Myocardial Infarct Size by Partial Reduction of Blood Supply Combined With Rapid Stimulation of the Gastrocnemius Muscle in the Rabbit,” Circulation, 1997.

[66] T. Oxman, M. Arad, R. Klein, N. Avazov, and B. Rabinowitz, “Limb ischemia preconditions the heart against reperfusion tachyarrhythmia.,” Am. J. Physiol., vol. 273, no. 4, pp. H1707–12, Oct. 1997.

[67] R. Kharbanda, U. Mortensen, P. White, S. Kristiansen, M. Schmidt, J. Hoschtitzky, M. Vogel, K. Sorensen, A. Redington, and R. MacAllister, “Transient limb ischemia induces remote ischemic preconditioning in vivo,” Circulation, vol. 106, no. 23, pp. 2881–2883, 2002.

[68] D. J. Hausenloy and D. M. Yellon, “Preconditioning and postconditioning: underlying mechanisms and clinical application.,” Atherosclerosis, vol. 204, no. 2, pp. 334–341, Jun. 2009.

[69] R. K. Kharbanda, T. T. Nielsen, and A. N. Redington, “Translation of remote ischaemic preconditioning into clinical practice.,” Lancet, vol. 374, no. 9700, pp. 1557–1565, Oct. 2009.

[70] E. Murphy, “Primary and secondary signaling pathways in early preconditioning that converge on the mitochondria to produce cardioprotection.,” Circ. Res., vol. 94, no. 1, pp. 7–16, Jan. 2004.

[71] D. J. Hausenloy and D. M. Yellon, “Remote ischaemic preconditioning: underlying mechanisms and clinical application.,” Cardiovasc. Res., vol. 79, no. 3, pp. 377–386, Aug. 2008.

[72] G. J. Gross, “Role of opioids in acute and delayed preconditioning.,” Journal of Molecular and Cellular Cardiology, vol. 35, no. 7, pp. 709–718, Jul. 2003.

[73] S. P. Jones and R. Bolli, “The ubiquitous role of nitric oxide in cardioprotection.,” Journal of Molecular and Cellular Cardiology, vol. 40, no. 1, pp. 16–23, Jan. 2006.

100

[74] Y. Fujio, T. Nguyen, D. Wencker, R. N. Kitsis, and K. Walsh, “Akt promotes survival of cardiomyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart.,” Circulation, vol. 101, no. 6, pp. 660–667, Feb. 2000.

[75] D. J. Hausenloy, A. Tsang, M. M. Mocanu, and D. M. Yellon, “Ischemic preconditioning protects by activating prosurvival kinases at reperfusion.,” Am. J. Physiol. Heart Circ. Physiol., vol. 288, no. 2, pp. H971–6, Feb. 2005.

[76] N. Tapuria, Y. Kumar, M. M. Habib, M. Abu Amara, A. M. Seifalian, and B. R. Davidson, “Remote ischemic preconditioning: a novel protective method from ischemia reperfusion injury--a review.,” J. Surg. Res., vol. 150, no. 2, pp. 304–330, Dec. 2008.

[77] P. Saxena, M. A. J. Newman, J. S. Shehatha, A. N. Redington, and I. E. Konstantinov, “Remote ischemic conditioning: evolution of the concept, mechanisms, and clinical application.,” J Card Surg, vol. 25, no. 1, pp. 127–134, 2010.

[78] C. Li, T. Ha, J. Kelley, X. Gao, Y. Qiu, R. L. Kao, W. Browder, and D. L. Williams, “Modulating Toll-like receptor mediated signaling by (1-->3)-beta-D-glucan rapidly induces cardioprotection.,” Cardiovasc. Res., vol. 61, no. 3, pp. 538–547, Feb. 2004.

[79] T. Fukao and S. Koyasu, “PI3K and negative regulation of TLR signaling,” Trends in Immunology, vol. 24, no. 7, pp. 358–363, 2003.

[80] R. K. Kharbanda, “Cardiac conditioning: a review of evolving strategies to reduce ischaemia-reperfusion injury.,” Heart, vol. 96, no. 15, pp. 1179–1186, Aug. 2010.

[81] D. J. Hausenloy and D. M. Yellon, “Reperfusion injury salvage kinase signalling: taking a RISK for cardioprotection.,” Heart Fail Rev, vol. 12, no. 3, pp. 217–234, Dec. 2007.

[82] A. Hashimoto, G. Miyakoda, Y. Hirose, and T. Mori, “Activation of endothelial nitric oxide synthase by cilostazol via a cAMP/protein kinase A- and phosphatidylinositol 3-kinase/Akt-dependent mechanism.,” Atherosclerosis, vol. 189, no. 2, pp. 350–357, Dec. 2006.

[83] A. M. Zeiher, S. Dimmeler, I. Fleming, B. Fisslthaler, C. Hermann, and R. Busse, “Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation,” Nature, vol. 399, no. 6736, pp. 601–605, Jun. 1999.

[84] H. Tong, K. Imahashi, C. Steenbergen, and E. Murphy, “Phosphorylation of glycogen synthase kinase-3beta during preconditioning through a phosphatidylinositol-3-kinase--dependent pathway is cardioprotective.,” Circ. Res., vol. 90, no. 4, pp. 377–379, Mar. 2002.

[85] M. Shimizu, M. Tropak, R. J. Diaz, F. Suto, H. Surendra, E. Kuzmin, J. Li, G. Gross, G. J. Wilson, J. Callahan, and A. N. Redington, “Transient limb ischaemia remotely preconditions through a humoral mechanism acting directly on the myocardium: evidence suggesting cross-species protection.,” Clin. Sci., vol. 117, no. 5, pp. 191–200, Sep. 2009.

[86] H. Tong, W. Chen, and C. Steenbergen, “Ischemic Preconditioning Activates Phosphatidylinositol-3-Kinase Upstream of Protein Kinase C,” Circ. Res., 2000.

[87] A. Tsang, D. J. Hausenloy, M. M. Mocanu, and D. M. Yellon, “Postconditioning: a form of ‘modified reperfusion’ protects the myocardium by activating the phosphatidylinositol 3-kinase-Akt pathway.,” Circ. Res., vol. 95, no. 3, pp. 230–232, Aug. 2004.

[88] A. B. Stein, X.-L. Tang, Y. Guo, Y.-T. Xuan, B. Dawn, and R. Bolli, “Delayed adaptation of the heart to stress: late preconditioning.,” Stroke, vol. 35, no. 11, pp. 2676–2679, Nov. 2004.

101

[89] M. Heidbreder, A. Naumann, K. Tempel, P. Dominiak, and A. Dendorfer, “Remote vs. ischaemic preconditioning: the differential role of mitogen-activated protein kinase pathways.,” Cardiovasc. Res., vol. 78, no. 1, pp. 108–115, Apr. 2008.

[90] C. P. Baines, J. Zhang, G.-W. Wang, Y.-T. Zheng, J. X. Xiu, E. M. Cardwell, R. Bolli, and P. Ping, “Mitochondrial PKCepsilon and MAPK form signaling modules in the murine heart: enhanced mitochondrial PKCepsilon-MAPK interactions and differential MAPK activation in PKCepsilon-induced cardioprotection.,” Circ. Res., vol. 90, no. 4, pp. 390–397, Mar. 2002.

[91] J. Raphael, “Physiology and Pharmacology of Myocardial Preconditioning,” Semin Cardiothorac Vasc Anesth, vol. 14, no. 1, pp. 54–59, May 2010.

[92] M. Marber, D. Latchman, and J. Walker, “Cardiac stress protein elevation 24 hours after brief ischemia or heat stress is associated with resistance to myocardial infarction,” Circulation, 1993.

[93] R. Bolli, “The late phase of preconditioning,” Circ. Res., vol. 87, no. 11, pp. 972–983, 2000.

[94] D. J. Hausenloy and D. M. Yellon, “The second window of preconditioning (SWOP) where are we now?,” Cardiovasc Drugs Ther, vol. 24, no. 3, pp. 235–254, Jun. 2010.

[95] C. Depre, J. Y. Park, Y.-T. Shen, X. Zhao, H. Qiu, L. Yan, B. Tian, S. F. Vatner, and D. E. Vatner, “Molecular mechanisms mediating preconditioning following chronic ischemia differ from those in classical second window.,” Am. J. Physiol. Heart Circ. Physiol., vol. 299, no. 3, pp. H752–62, Sep. 2010.

[96] A. Rizvi, X. L. Tang, Y. Qiu, Y. T. Xuan, H. Takano, A. K. Jadoon, and R. Bolli, “Increased protein synthesis is necessary for the development of late preconditioning against myocardial stunning.,” Am. J. Physiol., vol. 277, no. 3, pp. H874–84, Sep. 1999.

[97] Y. Nishino, “Ischemic preconditioning activates AMPK in a PKC-dependent manner and induces GLUT4 up-regulation in the late phase of cardioprotection,” Cardiovasc. Res., vol. 61, no. 3, pp. 610–619, Feb. 2004.

[98] B. Dawn, H. Takano, and X. Tang, “Role of Src protein tyrosine kinases in late preconditioning against myocardial infarction,” American Journal of …, 2002.

[99] H. Takano, R. Li, H. Han, Y. Qiu, J. Li, and R. Bolli, “Nuclear factor-B plays an essential role in the late phase of ischemic preconditioning in conscious rabbits,” Circ. Res., 1999.

[100] J. A. Romashkova and S. S. Makarov, “NF-kappaB is a target of AKT in anti-apoptotic PDGF signalling.,” Nature, vol. 401, no. 6748, pp. 86–90, Sep. 1999.

[101] Y. Wang, M. Kudo, M. Xu, A. Ayub, and M. Ashraf, “Mitochondrial KATPChannel as an End Effector of Cardioprotection During Late Preconditioning: Triggering Role of Nitric Oxide,” Journal of Molecular and Cellular Cardiology, vol. 33, no. 11, pp. 2037–2046, 2001.

[102] H. Patel, E. Gross, and J. Peart, “Sarcolemmal KATP channel triggers delayed ischemic preconditioning in rats,” American Journal of …, 2005.

[103] Y. Guo, W. J. Wu, Y. Qiu, X. L. Tang, Z. Yang, and R. Bolli, “Demonstration of an early and a late phase of ischemic preconditioning in mice,” Am. J. Physiol. Heart Circ. Physiol., vol. 275, no. 4, pp. H1375–H1387, 1998.

[104] R. Bolli, Q.-H. Li, X.-L. Tang, Y. Guo, Y.-T. Xuan, G. Rokosh, and B. Dawn, “The late phase of preconditioning and its natural clinical application--gene therapy.,” Heart Fail Rev, vol. 12, no. 3, pp. 189–199, Dec. 2007.

102

[105] M. M. H. Cheung, R. K. Kharbanda, I. E. Konstantinov, M. Shimizu, H. Frndova, J. Li, H. M. Holtby, P. N. Cox, J. F. Smallhorn, G. S. Van Arsdell, and A. N. Redington, “Randomized controlled trial of the effects of remote ischemic preconditioning on children undergoing cardiac surgery: first clinical application in humans.,” Journal of the American College of Cardiology, vol. 47, no. 11, pp. 2277–2282, Jun. 2006.

[106] D. J. Hausenloy, P. K. Mwamure, V. Venugopal, J. Harris, M. Barnard, E. Grundy, E. Ashley, S. Vichare, C. Di Salvo, S. Kolvekar, M. Hayward, B. Keogh, R. J. MacAllister, and D. M. Yellon, “Effect of remote ischaemic preconditioning on myocardial injury in patients undergoing coronary artery bypass graft surgery: a randomised controlled trial.,” Lancet, vol. 370, no. 9587, pp. 575–579, Aug. 2007.

[107] A. K. Roscoe, J. D. Christensen, and C. I. Lynch, “Isoflurane, but not Halothane, Induces Protection of Human Myocardium via Adenosine A1 Receptors and Adenosine Triphosphate–sensitive Potassium Channels,” Anesthesiology, vol. 92, no. 6, p. 1692, Jun. 2000.

[108] S. G. De Hert, “Volatile Anesthetics and Cardiac Function,” Semin Cardiothorac Vasc Anesth, vol. 10, no. 1, pp. 33–42, Mar. 2006.

[109] V. Piriou, P. Chiari, F. Lhuillier, O. Bastien, J. Loufoua, O. Raisky, J. S. David, M. Ovize, and J. J. Lehot, “Pharmacological preconditioning: comparison of desflurane, sevoflurane, isoflurane and halothane in rabbit myocardium,” British Journal of Anaesthesia, vol. 89, no. 3, pp. 486–491, Sep. 2002.

[110] D. F. Stowe and L. G. Kevin, “Cardiac preconditioning by volatile anesthetic agents: a defining role for altered mitochondrial bioenergetics.,” Antioxid. Redox Signal., vol. 6, no. 2, pp. 439–448, Apr. 2004.

[111] D. K. Cope, W. K. Impastato, M. V. Cohen, and J. M. Downey, “Volatile anesthetics protect the ischemic rabbit myocardium from infarction.,” Anesthesiology, vol. 86, no. 3, pp. 699–709, Mar. 1997.

[112] A. Wood, J. Campagna, and K. Miller, “Mechanisms of Actions of Inhaled Anesthetics — NEJM,” New England Journal …, 2003.

[113] J. G. Bovill, “Inhalation anaesthesia: from diethyl ether to xenon.,” Handb Exp Pharmacol, no. 182, pp. 121–142, 2008.

[114] D. M. Wheeler, A. Katz, R. T. Rice, and R. G. Hansford, “Volatile anesthetic effects on sarcoplasmic reticulum Ca content and sarcolemmal Ca flux in isolated rat cardiac cell suspensions.,” Anesthesiology, vol. 80, no. 2, pp. 372–382, Feb. 1994.

[115] R. Kato and P. Foëx, “Myocardial protection by anesthetic agents against ischemia-reperfusion injury: An update for anesthesiologists,” Can J Anesth/J Can Anesth, vol. 49, no. 8, pp. 777–791, Oct. 2002.

[116] G. Torri, “Inhalation anesthetics: a review.,” Minerva anestesiologica, vol. 76, no. 3, p. 215, 2010.

[117] D. C. Saha, A. C. Saha, G. Malik, M. E. Astiz, and E. C. Rackow, “Comparison of cardiovascular effects of tiletamine-zolazepam, pentobarbital, and ketamine-xylazine in male rats.,” J. Am. Assoc. Lab. Anim. Sci., vol. 46, no. 2, pp. 74–80, Mar. 2007.

[118] X. Jiang, L. Gao, Y. Zhang, G. Wang, Y. Liu, C. Yan, and H. Sun, “A comparison of the effects of ketamine, chloral hydrate and pentobarbital sodium anesthesia on isolated rat hearts and cardiomyocytes.,” J Cardiovasc Med (Hagerstown), vol. 12, no. 10, pp. 732–735, Oct. 2011.

[119] S. G. De Hert, F. Turani, S. Mathur, and D. F. Stowe, “Cardioprotection with volatile anesthetics: mechanisms and clinical implications.,” Anesth. Analg., vol. 100, no. 6, pp. 1584–1593, Jun. 2005.

103

[120] Z.-Y. Hu and J. Liu, “Mechanism of cardiac preconditioning with volatile anaesthetics.,” Anaesth Intensive Care, vol. 37, no. 4, pp. 532–538, Jul. 2009.

[121] M. Zaugg, E. Lucchinetti, D. R. Spahn, T. Pasch, and M. C. Schaub, “Volatile anesthetics mimic cardiac preconditioning by priming the activation of mitochondrial KATP channels via multiple signaling pathways,” Anesthesiology, vol. 97, no. 1, p. 4, 2002.

[122] C. H. Chen, J. H. Chuang, K. Liu, and J. Y. H. Chan, “Nitric oxide triggers delayed anesthetic preconditioning-induced cardiac protection via activation of nuclear factor-kappaB and upregulation of inducible nitric oxide synthase.,” Shock, vol. 30, no. 3, pp. 241–249, Sep. 2008.

[123] C. Zhong, Y. Zhou, and H. Liu, “Nuclear factor kappaB and anesthetic preconditioning during myocardial ischemia-reperfusion.,” Anesthesiology, vol. 100, no. 3, pp. 540–546, Mar. 2004.

[124] P. Sergeev, R. da Silva, and E. Lucchinetti, “Trigger-dependent Gene Expression Profiles in Cardiac Precon... : Anesthesiology,” …, 2004.

[125] J. Marinovic, Z. J. Bosnjak, and A. Stadnicka, “Distinct roles for sarcolemmal and mitochondrial adenosine triphosphate-sensitive potassium channels in isoflurane-induced protection against oxidative stress.,” Anesthesiology, vol. 105, no. 1, pp. 98–104, Jul. 2006.

[126] W. G. Toller, E. R. Gross, J. R. Kersten, P. S. Pagel, G. J. Gross, and D. C. Warltier, “Sarcolemmal and Mitochondrial Adenosine Triphosphate– dependent Potassium Channels: Mechanism of Desflurane-induced Cardioprotection,” Anesthesiology, vol. 92, no. 6, p. 1731, Jun. 2000.

[127] J. Müllenheim, D. Ebel, J. Frässdorf, B. Preckel, V. Thämer, and W. Schlack, “Isoflurane preconditions myocardium against infarction via release of free radicals.,” Anesthesiology, vol. 96, no. 4, pp. 934–940, Apr. 2002.

[128] H.-P. Yu, “Role of Anesthetic Agents on Cardiac and Immune Systems,” Shock, vol. 36, no. 6, pp. 532–541, Dec. 2011.

[129] G. Hu, M. R. Salem, and G. J. Crystal, “Isoflurane and Sevoflurane Precondition against Neutrophil-induced Contractile Dysfunction in Isolated Rat Hearts,” Anesthesiology, vol. 100, no. 3, p. 489, Mar. 2004.

[130] B. Heindl, F. M. Reichle, S. Zahler, P. F. Conzen, and B. F. Becker, “Sevoflurane and isoflurane protect the reperfused guinea pig heart by reducing postischemic adhesion of polymorphonuclear neutrophils.,” Anesthesiology, vol. 91, no. 2, pp. 521–530, Aug. 1999.

[131] G. Hu, T. Vasiliauskas, M. R. Salem, D. P. Rhone, and G. J. Crystal, “Neutrophils Pretreated with Volatile Anesthetics Lose Ability to Cause Cardiac Dysfunction,” Anesthesiology, vol. 98, no. 3, p. 712, Mar. 2003.

[132] G. Hu, M. R. Salem, and G. J. Crystal, “Role of Adenosine Receptors in Volatile Anesthetic Preconditioning against Neutrophil-induced Contractile Dysfunction in Isolated Rat Hearts,” Anesthesiology, vol. 103, no. 2, p. 287, Aug. 2005.

[133] J. R. Kersten, T. J. Schmeling, D. A. Hettrick, P. S. Pagel, G. J. Gross, and D. C. Warltier, “Mechanism of Myocardial Protection by Isoflurane: Role of Adenosine Triphosphate‐regulated Potassium (KATP) Channels,” Anesthesiology, vol. 85, no. 4, p. 794, Oct. 1996.

[134] M. Uecker, R. da Silva, T. Grampp, T. Pasch, M. C. Schaub, and M. Zaugg, “Translocation of protein kinase C isoforms to subcellular targets in ischemic and anesthetic preconditioning.,” Anesthesiology, vol. 99, no. 1, pp. 138–147, Jul. 2003.

104

[135] L. G. Kevin and E. Novalija, “Anesthetic preconditioning decreases arrhythmias and improves regional conduction in isolated hearts.,” J. Cardiothorac. Vasc. Anesth., vol. 22, no. 2, pp. 217–224, Apr. 2008.

[136] J. Raphael, S. Abedat, J. Rivo, K. Meir, R. Beeri, T. Pugatsch, Z. Zuo, and Y. Gozal, “Volatile anesthetic preconditioning attenuates myocardial apoptosis in rabbits after regional ischemia and reperfusion via Akt signaling and modulation of Bcl-2 family proteins.,” J. Pharmacol. Exp. Ther., vol. 318, no. 1, pp. 186–194, Jul. 2006.

[137] K. Tanaka, L. M. Ludwig, J. G. Krolikowski, D. Alcindor, P. F. Pratt, J. R. Kersten, P. S. Pagel, and D. C. Warltier, “Isoflurane produces delayed preconditioning against myocardial ischemia and reperfusion injury: role of cyclooxygenase-2.,” Anesthesiology, vol. 100, no. 3, pp. 525–531, Mar. 2004.

[138] M. Tonkovic-Capin and G. Gross, “Delayed cardioprotection by isoflurane: role of KATP channels,” American Journal of …, 2002.

[139] M. Wakeno-Takahashi and H. Otani, “Isoflurane induces second window of preconditioning through upregulation of inducible nitric oxide synthase in rat heart,” American Journal of …, 2005.

[140] Y. M. Tsutsumi, H. H. Patel, D. Huang, and D. M. Roth, “Role of 12-lipoxygenase in volatile anesthetic-induced delayed preconditioning in mice.,” Am. J. Physiol. Heart Circ. Physiol., vol. 291, no. 2, pp. H979–83, Aug. 2006.

[141] J. F. Coetzee, P. J. le Roux, S. Genade, and A. Lochner, “Reduction of postischemic contractile dysfunction of the isolated rat heart by sevoflurane: comparison with halothane.,” Anesth. Analg., vol. 90, no. 5, pp. 1089–1097, May 2000.

[142] W. Schlack, M. Hollmann, J. Stunneck, and V. Thämer, “Effect of halothane on myocardial reoxygenation injury in the isolated rat heart.,” British Journal of Anaesthesia, vol. 76, no. 6, pp. 860–867, Jun. 1996.

[143] M. Naylor, K. K. Bowen, K. A. Sailor, R. J. Dempsey, and R. Vemuganti, “Preconditioning-induced ischemic tolerance stimulates growth factor expression and neurogenesis in adult rat hippocampus.,” Neurochem. Int., vol. 47, no. 8, pp. 565–572, Dec. 2005.

[144] K. R. Dave, I. Saul, R. Busto, M. D. Ginsberg, T. J. Sick, and M. A. P rez-Pinz n, “Ischemic Preconditioning Preserves Mitochondrial Function After Global Cerebral Ischemia in Rat Hippocampus,” J. Cereb. Blood Flow Metab., vol. 21, no. 12, pp. 1401–1410, Dec. 2001.

[145] E. Novalija, S. Fujita, J. P. Kampine, and D. F. Stowe, “Sevoflurane mimics ischemic preconditioning effects on coronary flow and nitric oxide release in isolated hearts.,” Anesthesiology, vol. 91, no. 3, pp. 701–712, Sep. 1999.

[146] V. Piriou, P. Chiari, O. Gateau-Roesch, L. Argaud, D. Muntean, D. Salles, J. Loufouat, P.-Y. Gueugniaud, J.-J. Lehot, and M. Ovize, “Desflurane-induced Preconditioning Alters Calcium-induced Mitochondrial Permeability Transition,” Anesthesiology, vol. 100, no. 3, p. 581, Mar. 2004.

[147] X. Lu, H. Liu, L. Wang, and S. Schaefer, “Activation of NF-kappaB is a critical element in the antiapoptotic effect of anesthetic preconditioning.,” Am. J. Physiol. Heart Circ. Physiol., vol. 296, no. 5, pp. H1296–304, May 2009.

[148] R. R. Lamberts, G. Onderwater, N. Hamdani, M. J. A. Vreden, J. Steenhuisen, E. C. Eringa, S. A. Loer, G. J. M. Stienen, and R. A. Bouwman, “Reactive oxygen species-induced stimulation of 5'AMP-activated protein kinase mediates sevoflurane-induced cardioprotection.,” Circulation, vol. 120, no. 11, pp. S10–5, Sep. 2009.

105

[149] P. C. Chiari, P. S. Pagel, K. Tanaka, J. G. Krolikowski, L. M. Ludwig, R. A. Trillo, N. Puri, J. R. Kersten, and D. C. Warltier, “Intravenous emulsified halogenated anesthetics produce acute and delayed preconditioning against myocardial infarction in rabbits.,” Anesthesiology, vol. 101, no. 5, pp. 1160–1166, Nov. 2004.

[150] C. Lotz, M. Lange, A. Redel, J. Stumpner, J. Schmidt, T. Tischer-Zeitz, N. Roewer, and F. Kehl, “Peroxisome-proliferator-activated receptor γ mediates the second window of anaesthetic-induced preconditioning.,” Exp. Physiol., vol. 96, no. 3, pp. 317–324, Mar. 2011.

[151] R. G. Merin, “Effect of anesthetic drugs on myocardial performance in man.,” Annu. Rev. Med., vol. 28, pp. 75–83, 1977.

[152] T. Koyama and K. Fukuda, “Involvement of the kappa-opioid receptor in nitrous oxide-induced analgesia in mice.,” J Anesth, vol. 24, no. 2, pp. 297–299, Apr. 2010.

[153] D. E. Emmanouil, A. S. Dickens, R. W. Heckert, Y. Ohgami, E. Chung, S. Han, and R. M. Quock, “Nitrous oxide-antinociception is mediated by opioid receptors and nitric oxide in the periaqueductal gray region of the midbrain.,” Eur Neuropsychopharmacol, vol. 18, no. 3, pp. 194–199, Mar. 2008.

[154] N. C. Weber, O. Toma, S. Awan, J. Frässdorf, B. Preckel, and W. Schlack, “Effects of nitrous oxide on the rat heart in vivo: another inhalational anesthetic that preconditions the heart?,” Anesthesiology, vol. 103, no. 6, pp. 1174–1182, Dec. 2005.

[155] C. Jin, S. Sonoda, L. Fan, M. Watanabe, T. Kugimiya, and T. Okada, “Sevoflurane and nitrous oxide exert cardioprotective effects against hypoxia-reoxygenation injury in the isolated rat heart.,” J Physiol Sci, vol. 59, no. 2, pp. 123–129, Mar. 2009.

[156] S.-H. Ko, S.-K. Lee, Y.-J. Han, H. Choe, Y.-G. Kwak, S.-W. Chae, K.-P. Cho, and H.-S. Song, “Blockade of Myocardial ATP‐sensitive Potassium Channels by Ketamine,” Anesthesiology, vol. 87, no. 1, p. 68, Jul. 1997.

[157] J. Han, N. Kim, H. Joo, and E. Kim, “Ketamine abolishes ischemic preconditioning through inhibition of K(ATP) channels in rabbit hearts.,” Am. J. Physiol. Heart Circ. Physiol., vol. 283, no. 1, pp. H13–21, Jul. 2002.

[158] A. Molojavyi, B. Preckel, T. Comfère, J. Müllenheim, V. Thämer, and W. Schlack, “Effects of ketamine and its isomers on ischemic preconditioning in the isolated rat heart.,” Anesthesiology, vol. 94, no. 4, pp. 623–9; discussion 5A–6A, Apr. 2001.

[159] J. Müllenheim, J. Frädorf, B. Preckel, and V. Thämer, “Ketamine, but Not S (+)-ketamine, Blocks Ischemic Preconditi... : Anesthesiology,” …, 2001.

[160] J.-L. Hanouz, L. Zhu, E. Persehaye, M. Massetti, G. Babatasi, A. Khayat, P. Ducouret, B. Plaud, and J.-L. Gérard, “Ketamine preconditions isolated human right atrial myocardium: roles of adenosine triphosphate-sensitive potassium channels and adrenoceptors.,” Anesthesiology, vol. 102, no. 6, pp. 1190–1196, Jun. 2005.

[161] L. Wang, N. Oka, M. Tropak, J. Callahan, J. Lee, G. Wilson, A. Redington, and C. A. Caldarone, “Remote ischemic preconditioning elaborates a transferable blood-borne effector that protects mitochondrial structure and function and preserves myocardial performance after neonatal cardioplegic arrest,” The Journal of Thoracic and Cardiovascular Surgery, vol. 136, no. 2, pp. 335–342, Aug. 2008.

[162] M. Galagudza, J. Vaage, and G. Valen, “Isoflurane and other commonly used anaesthetics do not protect the isolated buffer perfused mouse heart from ischemia-reperfusion injury.,” Clin. Exp. Pharmacol. Physiol., vol. 33, no. 4, pp. 315–319, Apr. 2006.

[163] R. Bolli, K. Shinmura, X.-L. Tang, E. Kodani, Y.-T. Xuan, Y. Guo, and B. Dawn, “Discovery of a new function of cyclooxygenase (COX)-2: COX-2 is a

106

cardioprotective protein that alleviates ischemia/reperfusion injury and mediates the late phase of preconditioning.,” Cardiovasc. Res., vol. 55, no. 3, pp. 506–519, Aug. 2002.

[164] X. Ren, Y. Wang, and W. K. Jones, “TNF-α is required for late ischemic preconditioning but not for remote preconditioning of trauma,” Journal of Surgical Research, vol. 121, no. 1, pp. 120–129, Sep. 2004.

[165] T. Steensrud, J. Li, X. Dai, C. Manlhiot, R. K. Kharbanda, M. Tropak, and A. Redington, “Pretreatment with the nitric oxide donor SNAP or nerve transection blocks humoral preconditioning by remote limb ischemia or intra-arterial adenosine.,” Am. J. Physiol. Heart Circ. Physiol., vol. 299, no. 5, pp. H1598–603, Nov. 2010.

[166] Y. Guo, W. K. Jones, Y. T. Xuan, X. L. Tang, W. Bao, W. J. Wu, H. Han, V. E. Laubach, P. Ping, Z. Yang, Y. Qiu, and R. Bolli, “The late phase of ischemic preconditioning is abrogated by targeted disruption of the inducible NO synthase gene.,” Proc. Natl. Acad. Sci. U.S.A., vol. 96, no. 20, pp. 11507–11512, Sep. 1999.

[167] Y. T. Xuan, Y. Guo, Y. Zhu, H. Han, R. Langenbach, B. Dawn, and R. Bolli, “Mechanism of cyclooxygenase-2 upregulation in late preconditioning,” Journal of Molecular and Cellular Cardiology, vol. 35, no. 5, pp. 525–537, 2003.

[168] Y.-T. Xuan, Y. Guo, Y. Zhu, O.-L. Wang, G. Rokosh, and R. Bolli, “Endothelial nitric oxide synthase plays an obligatory role in the late phase of ischemic preconditioning by activating the protein kinase C epsilon p44/42 mitogen-activated protein kinase pSer-signal transducers and activators of transcription1/3 pathway.,” Circulation, vol. 116, no. 5, pp. 535–544, Jul. 2007.

[169] S. Loukogeorgakis, R. Williams, and A. Panagiotidou, “Transient Limb Ischemia Induces Remote Preconditioning and Remote Postconditioning in Humans by a KATP Channel–Dependent Mechanism,” Circulation, 2007.

[170] I. A. Rahman, J. G. Mascaro, R. P. Steeds, M. P. Frenneaux, P. Nightingale, P. Gosling, P. Townsend, J. N. Townend, D. Green, and R. S. Bonser, “Remote ischemic preconditioning in human coronary artery bypass surgery: from promise to disappointment?,” Circulation, vol. 122, no. 11, pp. S53–9, Sep. 2010.

[171] M. Shimizu, I. E. Konstantinov, R. K. Kharbanda, M. H. Cheung, and A. N. Redington, “Effects of intermittent lower limb ischaemia on coronary blood flow and coronary resistance in pigs,” Acta Physiol, vol. 190, no. 2, pp. 103–109, Jun. 2007.

[172] K. Enko, K. Nakamura, K. Yunoki, T. Miyoshi, S. Akagi, M. Yoshida, N. Toh, M. Sangawa, N. Nishii, S. Nagase, K. Kohno, H. Morita, K. F. Kusano, and H. Ito, “Intermittent arm ischemia induces vasodilatation of the contralateral upper limb.,” J Physiol Sci, vol. 61, no. 6, pp. 507–513, Nov. 2011.

[173] M. Wei, P. Xin, S. Li, J. Tao, Y. LI, J. Li, M. Liu, J. Li, W. Zhu, and A. N. Redington, “Repeated Remote Ischemic Postconditioning Protects Against Adverse Left Ventricular Remodeling and Improves Survival in a Rat Model of Myocardial InfarctionNovelty and Significance,” Circ. Res., vol. 108, no. 10, pp. 1220–1225, 2011.

[174] E. Jean St Michel, C. Manlhiot, J. Li, M. Tropak, M. M. MICHELSEN, M. R. SCHMIDT, B. W. MCCRINDLE, G. D. WELLS, and A. N. Redington, “Remote preconditioning improves maximal performance in highly trained athletes.,” Med Sci Sports Exerc, vol. 43, no. 7, pp. 1280–1286, Jul. 2011.

[175] M. E. Reichelt, L. Willems, B. A. Hack, J. N. Peart, and J. P. Headrick, “Cardiac and coronary function in the Langendorff-perfused mouse heart model.,” Exp. Physiol., vol. 94, no. 1, pp. 54–70, Jan. 2009.

107

[176] F. J. Sutherland and D. J. Hearse, “The isolated blood and perfusion fluid perfused heart.,” Pharmacol. Res., vol. 41, no. 6, pp. 613–627, Jun. 2000.

[177] S. Dhein, “The Langendorff Heart,” Practical methods in cardiovascular research, pp. 155–172, 2005.

[178] J. P. Headrick, J. Peart, B. Hack, A. Flood, and G. P. Matherne, “Functional properties and responses to ischaemia-reperfusion in Langendorff perfused mouse heart.,” Exp. Physiol., vol. 86, no. 6, pp. 703–716, Nov. 2001.

[179] F. Labruto, J. Vaage, G. Li, and G. Valen, “Intraperitoneal injection induces a delayed preconditioning-like effect in mice.,” Lab. Anim., vol. 39, no. 3, pp. 298–307, Jul. 2005.

[180] M. C. Fishbein, S. Meerbaum, J. Rit, U. Lando, K. Kanmatsuse, J. C. Mercier, E. Corday, and W. Ganz, “Early phase acute myocardial infarct size quantification: validation of the triphenyl tetrazolium chloride tissue enzyme staining technique.,” Am. Heart J., vol. 101, no. 5, pp. 593–600, May 1981.

[181] N. Varma, F. R. Eberli, and C. S. Apstein, “Increased diastolic chamber stiffness during demand ischemia: response to quick length change differentiates rigor-activated from calcium-activated tension.,” Circulation, vol. 101, no. 18, pp. 2185–2192, May 2000.

[182] N. Varma, F. R. Eberli, and C. S. Apstein, “Left ventricular diastolic dysfunction during demand ischemia: rigor underlies increased stiffness without calcium-mediated tension. Amelioration by glycolytic substrate.,” Journal of the American College of Cardiology, vol. 37, no. 8, pp. 2144–2153, Jun. 2001.

[183] K. Ishikawa, E. R. Chemaly, L. Tilemann, K. Fish, D. Ladage, J. Aguero, T. Vahl, C. Santos-Gallego, Y. Kawase, and R. J. Hajjar, “Assessing left ventricular systolic dysfunction after myocardial infarction: are ejection fraction and dP/dtmax complementary or redundant?,” Am. J. Physiol. Heart Circ. Physiol., vol. 302, no. 7, pp. H1423–8, Apr. 2012.

[184] L. A. Ahmed, H. A. Salem, A. S. Attia, and A. M. Agha, “Comparative study of the cardioprotective effects of local and remote preconditioning in ischemia/reperfusion injury.,” Life Sci., vol. 90, no. 7, pp. 249–256, Feb. 2012.

[185] R. da Silva, T. Grampp, T. Pasch, M. C. Schaub, and M. Zaugg, “Differential Activation of Mitogen-activated Protein Kinases in Ischemic and Anesthetic Preconditioning,” Anesthesiology, vol. 100, no. 1, p. 59, Jan. 2004.

[186] M. S. McKinney and J. Fee, “Cardiovascular effects of 50% nitrous oxide in older adult patients anaesthetized with isoflurane or halothane,” British Journal of Anaesthesia, vol. 80, no. 2, pp. 169–173, Feb. 1998.

[187] D. P. Jenkins, W. B. Pugsley, and D. M. Yellon, “Ischaemic preconditioning in a model of global ischaemia: infarct size limitation, but no reduction of stunning.,” Journal of Molecular and Cellular Cardiology, vol. 27, no. 8, pp. 1623–1632, Aug. 1995.

[188] L. Xi, M. L. Hess, and R. C. Kukreja, “Ischemic preconditioning in isolated perfused mouse heart: reduction in infarct size without improvement of post-ischemic ventricular function.,” Mol. Cell. Biochem., vol. 186, no. 1, pp. 69–77, Sep. 1998.

[189] D. M. Bers, “Cardiac excitation–contraction coupling,” Nature, vol. 415, no. 6868, pp. 198–205, 2002.

[190] D. H. MacLennan and E. G. Kranias, “Phospholamban: a crucial regulator of cardiac contractility.,” Nat. Rev. Mol. Cell Biol., vol. 4, no. 7, pp. 566–577, Jul. 2003.

108

[191] D. K. Rorie, G. M. Tyce, and J. C. Sill, “Increased norepinephrine release from dog pulmonary artery caused by nitrous oxide.,” Anesth. Analg., vol. 65, no. 6, pp. 560–564, Jun. 1986.

[192] T. J. Ebert and J. P. Kampine, “Nitrous oxide augments sympathetic outflow: direct evidence from human peroneal nerve recordings.,” Anesth. Analg., vol. 69, no. 4, pp. 444–449, Oct. 1989.

[193] Y. Zhang, L. P. Quock, E. Chung, Y. Ohgami, and R. M. Quock, “Involvement of a NO-cyclic GMP-PKG signaling pathway in nitrous oxide-induced antinociception in mice.,” Eur. J. Pharmacol., vol. 654, no. 3, pp. 249–253, Mar. 2011.

[194] A. Gorbe, Z. Giricz, A. Szunyog, T. Csont, D. S. Burley, G. F. Baxter, and P. Ferdinandy, “Role of cGMP-PKG signaling in the protection of neonatal rat cardiac myocytes subjected to simulated ischemia/reoxygenation.,” Basic Res. Cardiol., vol. 105, no. 5, pp. 643–650, Sep. 2010.

[195] D. J. Hausenloy, M. M. Mocanu, and D. M. Yellon, “Cross-talk between the survival kinases during early reperfusion: its contribution to ischemic preconditioning.,” Cardiovasc. Res., vol. 63, no. 2, pp. 305–312, Aug. 2004.

[196] R. S. Crawford, F. F. Hashmi, J. E. Jones, H. Albadawi, M. McCormack, K. Eberlin, F. Entabi, M. D. Atkins, M. F. Conrad, W. G. Austen, and M. T. Watkins, “A novel model of acute murine hindlimb ischemia.,” Am. J. Physiol. Heart Circ. Physiol., vol. 292, no. 2, pp. H830–7, Feb. 2007.

[197] J. Hoff, “Methods of Blood Collection in the Mouse,” Lab Animal, vol. 29, no. 10, pp. 47–53, 2000.

[198] N. Tanaka, N. Dalton, L. Mao, H. A. Rockman, K. L. Peterson, K. R. Gottshall, J. J. Hunter, K. R. Chien, and J. Ross, “Transthoracic Echocardiography in Models of Cardiac Disease in the Mouse,” vol. 94, no. 5, pp. 1109–1117, Sep. 1996.

[199] C. R. Frasier, R. L. Moore, and D. A. Brown, “Exercise-induced cardiac preconditioning: how exercise protects your achy-breaky heart.,” J. Appl. Physiol., vol. 111, no. 3, pp. 905–915, Sep. 2011.

[200] S. Golbidi and I. Laher, “Molecular mechanisms in exercise-induced cardioprotection.,” Cardiol Res Pract, vol. 2011, p. 972807, 2011.

109

Chapter 9 Appendices

Appendix 1. Power-lab Acquisition Software: Cardiac Output Chart

110

Appendix 2. Protein extraction-lysis-buffer Final Concentrations: Tris. HCL – 20 mM (pH 7.5)

NaCL – 150 mM

EDTA – 1 mM

EGTA – 1mM

NP-40 – 1%

Na4O7P2 – 2.5 mM

β - Glycerolphosphate – 1 mM

Na3VO4 – 1mM

ddH2O

1 Tablet of Proteinase Inhibitor Cocktail for every 10 mL (Roche Inc.)