by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El...

108
METOPROLOL IMPAIRS MESENTERIC AND POSTERIOR CEREBRAL ARTERY FUNCTION IN MICE by Mostafa Hossam El Beheiry A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Physiology University of Toronto © Copyright by Mostafa Hossam El Beheiry 2010

Transcript of by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El...

Page 1: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

METOPROLOL IMPAIRS MESENTERIC AND POSTERIOR CEREBRAL ARTERY FUNCTION IN

MICE

by

Mostafa Hossam El Beheiry

A thesis submitted in conformity

with the requirements for the degree of Master of Science

Graduate Department of Physiology

University of Toronto

© Copyright by Mostafa Hossam El Beheiry 2010

Page 2: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

ii

1 Abstract

METOPROLOL IMPAIRS MESENTERIC AND POSTERIOR CEREBRAL ARTERY FUNCTION IN

MICE Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale: In addition to their established cardioprotective role, β-adrenergic

antagonists also increase the risk of stroke and mortality. We propose that a vascular mechanism

could contribute to cerebral tissue ischemia in β-blocked patients.

Methods: Cardiac output (CO), mean arterial pressure (MAP) and microvascular brain oxygen

tension (PBrmvO2) were measured in anesthesized mice treated with metoprolol (3mg·kg-1, i.v.).

Dose-response curves (DRCs) for adrenergic-agonists were generated in mesenteric resistance

arteries (MRAs; isoproterenol, clenbuterol) and posterior cerebral arteries (PCAs;

phenylephrine, isoproterenol) before and after metoprolol treatment.

Results: Metoprolol reduced CO, maintained MAP and increased systemic vascular resistance

(SVR) resulting in a decreased PBrmvO2 in mice. Metoprolol attenuated β-adrenergic mediated

vasodilation in both MRAs and PCAs.

Conclusions: Metoprolol reduced brain perfusion in mice. A decrease in CO contributed

however, metoprolol also inhibited β-adrenergic vasodilation of mesenteric and cerebral arteries.

This provides evidence in support of a vascular mechanism for cerebral ischemia in β-blocked

patients.

Page 3: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

iii

2 Acknowledgements

First and foremost I must sincerely thank Drs. Gregory Hare and Steffen-Sebastian Bolz

for their invaluable guidance and support. They have helped me to understand and appreciate

the joys and the sacrifices involved in the scientific process. I am eternally thankful for being

given the opportunity to work with them.

Thanks as well to Dr. Scott Heximer for giving his time to be a part of my advisory

committee. His input was helped to steer this project in the right direction.

Additional thanks must be given to Sharon Klimosco at Department of Anesthesia at St.

Michael’s Hospital as well as the department itself for supporting my salary and funding my

travel and attendance at several scientific conferences. Thanks again to Dr. Bolz for providing

operating funds.

I would like to thank Drs. Jenny Zhang for her surgical expertise in collecting mean

arterial blood pressure data. Thanks as well to Drs. Golam Kabir and Kim Connelly for their

help in collecting and analyzing the left ventricular function of my β-blocked mice.

Special thanks to the people in the Bolz and Hare labs who made the last two years an

amazing and memorable experience.

Finally, thanks to my family and friends for their support outside of the laboratory.

Without these people I would have surely lost my sanity in the weeks where the science gods

were not on my side.

Page 4: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

iv

3 Table of contents

1  ABSTRACT ..................................................................................................................................................... II 

2  ACKNOWLEDGEMENTS .......................................................................................................................... III 

3  TABLE OF CONTENTS .............................................................................................................................. IV 

4  LIST OF TABLES ........................................................................................................................................ VII 

5  LIST OF FIGURES .................................................................................................................................... VIII 

6  OVERVIEW ..................................................................................................................................................... 1 

7  INTRODUCTION ............................................................................................................................................ 3 

7.1  CLINICAL EFFICACY OF BETA-BLOCKERS ........................................................................................................ 3 

7.1.1  Beta-blockers alleviate angina symptoms and reduce morbidity and mortality ................................... 5 

7.1.2  Beta-blockers reduce reinfarction and mortality post-myocardial infarction ...................................... 6 

7.1.3  Beta-blockers reduce morbidity and mortality in chronic heart failure ............................................... 7 

7.1.4  Beta-blockers reduce blood pressure and risk of stroke in hypertensive patients ................................ 9 

7.1.5  Beta-blockers reduce the risk of MI in perioperative patients at cardiac risk ................................... 11 

7.2  INCREASED RISK OF MORTALITY AND STROKE ASSOCIATED WITH BETA-BLOCKADE ..................................... 12 

7.3  BETA-ADRENERGIC SIGNALLING IN REGULATING THE CARDIOVASCULAR SYSTEM ...................................... 14 

7.3.1  Cardiac beta-adrenergic signal transduction pathways .................................................................... 14 

7.3.1.1  Beta-adrenergic modulation of chronotropy .................................................................................................. 15 

7.3.1.2  Beta-adrenergic modulation of contractility ................................................................................................... 18 

7.3.2  Vascular beta-adrenergic signal transduction pathways ................................................................... 20 

7.3.2.1  Distribution across vascular beds ................................................................................................................... 24 

7.4  POTENTIAL OF A VASCULAR MECHANISM IN BETA-BLOCKER PATHOLOGY .................................................... 26 

7.4.1  Vasodilators have better risk reductions for stroke and mortality than beta-blockers ....................... 26 

7.4.2  Cardioselective beta-blockers may act on beta2-adrenergic receptors .............................................. 28 

7.4.3  Metoprolol impairs cerebral oxygen delivery in acutely anemic rats ................................................ 29 

8  HYPOTHESIS AND AIMS ........................................................................................................................... 31 

Page 5: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

v

9  METHODS ...................................................................................................................................................... 32 

9.1  EFFECT OF METOPROLOL ON BRAIN O2 TENSION AND HEMODYNAMICS IN VIVO ............................................ 32 

9.1.1  Animals ............................................................................................................................................... 32 

9.1.2  Experimental protocol ........................................................................................................................ 32 

9.1.3  Heart rate and mean arterial blood pressure ..................................................................................... 32 

9.1.4  Microvascular brain oxygen tension .................................................................................................. 34 

9.1.5  Cardiac responsiveness and left ventricular function ........................................................................ 34 

9.2  EFFECT OF METOPROLOL IN THE MESENTERIC RESISTANCE ARTERY IN VITRO ............................................... 35 

9.2.1  Animals ............................................................................................................................................... 35 

9.2.2  Mesenteric resistance artery isolation ............................................................................................... 35 

9.2.3  Pressure myography ........................................................................................................................... 37 

9.2.4  Effect of metoprolol on beta1,2-adrenergic mediated vasodilation ................................................... 39 

9.2.5  Effect of metoprolol on beta2-adrenergic mediated vasodilation ...................................................... 39 

9.3  EFFECT OF METOPROLOL IN THE POSTERIOR CEREBRAL ARTERY IN VITRO .................................................... 40 

9.3.1  Animals ............................................................................................................................................... 40 

9.3.2  Posterior cerebral artery isolation ..................................................................................................... 40 

9.3.3  Pressure myography ........................................................................................................................... 40 

9.3.4  Effect of metoprolol on adrenergic mediated vasomotor function ..................................................... 41 

9.4  DRUGS AND SOLUTIONS ................................................................................................................................ 41 

9.5  STATISTICAL ANALYSIS ................................................................................................................................ 42 

9.5.1  Effect of metoprolol on brain O2 tension and hemodynamics in vivo ................................................. 42 

9.5.2  Effect of metoprolol in mouse arteries ............................................................................................... 42 

10  RESULTS ........................................................................................................................................................ 44 

10.1  EFFECT OF METOPROLOL ON BRAIN O2 TENSION AND HEMODYNAMICS ................................................... 44 

10.1.1  Metoprolol injection reduced brain O2 tension and heart rate ...................................................... 44 

10.1.2  Metoprolol injection reduced CO and increased SVR ................................................................... 45 

10.2  METOPROLOL INHIBITS ISOPROTERENOL MEDIATED VASODILATION MRAS ............................................ 51 

10.3  METOPROLOL INHIBITS ISOPROTERENOL MEDIATED VASODILATION INPCAS .......................................... 58 

Page 6: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

vi

11  DISCUSSION .................................................................................................................................................. 63 

11.1  BETA-ADRENERGIC VASODILATION IS REQUIRED TO MAINTAIN VITAL ORGAN PERFUSION ...................... 64 

11.2  METOPROLOL INHIBITS ADRENERGIC VASODILATION IN MESENTERIC AND CEREBRAL ARTERIES ............ 67 

11.3  METOPROLOL MAY BE ASSOCIATED WITH INCREASED MORBIDITY AND MORTALITY ............................... 67 

11.4  DIFFERENCES BETWEEN MESENTERIC AND CEREBRAL ARTERIES ............................................................. 69 

11.5  LIMITATIONS ............................................................................................................................................ 70 

12  SUMMARY ..................................................................................................................................................... 73 

13  APPENDIX A – BETA-BLOCKADE: A HISTORICAL PERSPECTIVE ............................................... 74 

14  APPENDIX B – CHARACTERIZATION OF PCA PHENYLEPHRINE RESPONSES ........................ 79 

14.1  DETERMINING OPTIMAL PHENYLEPHRINE PRECONSTRICTION DOSE IN PCAS ........................................... 79 

14.1.1  Methods ......................................................................................................................................... 79 

14.1.2  Results ........................................................................................................................................... 80 

14.2  EFFECT OF METOPROLOL ON PHENYLEPHRINE DOSE-RESPONSE CURVE IN PCAS ..................................... 83 

14.2.1  Results ........................................................................................................................................... 83 

14.2.2  Interpretation: Phenylephrine responses are time-dependent in the PCA .................................... 86 

15  REFERENCE LIST ....................................................................................................................................... 88 

Page 7: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

vii

4 List of Tables

TABLE 7.1 PHARMACOLOGICAL PROPERTIES OF CLINICALLY USED BETA-BLOCKERS .................................................... 4 

TABLE 10.1 HALF MAXIMAL CONCENTRATIONS FOR MRA DOSE-RESPONSE CURVES. ................................................ 53 

TABLE 10.2 HALF MAXIMAL CONCENTRATIONS OF ISOPROTERENOL IN PCAS. ........................................................... 62 

TABLE 13.1 ORIGINAL CHARACTERIZATION OF ADRENERGIC RECEPTOR SUBTYPES .................................................... 76 

TABLE 13.2 STRUCTURES OF BETA-ADRENERGIC RECEPTOR LIGANDS ........................................................................ 77 

TABLE 14.1 HALF MAXIMAL CONCENTRATIONS OF PHENYLEPHRINE IN PCAS. ........................................................... 85 

Page 8: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

viii

5 List of Figures

FIGURE 7.3.1 SUMMARY OF CARDIAC BETA-ADRENERGIC SIGNALLING. ..................................................................... 17 

FIGURE 7.3.2 SUMMARY OF VASCULAR BETA-ADRENERGIC SIGNALLING. ................................................................... 21 

FIGURE 9.1.1 EXPERIMENTAL PROTOCOL FOR ASSESSING MAP AND PBRMVO2 WITH METOPROLOL ............................ 33 

FIGURE 9.2.1 PHOTOGRAPHS OF ARTERY ISOLATION AND CANNULATION. .................................................................. 36 

FIGURE 9.2.2 SET UP OF THE PRESSURE MYOGRAPHY APPARATUS .............................................................................. 38 

FIGURE 10.1.1 TIME COURSE OF HYPOXIC CHALLENGE PROTOCOL WITH SALINE/METOPROLOL. ................................. 46 

FIGURE 10.1.2 MEAN HEART RATE, MAP AND PBRMVO2 IN HYPOXIC CHALLENGE WITH SALINE/METOPROLOL. ......... 47 

FIGURE 10.1.3 DELTA HEART RATE, MAP AND PBRMVO2 IN HYPOXIC CHALLENGE WITH SALINE/METOPROLOL. ........ 48 

FIGURE 10.1.4 HEMODYNAMIC CHANGES FOLLOWING SALINE AND SUBSEQUENT METOPROLOL INJECTIONS. ............ 49 

FIGURE 10.1.5 REPRESENTATIVE PRESSURE-VOLUME LOOP BEFORE AND AFTER METOPROLOL TREATMENT. ............. 50 

FIGURE 10.2.1 REPRESENTATIVE TRACING OF MRA EXPERIMENTAL PROTOCOL. ....................................................... 52 

FIGURE 10.2.2 EFFECT OF METOPROLOL ON ISOPROTERENOL DOSE-RESPONSE CURVES IN MESENTERIC ARTERIES. .... 54 

FIGURE 10.2.3 PERCENT DILATION TO EC50 DOSE (30µM) OF ISOPROTERENOL IN MESENTERIC ARTERIES. ................ 55 

FIGURE 10.2.4 PERCENT DILATION AT EMAX OF ISOPROTERENOL DOSE-RESPONSE CURVES. ......................................... 56 

FIGURE 10.2.5 EFFECT OF METOPROLOL ON CLENBUTEROL MEDIATED VASODILATION. ............................................. 57 

FIGURE 10.3.1 REPRESENTATIVE TRACING OF PCA PROTOCOL. ................................................................................. 60 

FIGURE 10.3.2 EFFECT OF METOPROLOL ON ISOPROTERENOL DOSE-RESPONSE IN PCAS. ............................................ 61 

FIGURE 14.1.1 PHENYLEPHRINE DOSE-RESPONSE CURVES IN MESENTERIC AND CEREBRAL ARTERIES. ....................... 81 

FIGURE 14.1.2 EFFECT OF CONSECUTIVE DOSES OF PHENYLEPHRINE IN POSTERIOR CEREBRAL ARTERIES. ................. 82 

FIGURE 14.2.1 EFFECT OF METOPROLOL ON PHENYLEPHRINE DOSE-RESPONSE IN PCAS. ........................................... 84 

Page 9: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

1

6 Overview

The β-blocker class of antihypertensive drugs is routinely used in the treatment of

chronic disease (hypertenstion, heart failure, coronary artery disease). Additionally, it is

prescribed as a prophylactic intervention in patients at cardiovascular risk undergoing surgery.

Their efficacy in reducing cardiac morbidity and mortality was established in conditions of

myocardial ischemia and infarction. Without clear testing, their scope of treatment broadened

and they were used more liberally due to the perception that β-blockers had broad efficacy with

limited toxicity. In 2009 alone there were 130 million prescriptions for β-blockers in the US

placing it in the top 5 most prescribed drug class.

In addition to their ability to promote cardioprotection by reducing myocardial oxygen

demand, β-blockers became a first line therapy to treat high blood pressure. However, despite

their ability to control blood pressure, β-blockers were not as effective as Ca2+ channel blockers

(CCBs) and angiotensin converting enzyme inhibitors (ACE-Is) at reducing stroke and

mortality. In perioperative medicine, initial trials recommended their use as they coincided with

a reduction in adverse cardiac events (eg. nonfatal myocardial infarction or ischemia and cardiac

death). A recent randomized controlled trial in perioperative medicine (POISE) revealed that β-

blockade with the common β1-antagonist, metoprolol, reduced the incidence of myocardial

infarction. However, metoprolol administration was also associated with an increased risk of

stroke and death during the perioperative period. This emphasized that in certain clinical settings

(surgery, acute blood loss) the inhibition of cardiac responsiveness may have the consequence of

decreased vital organ perfusion In support of this assumption, a growing body of evidence has

demonstrated an increase in cerebral ischemia and mortality associated with β-blocker therapy

in a diverse number of clinical settings. Defining the mechanisms contributing to these negative

Page 10: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

2

outcomes is essential in informing clinical practice to develop effective preventative therapeutic

strategies.

The general premise of this in vivo and in vitro experimental study is that a vascular

mechanism could contribute to β-blocker induced cerebral ischemia and mortality. This premise

is based on the following lines of reasoning: 1) Anti-hypertensive therapy with primary

vasodilators (ACE inhibitors, Ca2+ channel blockers) are associated with improved survival and

fewer strokes when compared with β-blockers; 2) β-blockers with vasodilatory capacity

(carvedilol) are associated with a lower stroke incidence and mortality when compared to β-

blockers that do not cause vasodilation (metoprolol); 3) β-blockers with greater vascular β2-

adrenergic receptor cross reactivity (metoprolol) are associated with increased mortality when

compared to more β1-cardioselective β-blockers (atenolol, bisoprolol); 4) There is evidence that

shows that stimulation of both β1- and β2- adrenergic receptor subtypes initiate vasodilatory

mechanisms in vascular smooth muscle. Thus β-blockade may have a negative impact on the

ability of the resistance vasculature to dilate, either through a direct β1-effect or by cross-

reacting with the β2-receptor. We therefore propose the following hypothesis:

Cardioselective β1-adrenergic antagonists impair resistance artery vasodilation and increase

the risk of organ ischemia.

The β-blocker metoprolol was chosen as our study drug because of its extensive clinical

use in North America and its specific pharmacology which may promote increased morbidity

and mortality within this class of drugs.

Page 11: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

3

7 Introduction

7.1 Clinical efficacy of beta-blockers

The β-adrenergic receptor (β-AR) antagonist class of drugs has been growing in

popularity as a cardiovascular intervention over the last half century. With 128.3 million

prescriptions in 2009 they were the 5th most dispensed class of medicine in North America (IMS

Health). Though they are currently used for the treatment of a wide range of cardiovascular

ailments (hypertension, myocardial infarction, chronic heart failure) and various non-

cardiovascular morbidities (glaucoma, migraine headaches, essential tremors, alcohol

withdrawal)1, β-blockers were initially developed as a specific treatment for angina pectoris2.

By inhibiting sympathetic activation of the heart through the β-adrenergic receptor, these drugs

are able to limit myocardial oxygen demand in order to match supply. In this way they protect

the heart from ischemia and their use subsequently expanded to post-MI and chronic heart

failure treatments. In addition to an expansion of their clinical indications, β-blockers also

developed from first generation, non-selective β1/2-antagonists, such as propranolol, to the

second generation β1-, “cardioselective”, antagonists, such as metoprolol, and are now in their

third generation which includes both nonspecific and β1-specific antagonists that have

vasodilating capacity, such as carvedilol and nebivolol respectively (Table 7.1). Because of their

efficacy in treating ischemic heart disease, their use further expanded to the treatment of

hypertension and as a prophylactic against myocardial ischemia and infarction in the

perioperative setting.

Page 12: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

4

Drug Receptor Selectivity*

Current Clinical

Use*

Additional Actions* Fold Selectivity for β1-AR vs. β2-AR**

Nadolol β1/2 -

Penbutolol β1/2 -

Pindolol β1/2 -

Propranolol β1/2 + -

Timolol β1/2 -

Acebutolol β1 + - 2.4

Atenolol β1 ++ - 4.7

Bisoprolol β1 ++ - 13.5

Esmolol β1 + -

Metoprolol β1 +++ - 2.3

Nebivolol β1 Stimulates eNOS

Labetalol β1/2 α1-AR antagonist

Carvedilol β1/2 ++ α1-AR antagonist

Celiprolol β1 Partial β2-AR agonism

Table 7.1 Pharmacological properties of clinically used beta-blockers *Adapted from Pulido and Kor 20083. **Adapted from Baker 20054.

Page 13: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

5

7.1.1 Beta-blockers alleviate angina symptoms and reduce morbidity and mortality

Beta-blockers were initially developed as a targeted treatment for stable angina pectoris

which is characterized by myocardial ischemia due to an increase in oxygen demand without the

consequent increase in supply. Stable angina manifests in patients as severe chest pains during

periods of activity which disappear once activity has ceased. Propranolol was first indicated in

the management of stable angina in 1964, shortly after its discovery by Sir James Black (See

Appendix A). In a small trial (n=20), the authors were encouraged by a trend of increased

number of days without symptoms, reduced number of angina attacks and increased level of

patient satisfaction with β-blockade5. In another small (n=19) double blind crossover trial there

was a significant reduction in the number of angina attacks and in the number of nitrate tablets

(to treat angina) taken during the 8 weeks of study (4 weeks of placebo, 4 weeks propranolol)6.

The 306 patient double-blind placebo controlled Atenolol in Silent Ischemia Study Trial

(ASIST) remains to date the largest randomized controlled trial (RCT) assessing β-blocker

efficacy in angina7. Atenolol treatment resulted in a 56% reduction in adverse events (death, MI,

unstable angina, aggravation of angina, revascularization) during the 1 year long follow up

period7.

Larger trials assessing the efficacy of β-blockade in managing angina have been reserved

for studies of its effectiveness in comparison to other monotherapies and in combination

therapies. Two large trials (TIBET and APSIS) demonstrated that β-blockade alone resulted in

no better outcomes than Ca2+ channel blockade alone or in combination8;9. Without placebo

controls it is difficult to assess the efficacy of β-blockade in the long-term prognosis of angina

in these two trials.

With a lack of large randomized controlled trials, the indication for β-blockade in

patients with angina pectoris has been extrapolated from RCTs of related cardiovascular

Page 14: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

6

morbidities. As will be discussed further, β-blockers have been proven to improve outcomes in

post-MI and heart failure patients 10-12. For this reason they are currently suggested as first line

therapy by the European Society of Cardiology and the American Heart Association in post-MI,

acute coronary syndrome and heart failure patients diagnosed with stable angina pectoris13;14.

7.1.2 Beta-blockers reduce reinfarction and mortality post-myocardial infarction

Early experimental studies found that β-blockade, with propranolol, was able to reduce

myocardial infarct (MI) size by 56 and 28% before and 3 hours after left anterior descending

coronary artery occlusion15. In the clinical setting, β-blocker prophylaxis for acute MI is next to

impossible; trials have therefore assessed the effectiveness of β-blockade soon after MI or

suspected MI. In these trials MI is defined a rise in plasma troponin in addition to at least one of

the following: 1) ischemic symptoms (chest, epigastric, arm wrist or jaw pain or dyspnea); 2)

development of pathologic Q-waves on an ECG; 3) ST segment elevation or depression or T

wave inversion.

The earliest clinical trials revealed that immediate (within 12 hours after the onset of MI

symptoms) and sustained β-blockade until patient discharge was associated with a 50%

reduction in risk of developing definite infarct or reinfarction after 10 days and a 36% reduction

in mortality after 90 days16-18. In the much larger Norwegian β-Blocker Heart Attack Trial

(BHAT), patients with suspected MI were treated orally with placebo (n=1921) or propranolol

(n=1916) within 5 to 21 days post-MI. The BHAT demonstrated that propranolol reduced total

mortality by 28% and total fatal and nonfatal coronary heart disease by 23% over a 3 year

follow up period19;20. Since BHAT did not address the effect of immediate β-blocker

intervention, nor short-term patient outcomes, an even larger RCT, Metoprolol in Acute

Myocardial Infarction (MIAMI), was conducted. Patients eligible for the MIAMI trial

(presenting in hospital within 24 hours of the onset of MI symptoms) were randomized to

Page 15: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

7

placebo (n=2901) and metoprolol (n=2877) treatments. In high risk patients, metoprolol

treatment reduced 15 day post-MI mortality by 29% 21. The first International Study of Infarct

Survival (ISIS-1) was conducted in 16207 patients with 8037 receiving early intravenous

atenolol and 7990 receiving no treatment within 5 hours after the onset of suspected MI for 2

weeks. ISIS-1 found that there was a significant 15% reduction in mortality up to 7 days

following treatment with atenolol versus no treatment22. ISIS-1 also demonstrated that the

benefits of β-blockade are most prominent during the first 24 hours of treatment22 following MI,

prompting the question of whether early β-blockade is beneficial versus delayed β-blockade.

The Thrombolysis in Myocardial Infarction Phase II (TIMI-II) trial found a significant 60%

reduction in the incidence of death and reinfarction after 6 weeks in a subgroup of patients

treated with immediate metoprolol (n=185) versus those in which β-blockade was deferred until

6 days after symptom onset (n=190)23. In the largest RCT of β-blockers to date, the Clopidogrel

and Metoprolol in Myocardial Infarction Trial (COMMIT) showed a 25 and 20% relative risk

reduction of reinfarction and ventricular fibrillation, respectively, following metoprolol (n=22

929) vs. placebo (n=22 923) treatment within 24h of suspected MI24.

A clear benefit has therefore been associated with early β-blockade in post-MI patients.

The American Heart Association has consequently recommended β-blockade as a management

strategy in all patients without contraindications due to the apparent early and long-term

mortality risk reductions in post-MI patients10.

7.1.3 Beta-blockers reduce morbidity and mortality in chronic heart failure

Chronic heart failure (CHF) is generally characterized by ventricular dysfunction leading

to an impairment of cardiac output and consequently a reduced capacity of the heart to supply

blood throughout the systemic circulation. In order to compensate for the reduction in pumping

capacity of the heart, the sympathetic nervous system and renin-angiotension-aldosterone

Page 16: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

8

system are recruited to preserve systemic blood flow25. These initial benefits of sympathetic

activation (ie. positive inotropy) following myocardial insult led to the contraindication of β-

blockers (negative inotropic agent) in CHF. However, in the mid-1990s, a neurohormonal

hypothesis for the progression of CHF proposed that the increased concentration of circulating

catecholamines and angiotensin II eventually leads to long-term deleterious effects on cardiac

tissue and therefore the worsening of CHF26. This theory paved the way for the first-line usage

of angiotensin converting enzyme (ACE) inhibitors in CHF patients and eventually led to the

reconsideration of β-blockade as a therapeutic approach26.

The US Carvedilol Heart Failure Study Group was among the first large RCTs to

examine the effect of β-blockade with carvedilol on mortality in CHF patients. The study

enrolled patients with a left ventricular ejection fraction (LVEF) less than 35%. The overall

relative risk reductions with carvedilol treatment after 12 months was 65% in mortality and 27%

in risk of hospitalization for cardiovascular reasons27. In the Carvedilol Prospective Randomized

Cumulative Survival Study Group (COPERNICUS) trial, the efficacy of carvedilol in CHF was

tested specifically in severe HF patients (LVEF <25%). There was a significant 34% relative

risk reduction in 10 month mortality with carvedilol (n=1156) treatment compared to placebo

(n=1133)28. It has been suggested that carvedilol, a non-selective β-blocker, may be especially

beneficial due to its α1-adrenergic blocking ability and its antioxidant effect which could help to

reduce the loss of cardiomyocytes while the disease progresses. However, substantially larger

trials using β1-selective blockers have shown similar profound survival benefits to CHF patients.

The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II) examined the effect on 12

month survival of the β1-selective antagonist bisoprolol in patients (placebo n=1320; bisoprolol

n=1327) with an LVEF less than 35%. CIBIS-II demonstrated 32, 15 and 25% relative risk

reductions in all cause mortality, all cause hospital admissions and all cardiovascular deaths

Page 17: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

9

respectively29. Similar findings were revealed by the much larger Metoprolol CR/XL

Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Metoprolol (n=1990)

treatment was associated with a 38% reduction in total mortality as well as cardiovascular death

and a 48% reduction in risk of death from worsening heart failure compared to placebo

(n=2001)30. The mechanisms of cardioprotection remain unclear, however an anti-fibrillatory

effect of β1-blockade as well as reduced myocardial oxygen demand and apoptosis may

contribute29;30.

Despite early reservations, β-blockers have emerged as being overwhelmingly successful

in the management of CHF. In fact, in each of the RCTs outlined above, the respective data

monitoring committees were obligated to terminate the studies earlier than anticipated due to the

substantial reduction in mortality compared to placebo treatments27-30. Consequently, β-blockers

have been recommended by both the American College of Cardiologists/American Heart

Association and the European Society of Cardiology as first-line treatment in all CHF patients

without contraindications to the treatment11;12.

7.1.4 Beta-blockers reduce blood pressure and risk of stroke in hypertensive patients

Hypertension, defined as a systolic blood pressure over 140mmHg and a diastolic blood

pressure above 90mmHg, has become one of the more prevalent disease in the western world

and, if uncontrolled, can lead to future cardiovascular complications (stroke, MI, CHF, angina).

Few placebo-controlled trials in hypertension exist due to the ethical dilemma of

withholding anti-hypertensive treatment from hypertensive control patients. One of the largest

of such trials, The International Propsective Primary Prevention Study in Hypertension

(IPPPSH), tested the non-selective β-blocker oxprenolol (n=3185) against placebo (n=3172)

over a 3-5 year follow up period in patients 40-64 years old with diastolic blood pressures above

100mmHg31. Oxprenolol brought about a significant reduction in blood pressure; however it

Page 18: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

10

was no more effective at reducing stroke rates, MI and mortality than placebo treated patients31.

The Medical Research Council in England studied the effectiveness of propranolol (n=2285)

against placebo (n=4525) in managing mild hypertension (diastolic blood pressure between 90-

109mmHg) in 35-64 year olds. Propranolol treatment reduced systolic and diastolic blood

pressures by 10 and 6mmHg respectively after a 5 year follow up period32. They also reported a

27% relative risk reduction in all strokes (fatal and non-fatal) in patients treated with

propranolol32. In a study of elderly patients (60-79 years old; mean systolic and diastolic blood

pressures 196 and 99mmHg respectively), Coope et al. (1986) reported the antihypertensive

benefits of atenolol (n=419) versus placebo (n=469) during a mean 4.4 year follow up period.

Atenolol treatment reduced systolic and diastolic blood pressures by 20 and 11mmHg

respectively33. Additionally, atenolol treatment was associated with a 41% relative risk

reduction in the incidence of fatal and non-fatal strokes but did not confer any significant

reductions in adverse cardiovascular events33. In another study of atenolol as an anti-

hypertensive in the elderly (ages 65-74), the Medical Research Council found a similar

reduction in mean systolic and diastolic pressures (20 and 10 mmHg respectively) after a mean

5.6 year follow up period 34. Like in Coope’s trial, there was a significant reduction in the risk of

stroke (though only by 17%) and no apparent benefits in reducing adverse cardiovascular

events34.

These placebo controlled trials identified β-blockers as effective anti-hypertensives

despite their inability to improve long-term prognosis. Until recently β-blockers were accepted

as a first-line therapy, however they have fallen out of favour due to their inferiority to other

common cardiovascular pharmaceuticals (ACE inhibitors, Ca2+ channel blockers)35;36. The

reasons for their change in status from 1st line to 3rd line antihypertensive agents will be further

explored in section 7.4.1.

Page 19: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

11

7.1.5 Beta-blockers reduce the risk of MI in perioperative patients at cardiac risk

Following the success of β-blockade in various cardiovascular morbidities, the treatment

quickly moved into prophylactic use in surgical patients at cardiac risk. Two small early studies

guided the practice of perioperative β-blockade. In 1996, treatment with atenolol (n=99) was

tested against placebo (n=101) in patients with or at risk of coronary artery disease undergoing

non-cardiac surgery. Atenolol treatment was associated with significantly higher 2-year survival

rates (90 vs. 79%) and 2-year event free survival (83 vs. 68%) than placebo37. In 1999, similar

findings were found in patients with 1 or more cardiac risk factors and a positive dobutamine

stress test treated with bisoprolol (n=59) or placebo (n=53). Bisoprolol significantly reduced

death by cardiac causes (80% relative reduction in risk) and incidence of non-fatal myocardial

infarction (100% relative risk reduction)38. In a 2-year follow-up of these patients, there was a

63% relative risk reduction in cardiac events (cardiac death or non-fatal MI) with bisoprolol

treatment39. Though these were very small trials (n<200 patients), perioperative β-blockade

quickly became a standard clinical practice40. Future studies, however, would call into question

the value of this treatment strategy.

The 2005 POBBLE study tested metoprolol (n=55) versus placebo (n=48) in elderly

patients undergoing vascular surgery. Metoprolol treatment offered no significant survival or

cardioprotective benefits than placebo treatment41. In the metoprolol after vascular surgery

(MaVS) trial, there was no observed benefit in reducing the primary outcome (non-fatal MI,

angina, CHF, atrial or ventricular dysrhythmia or cardiac death)42. In the Diabetic Post-

Operative Mortality and Morbidity (DIPOM) trial, there was again no significant benefit to

metoprolol treatment for reduction in the incidence of mortality, non-fatal MI, angina or CHF

after an 18 month follow up period43. In the Swiss Beta-Blocker in Spinal Anesthesia (BBSA)

trial, patients undergoing non-cardiac surgery with spinal block were treated with bisoprolol

Page 20: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

12

(n=110) or placebo (n=109). Again, there was no observed benefit in reducing cardiac death,

non-fatal MI, angina or CHF in β-blocked patients44. In 2008, the Perioperative Ischemic

Evaluation (POISE) study group published the results of its multicenter, 8000 patient strong

prospective analysis of perioperative β-blockade. The POISE trial is the largest in perioperative

medicine and tested the effect of metoprolol (n=4174) against placebo (n=4177) in patients with

or at risk of atherosclerosis undergoing non-cardiac surgery. They found that metoprolol

treatment resulted in a significant 16% reduction in risk of reaching the primary outcome

(cardiovascular death [death due to cardiac arrest, MI, pulmonary embolus, stroke, hemorrhage

or following any cardiovascular procedure], non-fatal MI or non-fatal cardiac arrest) and that

this risk reduction was driven by a significant 26% reduction in the relative risk of non-fatal

MI45. Reduced cardiovascular morbidity risk was confirmed in a recent RCT demonstrating a

65% reduction in the relative risk of cardiac death and non-fatal MI46. A meta-analysis of

perioperative β-blocker trials further supports the cardioprotective effect of β-blockade47,

however, the POISE findings were controversial because of two alarming findings; increased

risks of all cause mortality and stroke in the metoprolol group45.

7.2 Increased risk of mortality and stroke associated with beta-blockade

Despite the proven cardioprotective benefits of β-blocker treatment in a wide range of

cardiovascular diseases, their use in certain patient populations is debatable. Specifically, in the

perioperative setting, where β-blocker therapy has seen a recent proliferation over the last two

decades, there is growing evidence of increased risk of harm in treated patients. The landmark

2008 POISE trial demonstrated that metoprolol treated patients experienced a 100% increase in

relative risk of developing fatal or non-fatal strokes45. 82% of these strokes were ischemic

strokes in nature while the remaining 18% were hemorrhagic or undefined. Of the ischemic

strokes, the investigators did not define whether they were hypo-perfusion or thrombosis related.

Page 21: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

13

Additionally, the POISE investigators reported a 35% increase in the relative risk of all-cause

mortality (this includes cardiovascular and non-cardiovascular deaths) in metoprolol treated

patients. Development of a stroke resulted in an 18-fold increase in risk of death45. Shortly after

the publication of POISE, a meta-analysis of 33 randomized placebo-controlled perioperative β-

blockade trials revealed an alarming 116% increase in the relative risk of developing a non-fatal

stroke with β-blocker therapy as well as a 12.5% relative risk increase in all-cause mortality47.

The POISE study also reported that increased harm was associated with β-blockade in low

cardiac risk patients (revised cardiac risk scores ≤ 2)45. This finding was previously reported in a

large retrospective analysis of over 300, 000 propensity matched patients undergoing major non-

cardiac surgery. Lindenauer and colleagues reported that in patients with an RCRI score of 0 or

1 there was an increased risk of mortality with β-blocker treatment48. Furthermore, in the MaVS

trial where the large majority of patients were classified with an RCRI of ≤ 2 there was no

observed benefit of perioperative β-blockade in reducing primary outcomes42. Similarly, in

DIPOM and POBBLE there were again no observed benefits to metoprolol treatment41;43.

Increased risk of negative outcomes persists outside of the perioperative setting. In the

CIBIS-II trial in patients with heart failure there was a 91% increased risk of stroke with

bisoprolol treatment compared to placebo29. In the COMMIT trial, despite the benefits in

reducing the risk of reinfarction and ventricular fibrillation, there was a 28% increase in the risk

of cardiogenic shock when post-MI patients were treated with metoprolol24. In hypertension, β-

blockers are associated with 26% increased relative risk of stroke49 when compared to other

commonly used cardiovascular pharmaceuticals (CCBs, ACE-Is)50-55. These findings suggest

that patient selection is crucial in optimizing the benefits and minimizing the harm with β-

blocker therapy. There may also be a large population of patients being subjected to unnecessary

risks through treatment with β-blockers. It is therefore important to elucidate the mechanism by

Page 22: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

14

which β-blockade could promote organ ischemia (ie. stroke) in order to better inform clinical

practice. In a recent meta-analysis and review, the risk of stroke and mortality associated with

prerioperative β blocker therapy was attributed to: 1) Degree of hepatic metabolism, 2) Dose

titration and 3) Relative degree of β1/2-AR selectivity56. Thus, a clearer understanding of the beta

receptor physiology is required.

7.3 Beta-Adrenergic signalling in regulating the cardiovascular system

Lands and colleagues, in 1967, discovered the two subtypes responsible for the

physiologic effects of β-AR activation. Using isolated cardiac, adipose, bronchiole and vascular

tissue, Lands et al. observed different orders of activity to stimulation with isoproterenol,

norepinephrine and epinephrine within these samples57;58. It was discovered that heart rate,

myocardial force of contraction and lipolysis in adipose tissue had a potency order of (most to

least potent) isoproterenol > norepinephrine = epinephrine and consequently designated these

responses as being β1-mediated58. Bronchodilation and vasodilation responses were found to

follow an activity order of isoproterenol > epinephrine >> norepinephrine and consequently

were designated as β2-mediated responses. For the purposes of this report, cardiac and vascular

β-adrenergic signalling will be of particular focus.

7.3.1 Cardiac beta-adrenergic signal transduction pathways

Sympathetic activation of the β-ARs expressed on the heart result in an increase in heart

rate (chronotropy), contractility (inotropy) and relaxation rate (lusitropy). By modulating these

latter two characteristics, β-AR activation results in increased stroke volume. Coupled with an

increase in heart rate, β-adrenergic stimulation increases cardiac output in order to maintain

adequate tissue perfusion in the typical sympathetic fight or flight response59. At the cellular

Page 23: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

15

level, the downstream effectors of β-AR activation must therefore modulate two fundamentally

different physiologic responses; pacemaking and myogenic force production.

The β-ARs are Gs-protein coupled receptors expressed on both atrial and ventricular

cardiomyocytes. The receptors are composed of 7 transmembrane regions with an extracellular

N-terminus and intracellular C-terminus. They are made up of approximately 400-500 amino

acids and the β1- and β2-ARs show 48.9% sequence homology60.The ratio of expression of the

β1-AR to β2-AR subtypes is approximately 80:20% in ventricular myocytes and 70:30% in atrial

myocytes61. When bound by the endogenous catecholamines (norepinephrine and epinephrine),

both β-AR subtypes catalyze the transfer of a phosphate from guanosine triphosphate (GTP) to

the Gαs subunit of the heterotrimeric G-protein complex. This causes the dissociation of the Gαs

subunit from the βɣ subunits. The Gαs subunit then goes on to activate membrane bound

adenylyl cyclase which is responsible for catalyzing the conversion of adenosine triphosphate

(ATP) to cyclic adenosine monophosphate (cAMP)62. The ubiquitous second messenger, cAMP,

binds to protein kinase A (PKA) allowing the dissociation of its catalytic units from its

regulatory units. The catalytic units of PKA go on to phosphorylate various downstream

molecules that consequently regulate chronotropy, inotropy and luistropy59. The signalling

pathways are summarized in Figure 7.3.1.

7.3.1.1 Beta-adrenergic modulation of chronotropy

The heart is unique in its ability to generate spontaneous contractile activity through the

specialized pacemaker myocytes localized within the sinoatrial node (SAN)63. Action potentials

generated within the SAN are propagated through the left and right atria to the atrioventricular

node (AVN) where the electrical signal is transmitted to the left and right ventricles through the

bundle of His and Purkinje fibres. With its high intrinsic rate of action potential generation, the

SAN serves as the primary pacemaker of the heart, although the AVN and Purkinje fibres also

Page 24: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

16

display automaticity, albeit at a slower rate64. It is understood that diastolic depolarization, a

feature distinctly lacking in the force generating cardiomyocytes, is responsible for the

spontaneous activity of pacemaker cells. Specifically, the inward “funny” current (If), activated

during diastole, is responsible for the slow depolarization of a pacemaker cell following the

termination of the previous action potential. As the cell is slowly depolarized by the If it will

eventually reach the threshold potential (-55mV) and subsequently cause the generation of a

new action potential63;64.

Page 25: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

17

AdenylylCyclase

ATPcAMP

Sarcoplasmic Reticulum

PLB

[Ca2+]sr

[Ca2+]i

SERCA

Gαs Gβγ

GTPGDP

Protein Kinase A

β1/2-AR

RyR

[Na+]ec If [Na+]i

[Ca2+]ec

Cav1.2

[Ca2+]ec

[Ca2+]i

Cav1.2

Figure 7.3.1 Summary of cardiac beta-adrenergic signalling. See text for details. β1/2-AR = β1/2-adrenergic receptor; If = funny current channel; [Na+]ec = extracellular Na+; [Na+]i = cytoplasmic Na+; [Ca2+]ec = extracellular Ca2+; [Ca2+]i = cytoplasmic; Ca2+; [Ca2+]sr = sarcoplasmic reticulum Ca2+; Cav1.2 = voltage gated L-Type Ca2+ channel; PLB = phospholamban; SERCA = sarcoplasmic/endoplasmic reticulum calcium ATPase; RyR = ryanodine receptor.

Page 26: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

18

Stimulation of the β-adrenergic receptor in preparations of SAN cells has shown that the

increases in heart rate from sympathetic activation are likely caused by a shortening and

steepening of the slope of diastolic depolarization65. This causes the threshold potential to be

reached faster during diastole and consequently will cause the generation of more action

potentials per minute. Critical to this phenomenon is the modulation of If activity. In early

experiments, it was discovered that application of epinephrine in whole cell and single If

channel patch clamp preparations increases channel open probabilities and therefore If 66. Beta-

adrenergic modulation of If is mediated by direct binding of cAMP to the If channel as opposed

to phosphorylation by PKA. In patch clamp preparations of SAN cells, application of the

catalytic subunit of PKA alone did not activate If ; however channel activation was observed

when cAMP (alone or with PKA) was applied67. Therefore, β-AR stimulation increases heart

rate through direct action of cAMP on If channels by increasing open probability and therefore

activation of If.

7.3.1.2 Beta-adrenergic modulation of contractility

In regulating cardiac inotropy and lusitropy, β-adrenergic signalling primarily acts on L-

type Ca2+ channels (primarily the Cav1.2 channel), ryanodine receptors, phosphlamban and the

troponin complex. These former three molecules, when activated, modulate intracellular Ca2+

concentrations ([Ca2+]i) which in turn modulates the actions of troponin. Increases in [Ca2+]i

cause contraction whereas decreases in [Ca2+]i promote relaxation64.

In cardiomyocytes, the Cav1.2 channel is primarily responsible for conductance of the

contraction inducing calcium current (ICa)68. Treatment of isolated rat ventricular myocytes with

the non-selective β-AR agonist, isoproterenol, has shown 3-fold increases in ICa through

phosphorylation at the serine 1928 residue on the C-terminal end of Cav1.2 channel69.

Incubation of cardiomyocytes with β-AR specific-antagonists revealed that the β1- and β2-ARs

Page 27: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

19

are responsible for 65% and 35% of the phosphorylation at S1928 respectively. Furthermore,

forskolin, an adenylyl cyclase agonist, mimicked the increases in phosphorylation and ICa,

implicating PKA in the phosphorylation and activation of Cav1.269. It is likely that the A kinase

anchoring protein 15 (AKAP15) is responsible for the differential phosphorylation patterns

observed with β1- or β2-AR activation. Via a leucine zipper motif, AKAP15 colocalizes with the

C-terminal end of the Cav1.2 channel, allowing targeted activation of PKA at the plasma

membrane and in close proximity to its intended effector molecule70. It is suggested that β1-AR

activation is responsible for activation of Cav1.2 throughout the cell by a general increase in

cytosolic cAMP whereas β2-AR activation produces increases in local cAMP at specific areas

near the cell surface69. These regions seem to be associated with cardiac t-tubules as well as

ryanodine receptors (RyRs).

The RyRs are integral in cardiac excitation-contraction coupling. Localized on the

membrane of the sarcoplasmic reticulum in cardiomyocytes, they are stimulated by cytoplasmic

Ca2+ to release sarcoplasmic Ca2+ thereby greatly increasing [Ca2+]i in a process known as Ca2+

induced Ca2+ release (CICR). The β-ARs therefore indirectly regulate RyRs through their

activation of L-type Ca2+ channels which are responsible for the initial increase in [Ca2+]i

necessary for CICR64. However, there is emerging evidence for a direct regulatory role of β-

AR/cAMP/PKA on RyR function. Using patch clamp techniques and confocal imaging in

isolated rat ventricular myocytes, it has been discovered that β-AR activation (isoproterenol) is

responsible for an increase in the number of RyRs that simulatenously respond to the Ca2+

transient of a single L-Type Ca2+ channel71. This synchronization of RyR activation is abolished

when cardiomyocytes were pretreated with a cAMP antagonist, implicating the importance of

PKA in this process. Modulation of the synchronization of RyR opening is suggested to be

integral in producing immediate increases in heart pumping power 71.

Page 28: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

20

RyRs cause Ca2+ efflux from intracellular stores; however, the

sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA) is responsible for pumping Ca2+

out of the cytoplasm and back into the sarcoplasmic reticulum. In so doing, the SERCA pump

reduces [Ca2+]i and therefore is important in cardiomyocyte relaxation64. Phosphlamban is a 52

residue sarcoplasmic reticulum membrane protein that is in direct contact with SERCA. In

resting conditions it is dephosphorylated and serves to inhibit the function of SERCA, thereby

reducing the rate at which [Ca2+]i is removed from the cytoplasm. Beta-adrenergic stimulation,

however, causes phosphorylation of phospholamban at its serine-16 residue by PKA72.

Phosphrylated phosphlamban no longer inhibits SERCA, allowing an increased rate of Ca2+

influx to the sarcoplasmic reticulum. The increased rate of [Ca2+]i efflux from the cytoplasm

allows for the increased rate of relaxation necessary for the heart to keep up with sympathetic

increases in chrontropy. Therefore by modulating Ca2+ cycling in cardiomyocytes, the β-ARs

are responsible for increasing cardiac output to meet the increased oxygen demand associated

with sympathetic nervous system activation.

7.3.2 Vascular beta-adrenergic signal transduction pathways

Lands et al. identified the β2-AR receptor as the primary subtype associated with smooth

muscle relaxation and specifically with vasodilation in vascular smooth muscle cells

(VSMCs)58. Though β2-vasodilation has been entrenched since Lands et al.’s findings, there is

evidence suggesting a tissue and species specific functional role of the β1-AR in promoting

vasodilation (see section 7.3.2.1). β-AR activation follows the same signalling pathways

regardless of receptor subtype being stimulated, and, similar to cardiomyocyte signalling, there

is a central role of cAMP/PKA in mediating VSMC relaxation. The vasodilatory β-adrenergic

signalling pathways, compared to the comparison to the vasoconstriction pathway of α1-

adrenergic signalling, are show in Figure 7.3.2.

Page 29: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

21

AdenylylCyclase

ATPcAMPSarcoplasmic Reticulum

MLC20-P(vasoconstriction)

MLC20

(vasodilation)

PLCPIP2

IP3

Calmodulin

PLB

[Ca2+]sr

IP3R

[Ca2+]iSERCA

Gαs Gβγ

GTPGDP

Protein Kinase A

MLCP MLCK

Gαq

β1/2-AR

GTPGDP

Gβγ

α1-AR

LCC [Ca2+]i

[K+]i

KCa

[K+]ec

KATP

[Ca2+]ec

Figure 7.3.2 Summary of vascular beta-adrenergic signalling. See text for details. β1/2-AR = β1/2-adrenergic receptor; α1-AR = α1-adrenergic receptor; [K+]ec = extracellular K+; [K+]i = cytoplasmic K+; [Ca2+]ec = extracellular Ca2+; [Ca2+]i = cytoplasmic; Ca2+; [Ca2+]sr = sarcoplasmic reticulum Ca2+; LCC = voltage gated L-Type Ca2+ channel; KCa = Ca2+ sensitive K+ channel; KATP = ATP-sensitive K+ channel. PLB = phospholamban; SERCA = sarcoplasmic/endoplasmic reticulum calcium ATPase; IP3 = inositol trisphosphate; IP3R = inositol trisphosphate receptor; PLC = phospholipase C; PIP2 = phosphoinositol diphosphate; MLC20 = myosin light chain; MLC20-P = phosphorylated MLC; MLCP = MLC phosphatase; MLCK = MLC kinase.

Page 30: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

22

As in cardiomyocytes, dissociation of the Gαs subunit from Gβɣ following β-AR

receptor activation causes stimulation of L-type Ca2+ channels (LCCs) in vascular smooth

muscle cells. It has been shown that in isolated rabbit vein myocytes, simultaneous PKA and

PKC inhibition completely abolishes isoproterenol induced LCC activation73. Partial inhibition

of the ICa is observed when myocytes are treated with one of the two protein kinase inhibitors.

Furthermore, when either Gβɣ or Gαs is inhibited, the ICa is reduced and is completely abolished

when both subunits are inhibited during isoproterenol treatment73. A PKC inhibitor blocks Gβɣ

increases in ICa and a PKA inhibitor blocks ICa increases by Gαs implicating these two second

messengers in the activation of LCCs.

LCC activation causes an influx of Ca2+ into the cell, and raises the question of how

increased [Ca2+]i by β-AR stimulation can promote vasodilation when increased Ca2+ is required

for vasoconstriction. The answer lies within the downstream effects of Gαs in both PKA-

independent and dependent processes. Gαs has been discovered to potentiate the efflux of K+

through the calcium activated K+ channel (KCa) by both PKA independent and dependent

mechanisms74. This causes membrane hyperpolarization which limits further increases in [Ca2+]i

by closing voltage gated Ca2+ channels75. Additionally, it has been shown that PKA activates the

vascular ATP-sensitive K+ channel (KATP) which could contribute to membrane

hyperpolarization and therefore VSMC relaxation76. In order to further limit [Ca2+]i, PKA,

similarly to cardiomyocytes, will also inhibit the actions of phospholamban on the SERCA

pump in VSMCs. SERCA pumps Ca2+ out of the cytoplasm and into the endoplasmic reticulum,

reducing global [Ca2+]i77. In addition to reducing [Ca2+]i, the increased SR Ca2+ load caused by

phospholamban inhibition causes an increased frequency of Ca2+ sparks which can activate

KCa77. This ultimately leads to increased hyperpolarization, and thus reduced [Ca2+]i. While

Page 31: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

23

PKA has an important role in hyperpolarizing VSMCs and globally reducing [Ca2+]i, it is also

central in reducing the Ca2+ sensitivity of the cell to further promote vasodilation.

In VSMCs, the actual contractile force is generated by ATP dependent cross-bridge

formation of myosin with actin. Myosin slides a short distance along the actin filament before

releasing and completing the cross-bridge cycle78. This cross-bridge cycling is modulated by

myosin light chain (MLC20), a component of the myosin filament that, when phosphorylated at

serine-19, activates the ATPase of myosin allowing cross-bridge formation78. MLC20 is itself

regulated by an MLC kinase and phosphatase (MLCK; MLCP). MLCK is activated by the Ca2+-

calmodulin (Ca2+/CaM) complex, causing phosphorylation of MLC20 and consequently

vasoconstriction. PKA exerts an inhibitory role on MLCK by phosphorylating serine-1760 and

thereby reducing the affinity of MLCK for Ca2+/CaM79;80. Furthermore, PKA has been shown to

phosphorylate serine-695 of the myosin targeting subunit (MYPT1) of MLCP81. This prevents

the Rho-associated kinase phosphorylation of MLCP and its subsequent deactivation82;83. PKA

therefore inhibits MLCK activity while promoting MLCP activity. This shifts the balance within

the VSMC from constriction to vasodilation by increasing the concentration of

dephosphorylated MLC20.

There is evidence that β-adrenergic mediated vasodilation has a nitric oxide component.

Though the direct signalling pathways are yet to be elucidated, it has been demonstrated that

endothelial nitric oxide synthase (eNOS) is upregulated with β-AR receptor stimulation84-86.

Ferror and colleagues propose that activation of eNOS results from PKA activation and by

Akt/P13K. They suggested that the Gβɣ subunit may activate Akt/P13K and consequently cause

NO to be released from the endothelium86.

Page 32: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

24

7.3.2.1 Distribution across vascular beds

Activation of the sympathetic nervous system fight or flight response involves

preferential distribution of blood flow to organs necessary for survival (brain, heart, skeletal

muscle). The endogenous adrenergic catecholamines (epinephrine, norepinephrine) can bind to

both the vasoconstricting α1-adrenergic receptor and the vasodilating β-ARs. The relative

expression and functionality of these receptor subtypes will therefore determine whether an

organ sees reduced or increased perfusion following sympathetic activation87.

The distribution of β-ARs has been well characterized in the cerebral circulation of large

mammals. Bovine anterior, internal carotid and middle cerebral artery strips demonstrate α1-

adrenergic activity (ie. contraction) when stimulated by norepinephrine while posterior cerebral,

posterior communicating and basilar arteries (caudal arteries) are mediated by the β-AR (ie.

relaxation with norepinephrine treatment)88. Furthermore, relaxation in these latter three arteries

can be reversed to contraction when the isolated artery strips are treated with propranolol. Using

metoprolol (β1-specific antagonist) and butoxamine (β2-specific antagonist) this same group

found that each of the caudal arteries were predominantly under the functional control of the β1-

adrenergic receptor89. In humans, radioligand binding assays revealed both β1- and β2-AR

expression in human basilar and middle cerebral arteries in a 40:60% ratio90;91. Interestingly,

pharmacological studies of isolated human pial artery strips show a predominant role of the β1-

AR in mediating vasodilation based on low sensitivity of arteries to the β2-selective agonist

tertbutaline92. In swine basilar artery rings, radioligand binding has demonstrated a 65:35% β1-

/β2-AR expression ratio as well as a functionally dominant role of the β1-AR in the vasodilatory

response of these arteries93. In feline middle cerebral artery rings, there is expression of both β-

AR subtypes with the β1-AR being predominantly functional based on reduced potency to

tertbutaline94. Radioligand binding in rat middle cerebral arteries reveal expression of both β-

Page 33: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

25

ARs91 and vasodilation in isolated rat middle cerebral, posterior cerebral and basilar artery rings

showed equivalent responses to β1- and β2-AR stimulation85. There is no reported functional

data on the primary β-AR involved in the vasodilatory response of mouse cerebral arteries.

Since sympathetic activation increases cardiac responsiveness and therefore myocardial

oxygen demand, there must be a consequent increase in oxygen supply to the tissues of the

heart. The coronary circulation therefore shows β-AR mediated vasodilation to ensure adequate

perfusion and oxygen delivery to the heart during times of increased oxygen demand95;96. In

helical strips of human left anterior coronary artery branches there is a predominant contribution

of the β1-AR subtype in relaxation as determined by metoprolol blockade of isoproterenol

mediated vasodilation as well as a low potency of β2-specific dilation by tertbutaline97. These

researchers also found the same relationship in Japanese monkey and mongrel dog left anterior

coronary artery strips97. These findings further confirmed a previous report that there is a

predominant role of β1-mediated vasodilation in dog coronary arteries98. Similarly, it has been

shown that there is a dominant functional role of the β1-AR in swine anterior descending

coronary artery strips and, through radioligand binding, there is a 68:32% ratio of β1-/β2-AR

expression in these strips99. In rodent models, rings of the rat left anterior coronary atery exhibit

predominantly β1-AR mediated vasodilation100 while isolated and pressurized, mouse

ventricular septal arteries display β-AR mediated vasodilation, though the subtype responsible is

unreported101. Despite the questions remaining in the mouse coronary circulation, it appears that

across species there is predominance for β1-AR mediated vasodilation in coronary arteries.

The evidence is less extensive in skeletal muscle arteries as to which β-AR subtype

promotes vasodilation. Based on potency orders to epinephrine and norepinephrine and on

isoproterenol mediated vasodilation in the presence of practolol (β1-AR antagonist), it was

established that helical segments of rat femoral artery branches are predominantly under β2-

Page 34: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

26

adrenergic receptor control98. Evidence in rodents is unclear; there is evidence for the expression

of both β-AR subtypes in rat gastrocnemius, soleus and vastus lateralis arterioles, though

receptor functionality is unknown102. In mice, using a β1- or β2-AR knockout model, the β1-AR

was discovered to be functionally predominant in femoral artery wire myograph preparations103.

Beta-AR signalling is therefore an important mediator of cardiovascular function. As

evidenced, β-blockers are very useful pharmacological tools for physiologists; however as

discussed, they are very valuable medications for clinical interventions. Their use proliferated

rapidly and only now are they being reconsidered in certain cardiovascular conditions. The

predominance, especially, of β1-AR receptor mediated vasodilation in vital organs suggests that

β-blockers may have important vascular effects that were not previously considered.

7.4 Potential of a vascular mechanism in beta-blocker pathology

Beta-blocker therapy reduces myocardial oxygen demand, a benefit which is invaluable

in diseased hearts where supply is pathologically limited. As described in section 7.3.2.1,

however, the cardioselective (ie. β1-AR specific) designation of β-blockers may be ambiguous

given that various vascular beds display a functional role of β1-AR mediated relaxation. If β-

blockers interfere with the vasodilatory capacity of blood vessels within vital organs (ie. brain)

they could consequently limit tissue perfusion during times of stress when circulating

catecholamines are at their zenith. Several lines of evidence point towards a potential vascular

effect contributing to the pathology of β-blocker administration.

7.4.1 Vasodilators have better risk reductions for stroke and mortality than beta-blockers

Clinical evidence comes from randomized trials assessing the efficacy of

“cardioselective” β-blockers against other cardiovascular drug treatments known to promote

vasodilation. In hypertension, specifically, there have been several trials of β-blockers versus

Page 35: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

27

calcium channel blocker (CCB) or renin-angiotensin system inhibitors (RAS-I) in improving the

long term outcomes of patients. A significantly stronger stroke signal has consistently been

found with β-blocker treatment50-55.

In the European Lacidipine Study on Atherosclerosis (ELSA), patients with systolic

blood pressures between 150 and 210 mmHg were treated and monitored over a 4 year period

with atenolol (n=1157) or the CCB lacidipine (n=1177). Atenolol treatment was associated with

a 33 and 36% increased relative risk of non-fatal stroke or mortality, respectively, compared to

lacidipine52. In the International Verapamil-Trandolapril Study (INVEST), hypertensive patients

were treated with atenolol (n=11309) or verapamil (n=11267) over a 2 year period. Atenolol

treatment was associated with a small 15% increase in relative risk of non-fatal stroke and no

appreciable difference in mortality53. In the Anglo-Scandinavian Cardiac Outcomes Trial

(ASCOT), treatment of atenolol (n=9618) was again tested against a CCB, amlodipine (n=9639)

in hypertensive patients over a mean 5.5 year follow up period. β-blockade was associated with

a significant 33% increase in relative risk of non-fatal stroke with no appreciable difference in

mortality51. Pooling all these results together, a Cochrane meta-analysis reported a significant

24% increase in relative risk of stroke with β-blocker versus CCB treatment in hypertensive

patients49. Similar findings were reported in patients treated with RAS-Is versus β-blockers.

In the Losartan Intervention For Endpoint (LIFE) reduction in hypertension trial, patients

with systolic blood pressures between 160 and 200mmHg were treated with atenolol (n=4588)

or the angiotensin II receptor blocker losartan (n=4605). Atenolol treatment was associated with

a 34% increased relative risk of fatal and non-fatal stroke54. Pooling these results with a smaller

trial (n=758) showing no differences between atenolol and the angiotensin coverting enzyme

inhibitor captopril55, a Cochrane meta-analysis found a significant 30% increase in the risk of

stroke in β-blocker treated patients49. These findings explain why β-blockers are no longer used

Page 36: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

28

as a first-line therapy in the treatment of hypertension36. However, the less favourable side effect

profile of β-blockers against vasodilators is not limited to its use in hypertension.

In the Carvedilol or Metoprolol European Trial (COMET), the 3rd generation,

vasodilating β-blocker, carvedilol (non-selective β-blocker with α1-AR antagonism) was tested

against metoprolol (2nd generation, β1-AR specific) in the treatment of chronic heart failure.

Patients with heart failure were given either carvedilol (n=1511) or metoprolol (n=1518) over a

5 year treatment and follow up period. Patients treated with metoprolol were at a significant 18

and 21% increased risk of all cause mortality and cardiovascular mortality respectively104.

Metoprolol treated patients were also more likely to experience a fatal stroke (190% increased

relative risk)105, a fatal or non-fatal MI (35% increased relative risk)106 and unstable angina

(35% increased relative risk)106. These findings all suggest a superior ability of the vasodilating

β-blocker against β1-specific blockade.

In the perioperative setting, acute β-blockade in preoperative anemic patients is

associated with a significant 116% increase in the relative risk of major adverse cardiac events

(MI, non-fatal cardiac arrest, in-hospital mortality) in a propensity matched retrospective

analysis107. Conversely, in a previous propensity matched retrospective analysis, it was reported

that ACE inhibitors and CCBs are associated with a significant reduction in the risk of

postoperative mortality in preoperative anemic patients 108. The inferiority of β-blockade

compared to vasodilating treatments is therefore of some concern and it may be a significant

mechanism of harm.

7.4.2 Cardioselective beta-blockers may act on beta2-adrenergic receptors

The COMET trial as well as Beattie’s retrospective analysis of preoperative anemic

patients raises an interesting question: are some β-blockers better than others? COMET showed

that metoprolol consistently did worse than carvedilol in reducing adverse events in patients104-

Page 37: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

29

106. In Beattie’s 2009 study, he found that metoprolol is associated with an increased risk of

death in anemic patients compared to treatment with atenolol or bisoprolol 108. A possible reason

for these discrepancies may have to do with the relative selectivity of these β-blockers at their

target receptors. In addition to directly inhibiting β1-adrenergic mediated vasodilation, a β-

blocker that cross-reacts with the β2-AR may compound the inhibitory action on vasodilation

thereby limiting tissue perfusion. Interestingly, bisoprolol and carvedilol, both showing better

outcomes than metoprolol, are 10 times more selective at the β1-AR than the reference β-blocker

propranolol109. Metoprolol shows a comparable potency to propranolol at the β1-AR109. In fact,

using radioligand binding in CHO cells expressing human β1- or β2-ARs, it was found that

metoprolol has the lowest selectivity for the β1-AR of all clinically used β-blockers tested4. This

is an alarming finding when considering that metoprolol is widely used based on its β1-AR

selectivity; it is currently the number one most prescribed β-blocker in North America with over

40 million prescriptions dispensed in 2009 (IMS Health).

7.4.3 Metoprolol impairs cerebral oxygen delivery in acutely anemic rats

Our lab examined the effect of the β-AR antagonist metoprolol in acutely anemic

(hemodiluted) rats. Administration of metoprolol preceding hemodilution resulted in no

significant change in mean arterial pressure (MAP) between β-blocked and control animals,

however, it significantly impaired the ability of rats to increase their cardiac output in response

to the stress associated with hemodilution110. When metoprolol administration preceded

hemodilution, there was a significant reduction in cerebral tissue and microvascular O2

tension110. These findings suggest that when the global blood supply is blunted by β-blockade,

there is reduced perfusion to the brain during anemia. The extent of the reduction in brain O2

tension, however, suggests that there could be a second mechanism contributing to reduced

Page 38: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

30

perfusion. Based on the evidence presented above, this other effect may be related to the

inhibition of the vasodilatory capacity of cerebral blood vessels.

Altogether, these observations suggest that a possible vascular role of β-blockers may

promote tissue ischemia. The effect of metoprolol administration in a mouse model has not been

previously assessed. A vascular mechanism of β-blockade in limiting tissue perfusion could be

indicated by examining the cerebral oxygen tension of β-blocked mice during hypoxic stress.

Acute hypoxia in mice causes a significant drop in mean arterial pressure but not CO suggesting

that there is global vasodilation (reduced systemic vascular resistance) in response to the

reduction in FiO2 (unpublished data). Cerebral tissue oxygen tension also drops in acute

hypoxia. If β-blockade with metoprolol inhibits cerebral vasodilation there may be a greater

drop in cerebral tissue PO2 in hypoxia due to an inability of the animal to adequately maintain

cerebral blood flow. This would provide a good rationale to examine how metoprolol affects

isolated resistance arteries. β-AR function has not been previously assessed in isolated mouse

cerebral arteries, though some studies have characterized their expression in small mesenteric

arteries. In the absence of β-blockade, the endogenous catecholamines will bind to both the

vasoconstricting α-adrenergic receptors and the vasodilating β-adrenergic receptors. If β-

blockers inhibit β-AR mediated vasodilation they could potentially shift the balance towards α-

AR vasoconstriction. This could therefore limit tissue perfusion during conditions of increased

oxygen demand. There is therefore a strong rationale to pursue the effects of the β-blocker

metoprolol on the function of these arteries.

Page 39: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

31

8 Hypothesis and Aims

Cardioselective β1-adrenergic antagonists impair resistance artery vasodilation and increase

the risk of organ ischemia.

Specific Aim #1

To determine the effect of a clinically utilized cardioselective β-blocker (metoprolol) on cardiac

output, systemic vascular resistance and cerebral tissue oxygen tension in mice.

Specific Aim #2

i) To determine the effect of metoprolol on β-adrenergic mediated vasodilation in mouse

mesenteric arteries.

ii) To characterize the response of mouse posterior cerebral arteries to adrenergic stimulation

(phenylephrine, isoproterenol) before and after metoprolol treatment.

Specific Aim #3

To determine the β-adrenergic receptor (β1 or β2) selectivity of metoprolol in mesenteric

arteries.

Page 40: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

32

9 Methods

9.1 Effect of metoprolol on brain O2 tension and hemodynamics in vivo

9.1.1 Animals

Wild type C57BL6/J mice were purchased from The Jackson Laboratory at 2-3 months

old (Bar Harbor, Maine, USA) and housed under normal husbandry conditions with food and

water ad libitum. All animal protocols were approved by the St. Michael’s Hospital Animal

Care Committee.

9.1.2 Experimental protocol

After establishing a baseline (FiO2=21%) for 10min, anesthetised (3% isoflurane) mice

were given an i.v. injection of vehicle (saline) or metoprolol (3mg·kg-1)111 and allowed to

achieve a post-drug baseline for an additional 10min. In order to test whether β-blockade

impaired the physiologic response to a stressor, mice were given a hypoxic challenge

(FiO2=15%) for 15min and were subsequently exposed to an FiO2 of 21% during a 10 min

recovery phase (Figure 9.1.1). Following the recovery phase, mice were euthanized by cervical

dislocation.

9.1.3 Heart rate and mean arterial blood pressure

Anesthetised mice had their femoral artery and vein exposed. The artery was cannulated

with PE10 tubing (427401 Intramedic, BD Biosciences, Mississauga, ON, Canada) filled with

heparinised saline and connected to a PowerLab pressure transducer (TCB-600, Millar

Instruments, Inc., Houston, TX, USA). The femoral vein was cannulated with PE10 tubing

attached to syringe to allow the injection of saline or metoprolol. PowerLab ECG electrodes

(ML224, ADInstruments, Colorado Springs, CO, USA) were inserted subcutaneously to

Page 41: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

33

PMOD

PowerLab

PBrmvO2

HR, Temp

Vein

Artery

PowerLabHR

MAP

Baseline Post-Drug

Saline/Metoprolol

HypoxiaFiO2 =15%

RecoveryFiO2=21%

10 min 10 min 15 min 10 min

Oxyphor G2Saline or Metoprolol

Saline or Metoprolol

Figure 9.1.1 Experimental protocol for assessing MAP and PBrmvO2 with metoprolol One group of mice was assigned to microvascular cerebral oxygen tension (PBrmvO2) measurements and a separate group was assessed for mean arterial pressure (MAP).

Page 42: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

34

measure heart rate. PowerLab data acquisition systems (Chart, ADInstruments) were used to

record these physiological outcomes during the protocol outlined in the previous section.

9.1.4 Microvascular brain oxygen tension

The tail veins of anesthetised mice were cannulated to allow injection of phosphorescent

dye and saline or metoprolol. PMOD 5000 LED emitting and phosphorescent detecting probes

(Oxygen Enterprises, Philadelphia, PA, USA) were placed approximately 0.5 cm apart directly

above the exposed skulls of mice. Mice were injected with Oyxphor G2-phosphorescent dye

(Pd-tetra-(4-carboxyphenyl) tetrabenzoporphyrin dendrimer) allowing measurements of

phosphorescence quenching by oxygen through the PMOD 5000 probes. Using

phosphorescence lifetime, the PMOD data acquisition system can calculate absolute measures of

microvascular oxygen tension. ECG probes were also inserted subcutaneously in the left and

right front paws and left hind leg of these mice. After the phosphorescent dye was injected, mice

went through the hypoxic challenge protocol outlined above in section 9.1.2. (n=5 for saline

treatment, n=6 for metoprolol treatment).

9.1.5 Cardiac responsiveness and left ventricular function

In a separate group of animals (n=6) we measured left-ventricular function during

metoprolol treatment. Animals were placed on a warming pad (37˚C), intubated, and ventilated

using positive pressure (110 beats/min) with 2% (v/w) isoflurane admixed with 100% O2. Mice

were secured in a recumbent position and the right jugular vein was cannulated. Pressure was

calibrated after warming the catheter in 0.9% NaCl at 37˚C for 30 minutes. The right internal

carotid was then identified and ligated cranially. The 1.2F miniaturized combined conductance

catheter-micro-manometer (#FT112B Scisense Inc, London, Canada) was inserted into the right

carotid artery and advanced into the left ventricle until stable PV loops were obtained. After 10

Page 43: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

35

minutes, 0.9% heparized saline was injected via the right jugular vein and a steady state

achieved. Following saline, 3mg·kg-1 of metoprolol was administered and another steady state

was reached. All loops (pre and post beta blockade) were obtained with the ventilator turned off

for 5 -10 seconds and the animal apnoeic. Data were then acquired under steady state

conditions.

  Using the pressure conductance data, a range of real-time functional parameters were

then calculated using the ADVantage systemTM (Scisence Inc). These included: end diastolic

pressure (EDP), end systolic pressure (ESP), end diastolic volume (EDV), end systolic volume

(ESV), cardiac output (CO), the time constant of relaxation (Tau ), arterial elastance (Ea) and

systemic vascular resistance (SVR).

9.2 Effect of metoprolol in the mesenteric resistance artery in vitro

9.2.1 Animals

Wild type C57BL6/J mice were purchased from Charles River Laboratories at 2-3 months old

(Montreal, QC, Canada) and housed under normal husbandry conditions (12:12h light-dark

cycle, chow and water ad libitum). All animal protocols were approved by the University

Animal Care Committee (University of Toronto, Toronto, ON, Canada).

9.2.2 Mesenteric resistance artery isolation

Mice were euthanized via cervical dislocation and the small and large intestines were

removed and placed in MOPS-buffered salt solution (pH 7.4, NaCl 145, KCl 4.7, CaCl2·2H2O

1.5, MgSO4·7H2O 1.17, NaH2PO4.2H2O 1.2, pyruvate 2.0, EDTA 0.02, MOPS 3.0, glucose 5.0

mmol/L) on ice. The large intestine distal to and including the cecum were dissected and

discarded as was the duodenum. The remaining portion of the intestine were then carefully

Page 44: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

36

A B

C

Figure 9.2.1 Photographs of artery isolation and cannulation. Mesenteric resistance artery (A) and posterior cerebral artery (B) locations within their respective organs. C) Posterior cerebral artery mounted on glass micropipettes and pressurized to 45mmHg.

Page 45: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

37

spread in a circular pattern and held in place by pins to expose the mesenteric circulation (Figure

9.2.1A). Second generation mesenteric resistance arteries (MRAs; diameter≈200µm) were

carefully dissected and cleaned and transferred to the MOPS containing organ baths of the

pressure myography chambers.

9.2.3 Pressure myography

Pressure myography chambers were fitted with a coverglass bottom and could

accommodate up to 5mL of buffer solution within the organ bath. Vessels were imaged with an

inverted light microscope (Leica, Richmond Hill, ON, Canada). A monochrome CCTV camera

(Panasonic, Mississauga, ON, Canada) was attached to the microscope and the output signal was

sent to a TV monitor (Living Systems Instrumentation, Burlington, VT, USA). A video

dimension analyzer (V-94; Living Systems Instrumentation) was used to accurately measure

real time changes in vessel lumen diameter by sensing optical density alterations in the vessel

image displayed on the monitor (Figure 9.2.2). Output from the dimension analyzer was

recorded by data acquisition systems (DI-720, DATAQ Instruments, Akron, OH, USA and

MP100, BIOPAC Systems Inc, Goleto, CA, USA).

Isolated arteries were tied to two glass cannulae in a pressure myography vessel chamber

(LS-C1-SH; Living Systems Instrumentation) and held at a constant transmural pressure using a

pressure servo controller with peristaltic pump (PS-200; Living Systems Instrumentation).

Mounted arteries were pressurized with no flow to 45mmHg and warmed to 37°C for 30

minutes (Figure 9.2.1C). After warming, MRAs were brought to a pressure of 60mmHg and

equilibrated for 10 min before undergoing a viability check. Vessels were constricted with 1µM

of the α1-adrenergic agonist phenylephrine for 4 minutes or until a steady state was reached.

Viable vessels (30-50% constriction from baseline) continued through their respective protocols,

Page 46: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

38

diameter

time

Figure 9.2.2 Set up of the pressure myography apparatus Mounted vessels in myography chambers are imaged using an inverted light microscope. The image is displayed on a TV monitor attached to data acquisition systems which measure changes in lumen diameter by detecting differences in contrast ratios of the vessel wall. This data is recorded on a computer.

Page 47: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

39

otherwise they were discarded. In all protocols, doses were added by removing 1mL of solution

from the organ bath and replacing it with 1mL of the desired dose.

9.2.4 Effect of metoprolol on beta1,2-adrenergic mediated vasodilation

Doses are applied by replacing 1mL of solution from the 5mL organ bath with the

desired dose of agonist. Viable vessels were initially preconstricted with 1 µM phenylephrine

for 2 min to induce steady state tone and then exposed to increasing concentrations of the non-

selective β1,2-adrenergic receptor (β-AR) agonist isoproterenol (0.3µM-200µM). Each individual

dose of isoproterenol contained a maintenance dose of phenylephrine (1µM) and was added to

the organ bath until a steady-state was reached (2 min). The vessel diameter after every given

dose was recorded. This isoproterenol dose-response curve served as the control curve.

Following the final dose of isoproterenol, the organ bath was replaced with 37°C MOPS buffer

and the vessel was allowed to equilibrate for 10 min before changing the bath with 0, 5, 10 or

50µM metoprolol in MOPS for 30 min incubation period. These concentrations were chosen as

they correspond to the plasma concentration of 3mg·kg-1 metoprolol (i.v.) measured by HPLC in

rats111. Following incubation, vessels were preconstricted and the isoproterenol dose-response

was repeated. During this second isoproterenol dose-response, phenylephrine (1µM) and

metoprolol (5, 10 or 50µM) concentrations were maintained in the organ bath. The organ bath

was then changed and vessels were equilibrated for 10 min before being incubated in a Ca2+-free

MOPS buffer for 30 min to determine the maximum passive diameter.

9.2.5 Effect of metoprolol on beta2-adrenergic mediated vasodilation

The above protocol in section 0 was repeated to assess the effect of 50µM metoprolol on

the vasodilatory dose-response curve of clenbuterol (0.3 µM-300µM), a β2-selective agonist,

instead of isoproterenol.

Page 48: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

40

9.3 Effect of metoprolol in the posterior cerebral artery in vitro

9.3.1 Animals

Wild type C57BL6/J mice were purchased from Charles River Laboratories at 2-3

months old (Montreal, QC, Canada) and housed under normal husbandry conditions (12:12h

light-dark cycle, chow and water ad libitum). All animal protocols were approved by the

University Animal Care Committee (University of Toronto, Toronto, ON, Canada).

9.3.2 Posterior cerebral artery isolation

Mice were euthanized via decapitation. The skull was exposed and then removed; the

brain was carefully dissected and placed in MOPS solution on ice. Ventrally, portions of the

posterior cerebrum, the cerebellum and the medulla were cut such that the resulting piece of

brain tissue encompassed the posterior cerebral artery (PCA; diameter≈150µm; Figure 9.2.1B).

The segment of cerebellum was carefully removed, exposing the PCA. The PCA was carefully

pulled free from the cerebral tissue and any remaining connective tissue was removed by blunt

dissection. This process was repeated for both left and right PCAs. Arteries were then

transferred to the MOPS filled organ baths of the perfusion myography chambers.

9.3.3 Pressure myography

Refer to section 9.2.3. Note that PCAs were maintained at a transmural pressure of

45mmHg and were checked for viability immediately after the 30 min warming period. Because

of reduced phenylephrine induced tone, the viability dose of phenylephrine was increased to

10µM in PCAs as opposed to 1µM in MRAs.

Page 49: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

41

9.3.4 Effect of metoprolol on adrenergic mediated vasomotor function

The preconstriction and maintenance dose of phenylephrine in PCAs was 3µM PE (See

Appendix B). In this series of experiments, PE dose-response curves (0.03-30µM) were first

generated in viable vessels. Each dose was held for 2 minutes to ensure a steady state was

reached. Following the PE dose-response, the bath was changed and the vessels were allowed to

equilibrate for 10 minutes. ISO dose-response curves (0.03-3000µM) were then generated with

each dose again being held for 2 minutes. The organ bath was then changed and after 10 minutes

was replaced with unaltered warm MOPS buffer (0µM metoprolol; n=5) or 50µM metoprolol

(n=7) in MOPS buffer and held for 30 minutes. Following this incubation period, PE and ISO

dose-response curves were repeated as above. During these dose-response curves, metoprolol

(50µM) concentrations were maintained in the organ bath. Additionally, phenylephrine (3µM)

concentration was maintained in the second ISO dose-response. The bath was changed after the

ISO curve and held for 10 minutes before being replaced with Ca2+ free MOPS buffer.

9.4 Drugs and solutions

The following drugs were used; L-phenylephrine hydrochloride (P6126), ±-metoprolol

(+)-tartrate salt (M5391), DL-isoproterenol hydrochloride (I5627), and clenbuterol

hydrochloride (C5423; Sigma-Aldrich, St. Louis, MO, USA).

Metoprolol tartrate solution as Betaloc® 1mg/mL (Prod. No. 1332, AstraZeneca Canada

Inc., Mississauga, ON, Canada) was used in whole animal studies.

Page 50: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

42

9.5 Statistical analysis

9.5.1 Effect of metoprolol on brain O2 tension and hemodynamics in vivo

Analysis of differences in heart rate, mean arterial pressure and cerebral microvascular

oxygen tension were performed by two-way repeated measures ANOVA. Analysis of cardiac

responsiveness and left ventricular function were performed by paired t-test. All data is

presented as mean ± SEM. Statistical analyses were done in SigmaPlot 11 (Systat Software Inc.,

Chicago, IL, USA).

9.5.2 Effect of metoprolol in mouse arteries

Acute diameter measurements (diameasured, representing steady state diameter following

dose of agonist) were normalized to represent either tone or percent dilation. Tone represents the

proportion of constriction relative to the Ca2+ free diameter (diamax). Tone is calculated as

follows:

Equation 9.5.1

100 dia

diadia)dia of (% tone

max

measuredmaxmax

An increase in tone corresponds to an increased degree of vasoconstriction in the vessel. A

reduction in tone would correspond to a reduced degree of vasoconstriction.

Percent dilation represents the percent change in diameter from the minimum diameter

observed (diamin) normalized to diamax. Percent dilation is calculated as follows:

Equation 9.5.2

100dia-dia

dia-diadilation %

minmax

minmeasured

An increase in percent dilation is indicative of increased vasodilation.

Page 51: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

43

Data are presented as mean ± SEM, n-values indicated number of animals or number of

vessels tested. Dose-response data of agonist before and after incubation with metoprolol (or

MOPS buffer) are tested by two-way repeated measures ANOVA. The concentration of agonists

that elicited half the maximal response of a dose-response curve is expressed as the EC50 value

and is represented as the log[EC50]. Additionally, dose-response curves will be assessed

quantitatively by their Emax value which represents the magnitude of the response at the

uppermost plateau of the dose-response curve. This value is extrapolated when it is not reached

experimentally. Mean EC50 and Emax values are tested by paired t-test where both curves were

generated in the same artery otherwise they were tested by unpaired student’s t-test.

Vasoconstrictor (tone) and vasodilator (% dilation) responses to any one dose of agonist before

and after metoprolol (or MOPS buffer) incubation are tested by paired t-tests. Statistical

analyses were performed in Prism 5 (GraphPad Software Inc., La Jolla, CA, USA). Differences

were significant at an alpha value less than 0.05.

Page 52: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

44

10 Results

10.1 Effect of metoprolol on brain O2 tension and hemodynamics

10.1.1 Metoprolol injection reduced brain O2 tension and heart rate

Metoprolol injection dropped heart rate and microvascular brain oxygen tension

(PBrmvO2) while mean arterial pressure (MAP) was maintained immediately after drug injection

(Figure 10.1.1). Hypoxic challenge increased the heart rate in saline treated animals which was

subsequently decreased during the recovery phase. β-blocked mice maintained a depressed heart

rate throughout the protocol. MAP and PBrmvO2 were decreased in both treatments following

hypoxic challenge and increased during the recovery phase (Figure 10.1.1).

Metoprolol treatment significantly depressed the heart rate compared to baseline (469 ±

13.2 vs. 548 ± 13.0 bpm; n=12, p<0.05) throughout the experimental protocol while saline

treated animals had a significant increase in heart rate compared to baseline (600 ± 32.7 vs. 552

± 20.2 bpm; n=11, p<0.05) only during hypoxic challenge (Figure 10.1.2A). There was no

difference in MAP between treatment groups (Figure 10.1.2B). Hypoxic challenge significantly

dropped MAP in both groups compared to baseline (saline: 62.0 ± 3.8 vs. 73.2 ± 1.7 mmHg;

n=6, p<0.05; metoprolol: 67.0 ± 3.0 vs. 71.7 ± 0.9 mmHg; n=6, p<0.05). Directly following

metoprolol injection, PBrmvO2 (Figure 10.1.2C) was significantly decreased compared to

baseline (60.8 ± 2.5 vs. 68.9 ± 1.6 mmHg; n=6; p<0.05) and compared to saline treatment (60.8

± 2.5 vs. 69.7 ± 2.1 mmHg; n=6, 5; p<0.05). There was no difference between metoprolol and

saline treatment during hypoxia or during the 21% FiO2 recovery phase. Hypoxic challenge

significantly dropped PBrmvO2 in both groups compared to baseline (p<0.05) and recovery at

21% FiO2 did not return PBrmvO2 to baseline levels (p<0.05) in both groups. Figure 10.1.3

depicts the degree of change from baseline of each condition in both treatment groups.

Page 53: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

45

10.1.2 Metoprolol injection reduced CO and increased SVR

Metoprolol injection significantly reduced cardiac responsiveness compared to saline

treatment in the same mouse (Figure 10.1.4). A representative tracing of a pressure-volume loop

is shown in Figure 10.1.5. The P-V loop indicates that preload (the lower right hand corner of

the curve, representing left ventricular end-diastolic pressure and volume) is increased following

metoprolol injection. The slope of the end-systolic pressure-volume relationship is smaller

following metoprolol injection, suggesting reduced ventricular contractility. The slope of the

end-diastolic pressure-volume relationship is steeper following metoprolol injection which

suggests decreased left ventricular compliance. Additionally, the area under the curve,

representing left ventricular stroke work, is smaller following metoprolol treatment. Heart rate

was significantly decreased 10 minutes after metoprolol injection compared to 10 minutes after

saline injection (399 ± 23.9 vs. 485 ± 23.3 bpm; n=6, p<0.05; Figure 10.1.4A). Similarly, stroke

volume was reduced following metoprolol compared to saline (19.7 ± 1.8 vs. 26.3 ± 1.5

µL/beat; n=6, p<0.05; Figure 10.1.4B). As a result of decreases in heart rate and stroke volume,

cardiac output was also significantly decreased following metoprolol treatment (7.96 ± 0.8 vs.

12.8 ± 1.1 mL/min; n=6, p<0.05; Figure 10.1.4C). Systemic vascular was consequently

increased in metoprolol treated animals (9.5 ± 0.6 vs. 5.3 ± 0.2 dynes/cm5 ; n=6, p<0.05; Figure

10.1.4D). There were no differences between left ventricular end-systolic and end-diastolic

pressures (Figure 10.1.4E, F) and volumes (Figure 10.1.4G, H).

Page 54: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

46

He

art

Rat

e, b

pm

300

400

500

600

700

Mea

n A

rter

ial P

ress

ure,

mm

Hg

40

50

60

70

80

90

100

40

60

80

100

Time, min

0 10 20 30 40 50

Mic

rova

scu

lar

Bra

in O

2 T

ensi

on,

mm

Hg

20

40

60

80

100

A

B

C

SalineMetoprolol (3 mg kg-1)

Saline/Metoprolol

FiO2=15% FiO2=21%

Saline/Metoprolol

FiO2=15% FiO2=21%

Saline/Metoprolol

FiO2=15% FiO2=21%

Figure 10.1.1 Time course of hypoxic challenge protocol with saline/metoprolol. Changes in heart rate (A), mean arterial pressure (B) and microvascular brain O2 tension (C) over time during before and after beta-blocker or saline administration, during hypoxic stress (FiO2=15%) and during recovery at FiO2=21%. Data points represent mean ± SEM.

Page 55: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

47

Hea

rt R

ate,

bpm

0

200

400

600

800

Mea

n A

rter

ial P

ress

ure,

mm

Hg

0

20

40

60

80

100

Baseline Drug FiO2 15% FiO2 21%

Mic

rova

scul

ar B

rain

O2

Ten

sion

, m

mH

g

0

20

40

60

80

Saline

Metoprolol (3 mg kg-1)

A

B

C

*, #

*

**

*, #

*, #

* *

*, #

*

*

Figure 10.1.2 Mean heart rate, MAP and PBrmvO2 in hypoxic challenge with saline/metoprolol. Graph of mean heart rate (A), MAP (B) and PBrmvO2 (C) during each experimental condition in hypoxic challenge protocol. Error bars represent SEM. * p<0.05 from baseline, # p<0.05 between treatment group; two-way repeated measures ANOVA.

Page 56: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

48

H

ea

rt R

ate

, bp

m

-150

-100

-50

0

50

100

150

200

MA

P,

mm

Hg

-20

-10

0

10

20

30

Drug FiO2 15% FiO2 21%

PB

r mvO

2,

mm

Hg

-60

-40

-20

0

20

40

SalineMetoprolol (3 mg kg-1)

A

B

C

*

*

*

#

*

**

Figure 10.1.3 Delta heart rate, MAP and PBrmvO2 in hypoxic challenge with saline/metoprolol. Mean change (Δ) in heart rate (A), MAP (B), and PBrmvO2 (C) from baseline condition. Error bars represent SEM. * p<0.05 between treatment groups, # p<0.05 within treatment group; two-way repeated measures ANOVA.

Page 57: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

49

He

art

Ra

te,

bpm

0

100

200

300

400

500

600

Str

oke

Vo

lum

e,

uL

/be

at

0

5

10

15

20

25

30

Car

dia

c O

utp

ut,

mL

/min

0

2

4

6

8

10

12

14

16

End

Sys

tolic

Pre

ssur

e, m

mH

g

0

20

40

60

80

100

120

En

d D

iast

olic

Pre

ssu

re,

mm

Hg

0

2

4

6

8

10

12

14

Saline Metoprolol

En

d S

ysto

lic V

olu

me,

uL

0

10

20

30

40

50

60

70

Saline Metoprolol

End

Dia

stol

ic V

olu

me

, u

L

0

20

40

60

80

100

* *

*

A B

C D

E F

G H

SV

R,

dyn

es/

cm5

0

2

4

6

8

10

12

*

Figure 10.1.4 Hemodynamic changes following saline and subsequent metoprolol injections. Heart rate (A), stroke volume (B), cardiac output (C) and systemic vascular resistance (SVR; panel D) are significantly reduced following metoprolol (3mg·kg-1) injection End-systolic and diastolic pressures and volumes (E, F, G, H) are not affected by metoprolol treatment. Error bars represent SEM. n=6; * p<0.05 by paired t-test.

Page 58: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

50

0

20

40

60

80

100

120

0 10 20 30 40 50 60 70 80

Left

ven

tric

ula

r pre

ssu

re, m

mH

g

Volume, uL

Saline Metoprolol (3mg/kg)

Figure 10.1.5 Representative pressure-volume loop before and after metoprolol treatment. Tracing of a pressure-volume loop recorded in a single mouse after 10 minutes of i.v. saline followed by 10 minutes of i.v. metoprolol.

Page 59: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

51

10.2 Metoprolol inhibits isoproterenol mediated vasodilation MRAs

A representative tracing of the entire time course of the experimental protocol is shown

in Figure 10.2.1. Isoproterenol (ISO) dose-response curves generated before incubation with

metoprolol are considered controls. There was no effect of time on the ISO dose-response curve

as incubation in 0µM metoprolol was no different from its control (Table 10.1; Figure 10.2.2A).

In MRAs the LogEC50 values of ISO in 5 and 10M metoprolol were not significantly different

than their respective control curves or from each other (Table 10.1; Figure 10.2.2B, C). A

significant right-shift in the ISO dose-response curve was observed at a metoprolol

concentration of 50µM (-4.1 ± 0.1 vs -4.4 ± 0.1; n=6; p<0.05; Figure 10.2.2D). The LogEC50 of

ISO at 50M metoprolol was significantly higher than at 0, and 5M metoprolol (-4.1 ± 0.1 vs -

4.6 ± 0.1 and -4.6 ± 0.04; n=6, 3, 7, p<0.05). In addition, the percent dilation at the control EC50

dose of isoproterenol (30µM or LogEC50=-4.5) was significantly attenuated by metoprolol at 10

(31 ± 4.2 vs. 40 ± 1.8%; n=7; p<0.05; Figure 10.2.3C) and at 50µM (17 ± 2.3 vs. 34 ± 2.0%;

n=6; p<0.05; Figure 10.2.3D). At all concentrations of metoprolol, dose-response curves

reached equivalent Emax plateaus (Figure 10.2.4).

The clenbuterol (β2-specific agonist) dose-response curve is shown in Figure 10.2.5A.

The LogEC50 values are not statistically different, though there is a slight increase at 50µM

metoprolol (from -4.9 ± 0.04 to -4.7 ± 0.1; Table 10.1). The percent dilation at the control EC50

dose (10µM or LogEC50=-5) is significantly attenuated at 50µM metoprolol (25 ± 1.6 vs. 34 ±

1.0; n=5, p<0.05; Figure 10.2.5B).

Page 60: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

52

0 2500 5000 7500 10000100

150

200

250

3001M PE

ISO DRC

50M Metoprolol

1M PECa2+ Free

ISO DRC

-6.5

-6

-5.5

-5.0

-4.5

-4.0

-6.5

-6 -5.5-5.0

-4.5

-4.0

time, sec

dia

met

er,m

Figure 10.2.1 Representative tracing of MRA experimental protocol. Time course of experimental protocol in mesenteric resistance arteries; the first dose-response curve (DRC) is considered the control to which the second dose-response curve (with metoprolol in the bath) is compared to. Dose numbers represent 10xµM of ISO. PE=phenylephrine; ISO=isoproterenol.

Page 61: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

53

Control (- Metoprolol) + Metoprolol

ISO ± 0µM Metoprolol (n=3) -4.5 ± 0.1 -4.6 ± 0.08

ISO ± 5µM Metoprolol (n=7) -4.6 ± 0.05 -4.6 ± 0.04

ISO ± 10µM Metoprolol (n=7) -4.6 ± 0.06 -4.4 ± 0.1

ISO ± 50µM Metoprolol (n=6) -4.4 ± 0.07 -4.1 ± 0.08*,#

CLEN in 50µM Metoprolol -4.9 ± 0.04 -4.7 ± 0.1

Table 10.1 Half maximal concentrations for MRA dose-response curves. LogEC50 values for isoproterenol (ISO) in 0, 5, 10 and 50µM metoprolol and clenbuterol (CLEN) in 50µM metoprolol. Control values are EC50s in the absence of the stated dose of metoprolol. * p<0.05 between columns (paired t-test); # p<0.05 within column (one-way ANOVA).

Page 62: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

54

-8 -6 -40

20

40

60

80 Control, n=3

0M Metoprolol, n=3

log [isoproterenol (mol/L)]

% d

ilat

ion

-7 -6 -5 -4 -30

20

40

60

80 Control, n=7

5M Metoprolol, n=7

log [isoproterenol (mol/L)]

% d

ilat

ion

-7 -6 -5 -4 -30

20

40

60

80 Control, n=7

10M Metoprolol, n=7

log [isoproterenol (mol/L)]

% d

ilat

ion

-7 -6 -5 -4 -30

20

40

60

80 Control, n=6

50M Metoprolol, n=6

* ***

**

log [isoproterenol (mol/L)]

% d

ilat

ion

A B

C D

Figure 10.2.2 Effect of metoprolol on isoproterenol dose-response curves in mesenteric arteries. Isoproterenol (ISO) dose-response curves were generated before (control) and after incubation with metoprolol in mesenteric resistance arteries. The ISO dose-response curve was unaffected by both time (A) and 5µM of metoprolol (B). There is a small rightward shift of the ISO dose-response curve at 10uM metoprolol, though the difference in EC50s is not significant (log[EC50]=-4.6 ± 0.06 vs -4.4 ± 0.1; n=7; panel C). There is a significant rightward shift in the ISO dose-response curve from –4.4 ± 0.07 to -4.1 ± 0.08 at 50µM metoprolol (paired t-test, p<0.05, n=6; panel D). Data points represent mean ± SEM. * p<0.05 by two-way repeated measures ANOVA.

Page 63: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

55

Control 5M Metoprolol0

20

40

60

% d

ilat

ion

at

ISO

EC

50

(30

M)

Control 0uM Metoprolol0

20

40

60%

dil

atio

n a

t IS

O E

C5

0 (3

0 M

)

Control 10uM Metoprolol0

20

40

60

*

% d

ilat

ion

at

ISO

EC

50 (

30M

)

Control 50uM Metoprolol0

20

40

60

*

% d

ilat

ion

at

ISO

EC

50 (

30M

)

A B

C D

Figure 10.2.3 Percent dilation to EC50 dose (30µM) of isoproterenol in mesenteric arteries. The percent dilation at the dose of isoproterenol (ISO) which elicits half the maximal response (30µM) from mesenteric resistance arteries is affected by time (A) or 5uM metoprolol (B). At 10uM metoprolol there is a significant reduction in the percent dilation to 30µM ISO (40 ± 1.8% vs. 31 ± 4.3%; paired t-test, p<0.05, n=7; panel C). There is a significant reduction in the percent dilation to 30µM ISO from 34 ± 2.0% to 17 ± 2.3% at 50uM metoprolol (paired t-test, p<0.05, n=6; panel D). Data presented as mean ± SEM.

Page 64: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

56

Control 0M Metoprolol0

20

40

60

80

100IS

O E

ma

x,

% d

ilat

ion

Control 5M Metoprolol0

20

40

60

80

100

ISO

Em

ax,

% d

ilat

ion

Control 10M Metoprolol0

20

40

60

80

100

ISO

Em

ax,

% d

ilat

ion

Control 50M Metoprolol0

20

40

60

80

100

ISO

Em

ax,

% d

ilat

ion

A B

C D

Figure 10.2.4 Percent dilation at Emax of isoproterenol dose-response curves. The % dilation to isoproterenol at Emax of the best-fit curves is not affected by time (A), 5 (B), 10 (C) or 50µM(D) metoprolol. Data presented as mean ± SEM. * p<0.05 by paired t-test.

Page 65: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

57

A

-7 -6 -5 -4 -30

20

40

60

80

100 Control, n=5

50M Metoprolol, n=5

*

*

log [clenbuterol (mol/L)]

% d

ilat

ion

Control 50uM Metoprolol0

20

40

60

80

100

*

% d

ilat

ion

at

CL

EN

EC

50

(10

M)

Control 50M Metoprolol0

20

40

60

80

100

CL

EN

Em

ax,

% d

ilat

ion

CB

Figure 10.2.5 Effect of metoprolol on clenbuterol mediated vasodilation. The percent dilation at intermediate doses of clenbuterol (β2-specific agonist) is significantly attenuated at 50µM metoprolol (panel A; * p<0.05 by two-way repeated measures ANOVA). The percent dilation to the EC50 dose of clenbuterol (10µM or LogEC50=-5; panel B) is significantly attenuated at 50µM metoprolol (25 ± 1.6 vs. 34 ± 1.0; n=5, p<0.05). Data presented as mean ± SEM.

Page 66: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

58

10.3 Metoprolol inhibits isoproterenol mediated vasodilation inPCAs

The effect of phenylephrine in PCAs has not been previously assessed; therefore it was

necessary to determine optimal phenylephrine preconstriction and maintenance doses before

generating isoproterenol dose-response curves. See Appendix B for work up of posterior

cerebral artery to determine optimal phenylephrine preconstriction dose in addition to the effect

of metoprolol on phenylephrine dose-response curves.

Isoproterenol (ISO) dose-response curves were generated before and after metoprolol

incubation (0µM metoprolol represents a time control, Figure 10.3.1). There was a significant

time effect on the logEC50 value of isoproterenol; the second ISO dose-response curve was

shifted to the right compared to the first (LogEC50=-4.1 ± 0.18 vs. -4.5 ± 0.10; n=5, p<0.05;

Table 10.2). However, two-way repeated measures ANOVA of the ISO dose-responses before

and after 0µM metoprolol showed no significant treatment or interaction effect (Figure

10.3.2A). Additionally, time had no effect on the percent dilation induced by the EC50 dose of

ISO (-4.5 or 30µM), as seen in Figure 10.3.2C, or on the Emax value of the ISO DRCs before and

after 0µM metoprolol (Figure 10.3.2D).

There was a significant treatment and interaction effect before and after incubation with

50µM metoprolol (two-way repeated measures ANOVA, n=7, p<0.05; Figure 10.3.2B). The

LogEC50 was right shifted at 50µM metoprolol (-4.0 ± 0.13 vs. -4.5 ± 0.24; n=7, p<0.05; Table

10.2). The % dilation at the EC50 dose of ISO was reduced at 50µM metoprolol, but the effect

was not significant (23 ± 4.8 vs. 37 ± 6.6 %; n=7; Figure 10.3.2C). The Emax of ISO was

significantly reduced at 50µM metoprolol (61 ± 5.5 vs. 79 ± 4.0 %; n=7, p<0.05; Figure

10.3.2D).

Taking into account a possible effect of time on the isoproterenol dose-response, the

DRC generated at 50µ metoprolol was compared to its time matched control (ie. the curve

Page 67: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

59

generated at 0µM metoprolol). LogEC50 values and the percent dilation at this dose were not

significantly different, however there was a significant reduction in Emax at 50µM metoprolol

compared to its time matched control (61 ± 5.5 vs. 79 ± 3.7; n=7, 5, p<0.05; Figure 10.3.2D).

Page 68: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

60

0 2500 5000 7500 10000 125000

50

100

150

20010M PE

PE DRC

10M PE

ISO DRC

0uM Metoprolol

10M PE

Ca2+ Free

PE DRC ISO DRC

-7.5-6.5

-5.5-4.5

-7.5 -5.5

-3.5

-7.5-6.5

-5.5

-7.5 -5.5

-3.5

-4.5

time, sec

dia

met

er,m

Figure 10.3.1 Representative tracing of PCA protocol. Time course of a single PCA showing dose-response curves (DRC) before and after metoprolol (0 or 50µM) incubations. Dose numbers represent 10xµM of PE or ISO. PE=phenylephrine; ISO=isoproterenol.

Page 69: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

61

-8 -6 -4 -20

20

40

60

80

100 Control, n=5

0M Metoprolol, n=5

log [isoproterenol (mol/L)]

% d

ilat

ion

-8 -6 -4 -20

20

40

60

80

100 Control, n=7

50M Metoprolol, n=7 **

log [isoproterenol (mol/L)]

% d

ilat

ion

M M

et

- 0

M M

et

+

0M

Met

- 5

0M

Met

+

50

0

20

40

60

80

100

, #

Em

ax,

% d

ilat

ion

M M

et

- 0

M M

et

+

0M

Met

- 5

0M

Met

+

50

0

20

40

60

80

100

EC

50

(30

M)

dil

atio

n,

%A B

C D

Figure 10.3.2 Effect of metoprolol on isoproterenol dose-response in PCAs. Isoproterenol (ISO) dose-response curves were generated before (control) and after incubation with metoprolol in posterior cerebral arteries The ISO dose-response curve is not affected by time (panel A) but is significantly blunted at high doses by 50µM metoprolol (panel B, D). Emax of isoproterenol at 50µM metoprolol is significantly lower than its internal control as well as its time control. Data presented as mean ± SEM. * p<0.05 by two-way repeated measures ANOVA; ɣ p<0.05 by paired t-test; # p<0.05 by unpaired t-test.

Page 70: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

62

Control (-Metoprolol) + Metoprolol

± 0µM Metoprolol (n=5) -4.5 ± 0.1 -4.1 ± 0.2*

± 50µM Metoprolol (n=7) -4.5 ± 0.2 -4.0 ± 0.1*

Table 10.2 Half maximal concentrations of isoproterenol in PCAs. LogEC50 values for ISO dose-response curves in PCAs. Data presented as mean ± SEM. *p<0.05 between columns (paired t-test); # p<0.05 within columns (unpaired-test).

Page 71: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

63

11 Discussion

The novel findings of this research demonstrate the dual impact of metoprolol on CO,

cerebral perfusion and adrenergic mediated vasodilation in isolated mesenteric and cerebral

arteries. Our experimental results support the hypothesis that metoprolol may impair vital organ

perfusion by limiting cardiac output (global ischemia) and adrenergic mediated vasodilation at

the level of the small resistance artery (organ specific ischemia). When metoprolol was

administered to anesthetized mice in vivo, MAP was maintained while HR, SV and CO were

reduced. The associated increase in SVR resulted in decreased brain perfusion as indicated by a

reduction in cerebral microvascular oxygen tension. However, the mechanism by which SVR

was increased could not be determined in a whole animal experiment. It may have been due to a

reflex increase in sympathetic tone, or as a direct action of metoprolol on resistance arteries. In

order to explore the latter mechanism, we performed studies in isolated mesenteric resistance

arteries. Results demonstrated that metoprolol inhibited isoproterenol (β1/2) mediated

vasodilation in a dose dependent manner. This was at least partially mediated via the β2-

adrenergic receptor confirmed by data demonstrating that metoprolol also inhibited clenbuterol

mediated vasodilation. Metoprolol had a more profound effect on shifting the EC50 of the

isoproterenol dose-response curve, suggesting an additional β1-AR blocking effect. Since

clinical studies have implicated metoprolol therapy as a risk factor for increased stroke, we then

assessed the impact of metoprolol on isoproterenol mediated vasodilation in cerebral arteries.

Our results demonstrated that metoprolol also impaired adrenergic mediated vasodilation in

isolated posterior cerebral arteries. Because adrenergic stimulation in isolated mouse posterior

cerebral arteries has not been previously characterized, we assessed the effect of time and

metoprolol administration on the phenylephrine DRC. Our results demonstrated that the

Page 72: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

64

posterior cerebral artery has a reduced phenylephrine dose-response over time which is not

affected by metoprolol.

Collectively, these data suggest that metoprolol could directly impair cerebral artery

dilation under conditions which require increased cerebral blood flow to maintain oxygen

delivery. The combination of reduced cardiac output coupled with organ specific inhibition of

resistance artery dilation may explain in part the mechanism of increased stokes and mortality

which occur in β-blocked perioperative patients.

11.1 Beta-adrenergic vasodilation is required to maintain vital organ perfusion

Immediately following metoprolol administration, mean arterial pressure as well as left-

ventricular pressures were maintained, CO was decreased and SVR was increased.

Subsequently, there was a reduction in PBrmvO2. Importantly, the reduction in brain perfusion

that was observed in this study occurred in β-blocked mice under resting conditions with low

metabolic requirements (ie. anesthetised). These results can be correlated to the clinical

condition of hypertension. During hypertension the health of the vasculature is altered and an

increased risk of organ injury is observed (stroke, MI)36. Beta-blockers concurrently reduced

MAP and the incidence of cardiovascular complications including stroke in hypertensive

patients49. However, vasodilatory anti-hypertensive medications (ACE-I, CCB) demonstrate a

further reduction in the incidence of stroke, relative to hypertensive patients treated with β-

blockers49. These clinical data suggested that anti-hypertensives with direct vasodilatory action

on vascular smooth muscle may improve cerebral perfusion. This argument is further supported

by the clinical finding that patients treated with β-blockers that possess vasodilatory capacity

(carvedilol) have fewer ischemic events (stroke, MI) than those treated with non-vasodilatory β-

blockers (metoprolol)104. A vascular mechanism for increased stroke incidence in hypertensive

patients treated with β-blockers is supported by our whole animal data. Mice treated with

Page 73: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

65

metoprolol had a reduced HR, SV and CO and increased SVR. This hemodynamic pattern

resulted in a reduction in cerebral microvascular oxygen tension. In addition to limiting global

cardiac output, the direct effect of metoprolol to reduce adrenergic mediated vasodilation of

mesenteric and cerebral arteries might also increase the risk of brain ischemia. Additional

studies in hypertensive animal models would be required to test this hypothesis.

The impact of metoprolol on artery function may be further exacerbated during times of

increased oxygen demand (sympathetic activation) and/or reduced oxygen supply. During these

conditions, the brain and heart, organs with high metabolic requirements, are preferentially

perfused by active vasodilatory mechanisms112;113. In the heart, sympathetic activation results in

parallel increases in myocardial oxygen consumption and delivery95;96. The latter is regulated by

β-adrenergic receptor mediated coronary vasodilation. In swine and dogs, coronary vasodilation

was mediated by both β1- and β2-ARs during times of increased oxygen demand (exercise)95;96.

Inhibition of these receptors during exercise led to reduced oxygen delivery to the myocardium

and a greater mismatch of coronary oxygen supply and consumption95. We have not assessed

the impact of metoprolol on coronary arteries. However, clinical studies suggest that β-blockade

paradoxically increased the incidence of MI in patients who experience acute blood loss and

fluid resuscitation (hemodilution)107;114. The cardioprotective benefit associated with β-blockade

may therefore be negated by their inhibitory effect on coronary vasodilation during conditions

of increased myocardial oxygen demand and limited supply.

In the brain, cerebral blood flow is increased under conditions of increased cerebral

metabolic rate (increased neuronal activity)115 and reduced oxygen supply (hemodilution)116.

These responses are mediated in-part by β-adrenergic mechanisms110;117. In the clinical setting

of acute hemodilutional anemia, reduced blood oxygen content (reduced O2 supply) has been

associated with an increase in the incidence of stroke118;119. In a subset of the POISE trial,

Page 74: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

66

significant blood loss (surgical anemia) in β-blocked patients was associated with a 2-fold

increased risk of stroke45. This suggests that β-blockers may increase the risk of ischemic organ

injury during times of reduced blood oxygen content. In parallel studies, our lab has previously

assessed the effect of β1-blockade (metoprolol) and β2-blockade (ICI 118, 551) in a model of

acute hemodilutional anemia in rats. Hemodilution causes an increase in oxygen demand to vital

tissue in order to maintain organ perfusion: CO is increased, MAP is maintained and SVR

decreases110. In animals pretreated with metoprolol, MAP was maintained, however the CO

response was blunted suggesting a global reduction in oxygen supply (in addition to reduced

blood O2 content)110. Animals treated with the β2-antagonist also maintained MAP during

hemodilution, however the heart rate response was no different from control (CO was not

measured)117. Metoprolol treatment reduced cerebral blood flow and oxygen tension and

increased cellular markers of tissue hypoxia (HIF-1α)110. This was due in part to the reduction in

cardiac output; however the magnitude of the reduction in oxygen tension suggests that a

cerebrovascular mechanism may have also contributed. This is supported by evidence from the

β2-blocked anemic rats. These animals also had reduced cerebral blood flow and tissue oxygen

tension during hemodilution, despite having similar HR responses to control117. As in the

coronary circulation, there may be a dual role for β1- and β2-AR mediated vasodilation in

maintaining cerebral perfusion. This assertion is supported by our findings in isolated arteries.

In the current study, we observed an inhibitory effect of metoprolol on β1/2-AR mediated

vasodilation (isoproterenol) in cerebral arteries. In addition, we demonstrated that metoprolol

can inhibit β2-specific mediated vasodilation (clenbuterol) in mesenteric arteries. Metoprolol

shifted the isoproterenol dose-response more profoundly than it did the clenbuterol dose-

response, suggesting a contribution of β1-AR mediated vasodilation. We have provided direct

evidence that metoprolol impairs cerebral artery dilation by a β-AR mediated mechanism in

Page 75: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

67

response to an adrenergic stimulus This may explain the observed increased incidence of stroke,

MI and mortality in patients treated with metoprolol who experience acute blood loss45;107.

11.2 Metoprolol inhibits adrenergic vasodilation in mesenteric and cerebral arteries

In the mesenteric and posterior cerebral arteries there was a significant inhibitory effect

of metoprolol on the isoproterenol dose-response curve. This confirms that in our isolated vessel

model, a cardioselective β-blocker can impair the normal vasodilatory response of blood vessels

during increased demand (ie. sympathetic stimulation).

The relevance of our isolated vessel studies are supported by our lab’s previous animal

studies in anemic rats110;117 and by clinical findings. As described in 11.1, impaired cerebral β-

AR mediated vasodilation could contribute to the increased risks of stroke in β-blocked

perioperative and hypertensive (relative to ACE-I and CCB treatment) patients50-55;120. In

addition to studying vessels from our organ of interest (brain), we also studied vessels from the

mesenteric vascular bed due to their being well characterized as well as their ease of access. Gut

ischemia is not a commonly reported outcome measure in most β-blocker clinical trials.

However, a trial in post-MI patients, identified an almost 200% increase in relative risk of

developing gastrointestinal problems (event rate 1 vs 0.3 % in treatment vs. control group)19

with propranolol treatment. It is unknown whether these events were ischemic in nature; future

trials should therefore consider the effect of β-blockade on other, non-vital organ systems.

11.3 Metoprolol may be associated with increased morbidity and mortality

This study demonstrated that metoprolol can cross-react with the β2-AR to inhibit

vasodilation. This finding is supported in cell culture studies where metoprolol was identified as

having the lowest β1-selectivity compared with other β-blockers. Metoprolol has 2.3 fold

selectivity for the β1-AR compared to the β2-AR while bisoprolol shows 13.5 fold β1-AR

Page 76: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

68

selectivity4. The low selectivity of metoprolol can explain why there is evidence of increased

morbidity compared to other cardioselective β-blockers. In a retrospective analysis of the impact

of anemia on mortality risks, metoprolol was associated with an increased risk of death while

atenolol and bisoprolol were not108. In a recent meta-analysis of perioperative β-blocker trials,

Badgett et al. found a significant negative correlation of mortality with increasing β1-

selectivity56. Patients treated with metoprolol were at a significantly greater risk of dying

perioperatively than a patient treated with bisoprolol. The authors did not find a significant

correlation between stroke and β-blocker selectivity. There is, however, a trend towards this

relationship. In POISE, metoprolol treatment was associated with increased stroke45 while the

DECREASE IV trial found that there was no significant difference between placebo and

bisoprolol treated patients in stroke incidence46. These are two different outcomes with two β-

blockers with significantly different β1-AR selectivity. However, they also differ in their

mechanisms of metabolism, which may also contribute to negative patient outcomes.

Certain β-blockers (metoprolol, carvedilol, propranolol, labetalol and timolol) are

metabolized by the cytochrome P450 CD6 (CYP2D6) enzyme121. The gene for CYP2D6 is

highly polymorphic resulting in differing effectiveness of the enzyme in individuals. Those who

are poor metabolizers tend to experience increased adverse outcomes with β-blockade121. There

is consequently a significant correlation of increased perioperative mortality risk with β-

blockers that rely on CYP2D6 metabolism56. In addition, a short titration period up to the target

dose is associated with increased mortality56. In POISE, patients were treated with metoprolol

(CYP2D6) over a short titration period (2-4 hours preoperatively)45 while in DECREASE-IV,

patients were given bisoprolol (non-CYP2D6) over a long titration period (34 days

preoperatively)46. These could potentially explain the negative outcomes associated with

metoprolol; however receptor selectivity is still a relevant mechanism. A patient with a CYP2D6

Page 77: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

69

polymorphism making them a poor metabolizer of metoprolol would be overdosed by the

standard β-blocker dosage. The overdose could lead to cross-reactivity with β2-AR4 in addition

to occupation of the β1-AR to limit cerebral and coronary vasodilation95;96;110;117. Our results

with clenbuterol support the hypothesis that cross-reactivity with the β2-AR may be a significant

mechanism of increased harm in patients.

11.4 Differences between mesenteric and cerebral arteries

This study used 3rd order mesenteric arteries which makes them more similar to

resistance arteries122. These arteries are integral in the control of blood pressure within the body

due to their location as precapillary arteries as well as their intrinsic ability to constrict in

response to increases in pressure (the myogenic response)123. The resistance arteries

consequently have more vascular smooth muscle cells in their tunica media layer than do the

elastic conduit arteries124;125. A larger smooth muscle layer, and therefore larger population of

VSMCs, would result in a greater force of vasoconstriction. Our results support this assertion

given that the posterior cerebral artery is a 1st order artery and is closer to an elastic conduit

artery than a resistance artery. Consequently, the PCAs demonstrated a reduced magnitude of

contractile response (Emax) to phenylephrine compared to the mesenteric arteries. The sensitivity

(EC50) to phenylephrine was not different between the two arteries suggesting similar α-AR

densities. However, previous experimental studies have demonstrated that receptor density dose

not determine the maximal response to phenylephrine stimulation in VSMCs126. This is in line

with our findings that differential phenylephrine response in mesenteric and cerebral arteries

may be due to structural rather than receptor population differences.

The myogenic response is important in maintaining blood flow and tissue perfusion as

well as protecting the capillaries from large increases in pressure127. In our model, we observed

no change in MAP with metoprolol administration, suggesting that myogenic autoregulation

Page 78: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

70

may not have been a very important factor in our results. However, it is possible that β-blockade

may inhibit this important autoregulatory mechanism which, in the brain, is an integral in

maintaining cerebral blood flow during times of increased demand or reduced supply128. The

importance of PKA (a downstream signalling molecule of β-AR stimulation) in the myogenic

response has not been extensively studied. However one study in isolated rat tail arteries has

reported that a PKA inhibitor attenuates the myogenic response at a transluminal pressure of

80mmHg129. Future work on the effect of β-blockade on not just the autonomic nervous

system’s role in autoregulation but also the myogenic contribution would be necessary to further

understand how β-blockade could affect tissue perfusion.

We found that the sensitivities and magnitudes of response to isoproterenol were

comparable in both mesenteric and posterior cerebral arteries (no differences in EC50 and Emax).

We also demonstrated that metoprolol inhibits dilation of resistance and conduit arteries which

could be relevant in regulating organ perfusion. In unpublished data in our lab, we have

demonstrated that CHF increases the myogenic response in mouse posterior cerebral arteries,

effectively making them important mediators of cerebral perfusion. A β-blocker effect on

conduit arteries could limit blood supply to the resistance arteries and could therefore be

additionally detrimental, especially with the presence of cardiovascular disease. This effect may

explain the reduced incidence of stroke with vasodilator (ACE-I, CCBs) treatment versus β-

blockers in hypertension49. This suggests, along with our findings, that a mechanism of conduit

and resistance artery dysfunction with β-blockade may play an important role in ischemic

injury.

11.5 Limitations

We assessed the impact of metoprolol in a mouse model which has a much higher

intrinsic heart rate than humans. Attempts were made to use a clinically relevant dose of

Page 79: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

71

metoprolol which reduced heart rate by about 20%. This reduction is comparable to the intended

effect size in the clinical setting. Though mouse cardiovascular physiology is not the same as in

humans, our model supports current clinical findings and is therefore valuable in elucidating the

mechanisms of increased morbidity and mortality with β-blockade.

Due to methodological constraints, we were unable to measure MAP and CO in the same

animals. However, in addition to measuring a reduction in CO, our flow-volume loops

demonstrated that left ventricular systolic pressures were maintained following metoprolol

treatment. This is consistent with our MAP data and therefore allowed an accurate measurement

of increased systemic vascular resistance following β-blockade.

Isoflurane anesthesia was used in all whole animal protocols. Isoflurane is a vasodilator

and increases cerebral blood flow when administered130. Isoflurane was maintained at 1.5% in

21% O2 in both control and treatment groups, therefore any differences in CBF and in PBrmvO2

were due to the difference in treatment groups rather than isoflurane.

In our hypoxic challenge experiments, mice were spontaneously breathing and under no

mechanical ventilation. Hypoxia causes hyperventilation due to a reduced arterial PO2 and this

consequently causes a reduction in arterial PCO2. CO2 is an important metabolic vasodilator128,

therefore low arterial PCO2 will limit tissue perfusion. This effect may have been responsible the

observation that there was no appreciable difference in PBrmvO2 between saline and β-blocker

treated mice during hypoxic challenge. We did not measure arterial blood gases and therefore

cannot confirm that there was a reduction in PCO2 in our animals, however this stimulus may

have been great enough to mask the effect of β-blockade on cerebral blood flow during hypoxia.

Mechanically ventilating our mice during hypoxic challenge would have been a possible

solution to this methodological limitation. More importantly, however, our finding that

metoprolol injection significantly lowered resting brain microvascular PO2 would not have been

Page 80: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

72

affected by this limitation and therefore still gave us a strong rationale to examine the effect of

β-blockers on isolated arteries.

Our isolated arteries are denervated and therefore lack the input from autonomic and

other perivascular nerves. All pharmacological agents are applied through the superfusate (ie.

external from the vessel lumen); which mimics prejunctional neurotransmitter release to our

postjunctional receptor targets. Although our agents’ pharmokinetics may differ in vivo than

what we see in vitro, our whole animal data support our findings in isolated arteries and provide

a closer tie to data from clinical studies.

We did not extend our study to determine the functionally predominant β-AR receptor

subtype in our two vessel types. Though we can make inferences based on sensitivities to

isoproterenol and clenbuterol, the assessment of a β1-AR agonist in these tissues would allow a

more complete understanding of the physiology at play. Additionally, assessing the

isoproterenol dose-response curve in the presence of a β2-specific antagonist (such as

butoxamine), would also greatly help the interpretation of our observed results. Our primary

aim, however, was to determine if metoprolol had an effect on mouse vascular tissue, and this

was clearly demonstrated.

The β-AR and α-AR have competitive signalling pathways. This may have been why we

observed a reduced phenylephrine dose-response following an isoproterenol dose-response

curve. However, since endogenous norepinephrine and epinephrine act on both α- and β-ARs in

vivo, our protocol would similarly mimic this dual receptor activation. These data therefore

remain relevant in linking our whole animal findings with those found in vitro.

Finally, we used different order of arteries from gut (3rd) and brain (1st). While the

mesenteric arteries are thought to be true resistance arteries, the cerebral vessels may have more

of a combined conduit/resistance artery function. The resistance arteries ultimately control tissue

Page 81: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

73

perfusion. However, we demonstrated that there is a significant metoprolol effect on larger

arteries which also can control blood flow to the precapillary arteries.

12 Summary

We have provided novel evidence that metoprolol can limit cardiac output and increase

systemic vascular resistance in a mouse model in vivo. This was associated with a reduction in

brain microvascular oxygen tension in anesthetized mice suggesting that basal brain perfusion

can be impaired by metoprolol. Isolated mesenteric and cerebral arteries were utilized to

demonstrate a direct effect of metoprolol to inhibit β-adrenergic mediated vasodilation. These

data support both a cardiac and vascular mechanism for the clinical observation of increased

incidences of organ ischemia and mortality in patients treated with metoprolol.

Page 82: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

74

13 Appendix A – Beta-blockade: A historical perspective

Sir James Black, working at Imperial Chemical Industries (ICI), invented the first

clinically used β-blocker, propranolol, in an era where knowledge of sympathetic nervous

transmission was in its infancy. Walter Cannon and Arturo Rosenblueth were the fathers of the

prevalent “sympathin theory” of adrenergic mediators131. They postulated that two circulating

molecules, sympathins E (excitatory) and I (inhibitory), when combined with epinephrine were

responsible for adrenergic activity in cells131. Though they could not actually prove the

existence of the sympathins in the body, their theory was held as physiologic dogma for almost

a decade until it was challenged by Raymond Ahlquist in 1948.

In an elegant series of experiments, across species and organ types, Ahlquist proposed

that there was only one adrenergic mediator. This was epinephrine (adrenaline), purified in 1901

by a Japanese chemist, Jokichi Takamine, and the first ever hormone isolated132. Ahlquist

proposed that epinephrine, an adrenal extract which strongly affected blood vessels, heart tissue

and skeletal muscle, bound to two different adrenergic receptors expressed on the target tissue.

Ahlquist used 6 different analogues of epinephrine (ethanolamine, isopropanolamine, dl- and l-

epinephrine, methyl isopropanolamine and isopropyl ethanolamine) and determined the relative

activity of each in producing a response in different organ systems from dogs, cats, rabbits and

rats. From the cardiovascular system, Ahlquist determined the order of activities of these amines

on vasoconstriction, vasodilation and myocardial excitation. He also determined how the

intestines, uterus, ureters, dilator papillae and nictitating membranes reacted to his

pharmacological manipulations. Ahlquist found that two different orders of activity emerged

based on both observed function and tissue type examined. For example; vasoconstriction in

renal, mesenteric, femoral and skin blood vessels followed an order of activity of l-epinephrine,

Page 83: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

75

dl-epinephrine, ethanolamine, isopropanolamine, methyl isopropanolamine and isopropyl

ethanolamine133 while vasodilation in skeletal muscle and coronary blood vessels followed an

order of isopropyl ethanolamine, l-epinephrine, methyl isopropanolamine, dl-epinephrine,

isopropanolamine and ethanolamine133. These two orders of activity were consistent across

tissue types leading Ahlquist to arbitrarily designate one order of activity as representative of the

alpha receptor and the other as the beta receptor. His findings are summarized in Table 13.1. It

is now known that the α-receptors are sub classified as α1A, α1B, α1D, α2A, α2B, and α2C and that

the β-receptor has three subtypes; β1, β2, and β3134. For the purposes of this study, only the β1-

and β2-adrenergic receptors will be discussed in detail.

Although Ahlquist’s initial conclusions have survived over a half-century of scientific

scrutiny, his seminal discovery could not dislodge Cannon and Rosenblueth’s entrenched

sympathin theory. As luck would have it, Sir James Black came across Ahlquist’s theory of

adrenergic transmission while reading Drill’s Pharmacology in Medicine which, as he put it,

was “surprising, because at the time [Ahlquist’s] proposal had been published only with great

difficulty – it just wasn’t in the medical dogma.”2 As a clinician scientist, what was most

interesting to Black was Ahlquist’s finding that the β-receptor was responsible for the

adrenergic excitation of the myocardium and how this could apply to the treatment of angina

pectoris. He reasoned that instead of increasing myocardial oxygen supply, as most anti-angina

drugs (vasodilators: nitrates, calcium channel blockers)131 were designed to do at the time, he

could develop a drug that reduced myocardial oxygen demand by acting on Ahlquist’s β-

receptors2. Black and his colleague, chemist John Stephenson, recognized that the well known

β-receptor specific agonist isoproterenol (Table 13.2) has no α-selectivity because the amine

methyl group of epinephrine was modified to the larger isopropyl group2. With this in mind,

Page 84: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

76

Alpha Receptors Beta Receptors Vasoconstriction

Renal Mesenteric Femoral

Vasodilation Coronary Skeletal muscle

Nictitating membrane Myocardium (excitatory) Uterus (excitatory)

Rabbit Dog

Uterus (inhibitory) Rat Cat Dog

Intestine (inhibitory) Dilator papillae

Table 13.1 Original characterization of adrenergic receptor subtypes Modified from Ahlquist, 1948133.

Page 85: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

77

Drug Structure Selectivity

Epinephrine HO

OH

HN

OH

α and β

Isoproterenol HO

OH

HN

OH

β

Dichloroisoproterenol Cl

Cl

HN

OH

β (partial agonist)

Propranolol HN

OH

O β (antagonist)

Table 13.2 Structures of beta-adrenergic receptor ligands Evolution of the chemical modifications to epinephrine eventually leading to the first clinically used β-blocker, propranolol.

Page 86: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

78

and knowing that the ring substituted dichloroisoproterenol was a partial β-agonist, Black and

Stephenson hypothesized that by further altering the ring structure of isoproterenol, they could

completely eliminate any agonist activity2. Propranolol, the world’s first β-blocker was

consequently invented by substituting a benzene group for the vicinal hydroxyl groups of

isoproterenol creating a double ring structure with complete antagonist activity at the β-

adrenergic receptor (Table 13.2). For this and other contributions to drug development, Sir

James Black was awarded the Nobel Prize in Medicine in 1988.

Page 87: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

79

14 Appendix B – Characterization of PCA phenylephrine responses

14.1 Determining optimal phenylephrine preconstriction dose in PCAs

Phenylephrine preconstriction and maintenance doses have not been characterized

previously in the posterior cerebral artery. It was therefore necessary to determine the PE dose

which gives an adequate degree of initial tone while keeping a consistent degree of background

(maintenance) tone through several doses.

14.1.1 Methods

PCAs (n=12) were exposed to increasing concentrations of the α1-adrenergic receptor

agonist phenylephrine (PE; 0.03-30µM) to generate a dose-response curve. In order to match the

degree of preconstriction in MRAs, the concentration of PE which induced 30-50% tone in

PCAs was determined. This concentration (10µM) was found at the upper plateau of the PCA

PE dose-response whereas the MRA preconstriction dose is located along the slope of the MRA

PE dose-response. Therefore, two concentrations of PE were tested as potential preconstriction

and maintenance doses for the isoproterenol dose-response curves; 10µM and 3µM which lies

along the slope of the PCA PE dose-response.

Following a viability check, PCAs were treated with 9 consecutive doses of 10 (n=4) or

3µM (n=3) PE to determine their effectiveness as preconstriction and maintenance doses.

Following this first series, organ baths were exchanged for 50µM metoprolol in MOPS and

incubated for 30 minutes in order to determine the effect of β-blockade on consecutive

applications of these two doses. Each dose of 10 or 3µM PE was held for 2 minutes before

recording the vessel diameter.

Page 88: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

80

14.1.2 Results

The phenylephrine dose-response curve in PCAs did not have significantly different

EC50 than in mesenteric resistance arteries (-6.0 ± 0.2 in MRAs vs -5.9 ± 0.08 in PCAs; n=5,

12). There was, however, a significant difference in the magnitude of response to PE in PCAs;

the degree of tone induced by the EC50 value of PE (1µM or LogEC50=-6.0) was significantly

lower in PCAs compared to MRAs (19 ± 3.0 vs. 37 ± 6.1%; n=12, 5, p<0.05; Figure 14.1.1B)

and the Emax % dilation was also significantly lower in PCAs (45 ± 1.7 vs. 60 ± 2.3; n=12, 5,

p<0.05; Figure 14.1.1C). This made the standard MRA preconstriction and maintenance dose

insufficient for the vasodilatory curves in PCAs.

Two potential concentrations of PE were tested for their ability to be effective

preconstriction and maintenance doses (Figure 14.1.2). Exposing PCAs to 9 consecutive doses

of 10µM PE resulted in a significant reduction in tone by the end of the series compared to the

tone induced by the first dose (from 40 ± 3.3 to 14 ± 6.4%; n=4, p<0.05; Figure 14.1.2A).

Consequently, consecutive doses of 10µM PE resulted in significant dilation of the PCA by the

end of the 9th dose (up to 66 ± 14%; Figure 14.1.2B). However, with 9 consecutive doses of

3µM PE, PCAs were able to maintain their initial degree of tone throughout the series and there

was no significant dilation (n=3; Figure 14.1.2C, D). Additionally, there was no difference in

these described responses at both concentrations PCAs before and after incubation with 50µM

metoprolol (Figure 14.1.2). With these findings, 3µM PE was chosen as the optimal

preconstriction and maintenance dose concentration in PCAs.

Page 89: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

81

-8 -7 -6 -5 -40

20

40

60

log [phenylephrine (mol/L)]

ton

e (%

dia

ma

x 45

mm

Hg

)

-8 -7 -6 -5 -40

20

40

60

log [phenylephrine (mol/L)]

ton

e (%

dia

ma

x 6

0mm

Hg

)A B

MRA PCA0

20

40

60

80

*

PE

Em

ax,

ton

e (%

dia

ma

x)

D

MRA PCA0

20

40

60

80

*

ton

e (%

dia

ma

x)

at P

E E

C5

0(1M

)

C

Figure 14.1.1 Phenylephrine dose-response curves in mesenteric and cerebral arteries. Dose-response curves to phenylephrine in mesenteric resistance (MRA; A) and posterior cerebral arteries (PCA; B). The degree of tone elicited by the EC50 dose of phenylephrine (C) was significantly higher in MRAs than PCAs (37 ± 6.1% vs. 19 ± 3.0%; n=5, 12, p<0.05). Emax values (D) were significantly lower in PCAs than in MRAs (45 ± 1.7 vs 60 ± 2.3; n=12,5, p<0.05) Data presented as mean ± SEM.

Page 90: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

82

0 2 4 6 8 100

20

40

60

80

100 Control, n=4

50M Metoprolol, n=4

* * * * * * *

# of 10M PE Doses

ton

e (%

dia

ma

x 45

mm

Hg

)

0 2 4 6 8 100

20

40

60

80

100 Control, n=4

50M Metoprolol, n=4

** * * * * *

# of 10M PE Doses

% d

ilat

ion

0 2 4 6 8 100

20

40

60

80

100 Control, n=3

50M Metoprolo, n=3

# of 3M PE Doses

ton

e (%

dia

ma

x 4

5mm

Hg

)

0 2 4 6 8 100

20

40

60

80

100 Control, n=3

50uM Metoprolol, n=3

# of 3M PE Doses

% d

ilat

ion

A B

C D

Figure 14.1.2 Effect of consecutive doses of phenylephrine in posterior cerebral arteries. In PCAs, tone is lost (A) and arteries dilate (B) in response to nine consecutive doses of 10µM phenylephrine. There is a significant dose effect, but there is no treatment and no interaction effect (two-way repeated measures ANOVA, p<0.05, n=4). Tone is maintained (C) and vessels do not dilate (D) in response to nine consecutive doses of 3µM phenylephrine. Data presented as mean ± SEM.

Page 91: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

83

14.2 Effect of metoprolol on phenylephrine dose-response curve in PCAs

14.2.1 Results

Phenylephrine dose-response curves (PE DRC) were generated before and after

metoprolol incubation (0µM metoprolol represents a time control; Figure 10.3.1). There was no

difference between phenylephrine LogEC50 values over time in the PCA (Table 14.1). There

was a significant inhibition of the magnitude of the response over time in posterior cerebral

arteries(Figure 14.2.1A, D). The tone generated at the EC50 dose of PE (1µM or logEC50=-6)

was not significantly altered by time Figure 14.2.1C. However, the tone at Emax (ie. at the top of

the PE DRC best-fit curve) was significantly inhibited by time (43 ± 3.0 in the first curve vs. 35

± 4 % tone in the second, 0µM metoprolol curve; n=5, p<0.05; Figure 14.2.1D). The same

relationship was found with application of 50µM metoprolol; no affect on LogEC50 value or on

the tone generated at this value (Table 14.1; Figure 14.2.1C) but a significant inhibition of the

degree of tone at Emax (47 ± 1.9 vs. 25 ± 3.9 %; n=7, p<0.05; Figure 14.2.1B, D). When

comparing the time matched PE DRC (ie. 0µM metoprolol) vs. 50µM metoprolol, there was no

significant difference in LogEC50 value, the tone at this concentration or in the degree of tone at

Emax(Figure 14.2.1).

Page 92: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

84

-8 -7 -6 -5 -40

20

40

60 Control, n=5

0M Metoprolol, n=5

* *

log [phenylephrine (mol/L)]

ton

e, %

dia

ma

x 4

5mm

Hg

-8 -7 -6 -5 -40

20

40

60 Control, n=7

50M Metoprolol, n=7

** *

log [phenylephrine (mol/L)]

ton

e, %

dia

ma

x 4

5mm

Hg

M M

et

- 0

M M

et

+

0M

Met

- 5

0M

Met

+

50

0

20

40

60

EC

50

(1M

)to

ne,

% d

iam

ax 4

5mm

Hg

M M

et

- 0

M M

et

+

0M

Met

- 5

0M

Met

+

50

0

20

40

60

Em

ax

ton

e,%

dia

ma

x 4

5mm

Hg

A B

C D

Figure 14.2.1 Effect of metoprolol on phenylephrine dose-response in PCAs. Phenylephrine (PE) dose-response curves were generated before (control) and after incubation with metoprolol in posterior cerebral arteries. PE mediated increases in tone at high doses are blunted with time (represented by 0µM metoprolol; panel A) and at 50µM metoprolol (panel B). The PE curve at 50µM metoprolol is not significantly different from its time matched control (panel C and D). Emax values (D) are significantly reduced by time (43 ± 3.0 vs. 35 ± 4% tone; n=5, p<0.05) and at 50µM metoprolol (47 ± 1.9 vs. 25 ± 3.9% tone; n=7, p<0.05). Data presented as mean ± SEM. * p<0.05 by two-way repeated measures ANOVA; ɣ p<0.05 by paired t-test; # p<0.05 by unpaired t-test.

Page 93: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

85

Control (-Met) + Metoprolol

± 0µM Metoprolol (n=5) -5.8 ± 0.1 -6.0 ± 0.2

± 50µM Metoprolol (n=7) -5.9 ± 0.1 -6.2 ± 0.2 Table 14.1 Half maximal concentrations of phenylephrine in PCAs. LogEC50 values for PE dose-response curves in PCAs. Data presented as mean ± SEM. *p<0.05 between columns (paired t-test); # p<0.05 within columns (unpaired-test).

Page 94: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

86

14.2.2 Interpretation: Phenylephrine responses are time-dependent in the PCA

The control phenylephrine dose-response (ie. the first dose-response in the protocol) is in

agreement with a previous study in mouse middle cerebral arteries (MCA)135. It is likely that

these vessels responded similarly to phenylephrine because of their proximity to the circle of

Willis as well as their 1st to 2nd order of branching.

There was a significant time-dependent inhibition of the magnitude of the phenylephrine

response in PCAs. In order to overcome this phenomenon, our analyses in PCAs were compared

to internal controls (ie. the same vessel) and to time-matched controls. Time-matched analyses

showed that the phenylephrine dose-response in the posterior cerebral artery was not

significantly affected by metoprolol.

The time-dependent decay of the phenylephrine dose-response may be due to a

desensitization of the α1-AR over the course of the experimental protocol. A study in rat

thoracic aorta has shown that prolonged exposure to phenylephrine reduces the responsiveness

to α1-AR mediated vasoconstriction136. This effect, however, was not due to receptor down-

regulation, but rather by an up-regulation of nitric oxide synthase (NOS) and consequently nitric

oxide (NO). As a potent vasodilator, NO could antagonize the vasoconstriction induced by

phenylephrine in the second dose-response curve (the time controlled curve).

It is also possible, considering the full protocol in PCAs (phenylephrine dose-response;

isoproterenol dose-response; phenylephrine dose-response with/without metoprolol;

isoproterenol dose-response with/without metoprolol), that the first isoproterenol curve may

influence the contractile response of the second phenylephrine dose-response curve. In addition

to a possible phenylephrine-induced increase in NOS, there may also be an isoproterenol-

induced increase in NOS and therefore NO. There is evidence that isoproterenol-mediated

vasodilation has a nitric oxide component, arising from attenuated isoproterenol responses in

Page 95: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

87

either endothelium denuded/L-NAME (NOS inhibitor) treated or eNOS-KO vessels84-86.

Furthermore, it has been demonstrated that isoproterenol has a dose-dependent inhibitory effect

on the phenylephrine dose-response curve in isolated rabbit common carotid arteries137. This

could likely suggest that with increasing doses of isoproterenol, there is an increase in NO. If it

is not fully removed when changing the bath, the NO could subsequently affect the

responsiveness of phenylephrine in the posterior cerebral arteries.

In our isoproterenol dose-response curves, there was no difference in the magnitude of

tone induced by our preconstriction dose (3µM) of phenylephrine before and after metoprolol.

The time dependent decay described above only affected higher concentrations of the

phenylephrine dose-response curve. This could explain why consecutive doses of 10µM

phenylephrine showed a slow reduction in tone (increased dilation) in PCAs. The mechanism

may be α1-AR mediated increases in NO, which would consequently reduce tone and promote

vasodilation 136. This effect was observed in our vessels and could contribute to reduced

responsiveness when high doses of phenylephrine are applied over a long period of time.

Conversely, consecutive doses of 3µM phenylephrine did maintain tone in our PCAs. For this

reason, this concentration of phenylephrine was chosen as our preconstriction and maintenance

dose for the isoproterenol dose-response curves.

Page 96: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

88

15 Reference List

1. Frishman WH: beta-Adrenergic blockers: a 50-year historical perspective. Am.J.Ther. 2008; 15: 565-76

2. Black J: Sir James Black: learning by doing. Interview. Mol.Interv. 2004; 4: 139-42

3. Pulido J, Kor D: Beta-Adrenergic Receptor Antagonists, Advances in Cardiovascular Pharmacology. Edited by Housmans P, Nuttall G. USA, Wolters Kluwer Health, Lippincott Williams and Wilkins, 2008, pp 43-66

4. Baker JG: The selectivity of beta-adrenoceptor antagonists at the human beta1, beta2 and beta3 adrenoceptors. Br.J.Pharmacol. 2005; 144: 317-22

5. SRIVASTAVA SC, DEWAR HA, NEWELL DJ: Double-blind trial of propranoll (Inderal) in angina of effort. Br.Med.J. 1964; 19;2: 724-5

6. KEELAN P: Double-blind trial of propranolol (Inderal) in angina pectoris. Br.Med.J. 1965; 1: 897-8

7. Pepine CJ, Cohn PF, Deedwania PC, Gibson RS, Handberg E, Hill JA, Miller E, Marks RG, Thadani U: Effects of treatment on outcome in mildly symptomatic patients with ischemia during daily life. The Atenolol Silent Ischemia Study (ASIST). Circulation. 1994; 90: 762-8

8. Dargie HJ, Ford I, Fox KM: Total Ischaemic Burden European Trial (TIBET). Effects of ischaemia and treatment with atenolol, nifedipine SR and their combination on outcome in patients with chronic stable angina. The TIBET Study Group. Eur.Heart J. 1996; 17: 104-12

9. Rehnqvist N, Hjemdahl P, Billing E, Bjorkander I, Eriksson SV, Forslund L, Held C, Nasman P, Wallen NH: Effects of metoprolol vs verapamil in patients with stable angina pectoris. The Angina Prognosis Study in Stockholm (APSIS). Eur.Heart J. 1996; 17: 76-81

10. Antman EM, Hand M, Armstrong PW, Bates ER, Green LA, Halasyamani LK, Hochman JS, Krumholz HM, Lamas GA, Mullany CJ, Pearle DL, Sloan MA, Smith SC, Jr., Anbe DT, Kushner FG, Ornato JP, Pearle DL, Sloan MA, Jacobs AK, Adams CD, Anderson JL, Buller CE, Creager MA, Ettinger SM, Halperin JL, Hunt SA, Lytle BW, Nishimura R, Page RL, Riegel B, Tarkington LG, Yancy CW: 2007 focused update of the ACC/AHA 2004 guidelines for the management of patients with ST-elevation myocardial infarction: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J.Am.Coll.Cardiol. 2008; 51: 210-47

11. Dickstein K, Cohen-Solal A, Filippatos G, McMurray JJ, Ponikowski P, Poole-Wilson PA, Stromberg A, van Veldhuisen DJ, Atar D, Hoes AW, Keren A, Mebazaa A,

Page 97: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

89

Nieminen M, Priori SG, Swedberg K, Vahanian A, Camm J, De CR, Dean V, Dickstein K, Filippatos G, Funck-Brentano C, Hellemans I, Kristensen SD, McGregor K, Sechtem U, Silber S, Tendera M, Widimsky P, Zamorano JL: ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2008: the Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2008 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association of the ESC (HFA) and endorsed by the European Society of Intensive Care Medicine (ESICM). Eur.Heart J. 2008; 29: 2388-442

12. Hunt SA, Abraham WT, Chin MH, Feldman AM, Francis GS, Ganiats TG, Jessup M, Konstam MA, Mancini DM, Michl K, Oates JA, Rahko PS, Silver MA, Stevenson LW, Yancy CW, Antman EM, Smith SC, Jr., Adams CD, Anderson JL, Faxon DP, Fuster V, Halperin JL, Hiratzka LF, Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B: ACC/AHA 2005 Guideline Update for the Diagnosis and Management of Chronic Heart Failure in the Adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure): developed in collaboration with the American College of Chest Physicians and the International Society for Heart and Lung Transplantation: endorsed by the Heart Rhythm Society. Circulation. 2005; %20;112: e154-e235

13. Fox K, Garcia MA, Ardissino D, Buszman P, Camici PG, Crea F, Daly C, De BG, Hjemdahl P, Lopez-Sendon J, Marco J, Morais J, Pepper J, Sechtem U, Simoons M, Thygesen K, Priori SG, Blanc JJ, Budaj A, Camm J, Dean V, Deckers J, Dickstein K, Lekakis J, McGregor K, Metra M, Morais J, Osterspey A, Tamargo J, Zamorano JL: Guidelines on the management of stable angina pectoris: executive summary: The Task Force on the Management of Stable Angina Pectoris of the European Society of Cardiology. Eur.Heart J. 2006; 27: 1341-81

14. Fraker TD, Jr., Fihn SD, Gibbons RJ, Abrams J, Chatterjee K, Daley J, Deedwania PC, Douglas JS, Ferguson TB, Jr., Gardin JM, O'Rourke RA, Williams SV, Smith SC, Jr., Jacobs AK, Adams CD, Anderson JL, Buller CE, Creager MA, Ettinger SM, Halperin JL, Hunt SA, Krumholz HM, Kushner FG, Lytle BW, Nishimura R, Page RL, Riegel B, Tarkington LG, Yancy CW: 2007 chronic angina focused update of the ACC/AHA 2002 guidelines for the management of patients with chronic stable angina: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines Writing Group to develop the focused update of the 2002 guidelines for the management of patients with chronic stable angina. J.Am.Coll.Cardiol. 2007; 50: 2264-74

15. Miura M, Thomas R, Ganz W, Sokol T, Shell WE, Toshimitsu T, Kwan AC, Singh BN: The effect of delay in propranolol administration on reduction of myocardial infarct size after experimental coronary artery occlusion in dogs. Circulation. 1979; 59: 1148-57

16. Yusuf S, Ramsdale D, Peto R, Furse L, Bennett D, Bray C, Sleight P: Early intravenous atenolol treatment in suspected acute myocardial infarction. Preliminary report of a randomised trial. Lancet. 1980; 2: 273-6

Page 98: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

90

17. Hjalmarson A, Elmfeldt D, Herlitz J, Holmberg S, Malek I, Nyberg G, Ryden L, Swedberg K, Vedin A, Waagstein F, Waldenstrom A, Waldenstrom J, Wedel H, Wilhelmsen L, Wilhelmsson C: Effect on mortality of metoprolol in acute myocardial infarction. A double-blind randomised trial. Lancet. 1981; 2: 823-7

18. The Norwegian Multicenter Study Group: Timolol-induced reduction in mortality and reinfarction in patients surviving acute myocardial infarction. N.Engl.J.Med. 1981; 304: 801-7

19. Beta-Blocker Heart Attack Trial Research Group: A randomized trial of propranolol in patients with acute myocardial infarction. I. Mortality results. JAMA. 1982; 247: 1707-14

20. Beta-Blocker Heart Attack Trial Research Group: A randomized trial of propranolol in patients with acute myocardial infarction. II. Morbidity results. JAMA. 1983; 250: 2814-9

21. The MIAMI Trial Research Group: Metoprolol in acute myocardial infarction (MIAMI). A randomised placebo-controlled international trial. The MIAMI Trial Research Group. Eur.Heart J. 1985; 6: 199-226

22. ISIS-1 Collaborative Group: Randomised trial of intravenous atenolol among 16 027 cases of suspected acute myocardial infarction: ISIS-1. First International Study of Infarct Survival Collaborative Group. Lancet. 1986; 2: 57-66

23. Roberts R, Rogers WJ, Mueller HS, Lambrew CT, Diver DJ, Smith HC, Willerson JT, Knatterud GL, Forman S, Passamani E, .: Immediate versus deferred beta-blockade following thrombolytic therapy in patients with acute myocardial infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI) II-B Study. Circulation. 1991; 83: 422-37

24. Chen ZM, Pan HC, Chen YP, Peto R, Collins R, Jiang LX, Xie JX, Liu LS: Early intravenous then oral metoprolol in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet. 2005; 366: 1622-32

25. Triposkiadis F, Karayannis G, Giamouzis G, Skoularigis J, Louridas G, Butler J: The sympathetic nervous system in heart failure physiology, pathophysiology, and clinical implications. J.Am.Coll.Cardiol. 2009; 54: 1747-62

26. Packer M: Evolution of the neurohormonal hypothesis to explain the progression of chronic heart failure. Eur.Heart J. 1995; 16 Suppl F:4-6.: 4-6

27. Packer M, Bristow MR, Cohn JN, Colucci WS, Fowler MB, Gilbert EM, Shusterman NH: The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group. N.Engl.J.Med. 1996; 334: 1349-55

28. Packer M, Coats AJ, Fowler MB, Katus HA, Krum H, Mohacsi P, Rouleau JL, Tendera M, Castaigne A, Roecker EB, Schultz MK, DeMets DL: Effect of carvedilol on survival in severe chronic heart failure. N.Engl.J.Med. 2001; 344: 1651-8

Page 99: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

91

29. CIBIS-II Investigators and Committees: The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet. 1999; 353: 9-13

30. MERIT-HF Study Group: Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet. 1999; 353: 2001-7

31. The IPPPSH Collaborative Group: Cardiovascular risk and risk factors in a randomized trial of treatment based on the beta-blocker oxprenolol: the International Prospective Primary Prevention Study in Hypertension (IPPPSH). The IPPPSH Collaborative Group. J.Hypertens. 1985; 3: 379-92

32. Medical Research Council Working Party: MRC trial of treatment of mild hypertension: principal results. Medical Research Council Working Party. Br.Med.J.(Clin.Res.Ed). 1985; 291: 97-104

33. Coope J, Warrender TS: Randomised trial of treatment of hypertension in elderly patients in primary care. Br.Med.J.(Clin.Res.Ed). 1986; 293: 1145-51

34. Medical Research Council Working Party: Medical Research Council trial of treatment of hypertension in older adults: principal results. MRC Working Party. BMJ. 1992; 304: 405-12

35. Bradley HA, Wiysonge CS, Volmink JA, Mayosi BM, Opie LH: How strong is the evidence for use of beta-blockers as first-line therapy for hypertension? Systematic review and meta-analysis. J.Hypertens. 2006; 24: 2131-41

36. Mansia G, De BG, Dominiczak A, Cifkova R, Fagard R, Germano G, Grassi G, Heagerty AM, Kjeldsen SE, Laurent S, Narkiewicz K, Ruilope L, Rynkiewicz A, Schmieder RE, Struijker Boudier HA, Zanchetti A: 2007 ESH-ESC Guidelines for the management of arterial hypertension: the task force for the management of arterial hypertension of the European Society of Hypertension (ESH) and of the European Society of Cardiology (ESC). Blood Press. 2007; 16: 135-232

37. Mangano DT, Layug EL, Wallace A, Tateo I: Effect of atenolol on mortality and cardiovascular morbidity after noncardiac surgery. Multicenter Study of Perioperative Ischemia Research Group. N.Engl.J.Med. 1996; 335: 1713-20

38. Poldermans D, Boersma E, Bax JJ, Thomson IR, van de Ven LL, Blankensteijn JD, Baars HF, Yo TI, Trocino G, Vigna C, Roelandt JR, van UH: The effect of bisoprolol on perioperative mortality and myocardial infarction in high-risk patients undergoing vascular surgery. Dutch Echocardiographic Cardiac Risk Evaluation Applying Stress Echocardiography Study Group. N.Engl.J.Med. 1999; 341: 1789-94

39. Poldermans D, Boersma E, Bax JJ, Thomson IR, Paelinck B, van de Ven LL, Scheffer MG, Trocino G, Vigna C, Baars HF, van UH, Roelandt JR: Bisoprolol reduces cardiac death and myocardial infarction in high-risk patients as long as 2 years after successful major vascular surgery. Eur.Heart J. 2001; 22: 1353-8

Page 100: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

92

40. Poldermans D, Bax JJ, Boersma E, De HS, Eeckhout E, Fowkes G, Gorenek B, Hennerici MG, Iung B, Kelm M, Kjeldsen KP, Kristensen SD, Lopez-Sendon J, Pelosi P, Philippe F, Pierard L, Ponikowski P, Schmid JP, Sellevold OF, Sicari R, Van den Berghe G, Vermassen F, Hoeks SE, Vanhorebeek I, Vahanian A, Auricchio A, Bax JJ, Ceconi C, Dean V, Filippatos G, Funck-Brentano C, Hobbs R, Kearn P, McDonag T, McGregor K, Popescu BA, Reiner Z, Sechtem U, Sirnes PA, Tendera M, Vardas P, Widimsky P, De CR, Agewall S, Al AN, Andreotti F, Anker SD, Baron-Esquivias G, Berkenboom G, Chapoutot L, Cifkova R, Faggiano P, Gibbs S, Hansen HS, Iserin L, Israel CW, Kornowski R, Eizagaechevarria NM, Pepi M, Piepoli M, Priebe HJ, Scherer M, Stepinska J, Taggart D, Tubaro M: Guidelines for pre-operative cardiac risk assessment and perioperative cardiac management in non-cardiac surgery: the Task Force for Preoperative Cardiac Risk Assessment and Perioperative Cardiac Management in Non-cardiac Surgery of the European Society of Cardiology (ESC) and endorsed by the European Society of Anaesthesiology (ESA). Eur.J.Anaesthesiol. 2010; 27: 92-137

41. Brady AR, Gibbs JS, Greenhalgh RM, Powell JT, Sydes MR: Perioperative beta-blockade (POBBLE) for patients undergoing infrarenal vascular surgery: results of a randomized double-blind controlled trial. J.Vasc.Surg. 2005; 41: 602-9

42. Yang H, Raymer K, Butler R, Parlow J, Roberts R: The effects of perioperative beta-blockade: results of the Metoprolol after Vascular Surgery (MaVS) study, a randomized controlled trial. Am.Heart J. 2006; 152: 983-90

43. Juul AB, Wetterslev J, Gluud C, Kofoed-Enevoldsen A, Jensen G, Callesen T, Norgaard P, Fruergaard K, Bestle M, Vedelsdal R, Miran A, Jacobsen J, Roed J, Mortensen MB, Jorgensen L, Jorgensen J, Rovsing ML, Petersen PL, Pott F, Haas M, Albret R, Nielsen LL, Johansson G, Stjernholm P, Molgaard Y, Foss NB, Elkjaer J, Dehlie B, Boysen K, Zaric D, Munksgaard A, Madsen JB, Oberg B, Khanykin B, Blemmer T, Yndgaard S, Perko G, Wang LP, Winkel P, Hilden J, Jensen P, Salas N: Effect of perioperative beta blockade in patients with diabetes undergoing major non-cardiac surgery: randomised placebo controlled, blinded multicentre trial. BMJ. 2006; 332: 1482

44. Zaugg M, Bestmann L, Wacker J, Lucchinetti E, Boltres A, Schulz C, Hersberger M, Kalin G, Furrer L, Hofer C, Blumenthal S, Muller A, Zollinger A, Spahn DR, Borgeat A: Adrenergic receptor genotype but not perioperative bisoprolol therapy may determine cardiovascular outcome in at-risk patients undergoing surgery with spinal block: the Swiss Beta Blocker in Spinal Anesthesia (BBSA) study: a double-blinded, placebo-controlled, multicenter trial with 1-year follow-up. Anesthesiology. 2007; 107: 33-44

45. Devereaux PJ, Yang H, Yusuf S, Guyatt G, Leslie K, Villar JC, Xavier D, Chrolavicius S, Greenspan L, Pogue J, Pais P, Liu L, Xu S, Malaga G, Avezum A, Chan M, Montori VM, Jacka M, Choi P: Effects of extended-release metoprolol succinate in patients undergoing non-cardiac surgery (POISE trial): a randomised controlled trial. Lancet. 2008; 371: 1839-47

46. Dunkelgrun M, Boersma E, Schouten O, Koopman-van Gemert AW, van PF, Bax JJ, Thomson IR, Poldermans D: Bisoprolol and fluvastatin for the reduction of perioperative cardiac mortality and myocardial infarction in intermediate-risk patients undergoing

Page 101: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

93

noncardiovascular surgery: a randomized controlled trial (DECREASE-IV). Ann.Surg. 2009; 249: 921-6

47. Bangalore S, Wetterslev J, Pranesh S, Sawhney S, Gluud C, Messerli FH: Perioperative beta blockers in patients having non-cardiac surgery: a meta-analysis. Lancet. 2008; 372: 1962-76

48. Lindenauer PK, Pekow P, Wang K, Mamidi DK, Gutierrez B, Benjamin EM: Perioperative beta-blocker therapy and mortality after major noncardiac surgery. N.Engl.J.Med. 2005; 353: 349-61

49. Wiysonge CS, Bradley H, Mayosi BM, Maroney R, Mbewu A, Opie LH, Volmink J: Beta-blockers for hypertension. Cochrane.Database.Syst.Rev. 2007; CD002003

50. Hansson L, Hedner T, Lund-Johansen P, Kjeldsen SE, Lindholm LH, Syvertsen JO, Lanke J, de FU, Dahlof B, Karlberg BE: Randomised trial of effects of calcium antagonists compared with diuretics and beta-blockers on cardiovascular morbidity and mortality in hypertension: the Nordic Diltiazem (NORDIL) study. Lancet. 2000; 356: 359-65

51. Dahlof B, Sever PS, Poulter NR, Wedel H, Beevers DG, Caulfield M, Collins R, Kjeldsen SE, Kristinsson A, McInnes GT, Mehlsen J, Nieminen M, O'Brien E, Ostergren J: Prevention of cardiovascular events with an antihypertensive regimen of amlodipine adding perindopril as required versus atenolol adding bendroflumethiazide as required, in the Anglo-Scandinavian Cardiac Outcomes Trial-Blood Pressure Lowering Arm (ASCOT-BPLA): a multicentre randomised controlled trial. Lancet. 2005; 366: 895-906

52. Zanchetti A, Bond MG, Hennig M, Neiss A, Mancia G, Dal PC, Hansson L, Magnani B, Rahn KH, Reid JL, Rodicio J, Safar M, Eckes L, Rizzini P: Calcium antagonist lacidipine slows down progression of asymptomatic carotid atherosclerosis: principal results of the European Lacidipine Study on Atherosclerosis (ELSA), a randomized, double-blind, long-term trial. Circulation. 2002; 106: 2422-7

53. Pepine CJ, Handberg EM, Cooper-DeHoff RM, Marks RG, Kowey P, Messerli FH, Mancia G, Cangiano JL, Garcia-Barreto D, Keltai M, Erdine S, Bristol HA, Kolb HR, Bakris GL, Cohen JD, Parmley WW: A calcium antagonist vs a non-calcium antagonist hypertension treatment strategy for patients with coronary artery disease. The International Verapamil-Trandolapril Study (INVEST): a randomized controlled trial. JAMA. 2003; 290: 2805-16

54. Sica DA: Cardiovascular morbidity and mortality in patients with diabetes in the Losartan Intervention For Endpoint reduction in hypertension study (LIFE): a randomised trial against atenolol. Curr.Hypertens.Rep. 2002; 4: 321-3

55. UK Prospective Diabetes Study Group: Efficacy of atenolol and captopril in reducing risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 39. UK Prospective Diabetes Study Group. BMJ. 1998; 317: 713-20

Page 102: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

94

56. Badgett RG, Lawrence VA, Cohn SL: Variations in Pharmacology of beta-Blockers May Contribute to Heterogeneous Results in Trials of Perioperative beta-Blockade. Anesthesiology. 2010;

57. Lands AM, Luduena FP, Buzzo HJ: Differentiation of receptors responsive to isoproterenol. Life Sci. 1967; 6: 2241-9

58. Lands AM, Arnold A, McAuliff JP, Luduena FP, Brown TG, Jr.: Differentiation of receptor systems activated by sympathomimetic amines. Nature. 1967; 214: 597-8

59. Saucerman JJ, McCulloch AD: Cardiac beta-adrenergic signaling: from subcellular microdomains to heart failure. Ann.N.Y.Acad.Sci. 2006; 1080:348-61.: 348-61

60. Tanaka Y, Horinouchi T, Koike K: New insights into beta-adrenoceptors in smooth muscle: distribution of receptor subtypes and molecular mechanisms triggering muscle relaxation. Clin.Exp.Pharmacol.Physiol. 2005; 32: 503-14

61. Brodde OE, Michel MC: Adrenergic and muscarinic receptors in the human heart. Pharmacol.Rev. 1999; 51: 651-90

62. Noble A, Thomas A, Johnson R, Bass P: Regulation of Cardiac Function, The Cardiovascular System: Basic Science and Clinical Conditions, 2 edition. Edited by Horne T, Stader L. Toronto, Churchill Livingstone, 2010, pp 39-48

63. DiFrancesco D: The role of the funny current in pacemaker activity. Circ.Res. 2010; %19;106: 434-46

64. Noble A, Thomas A, Johnson R, Bass P: Cardiac Muscle Structure and Function, The Cardiovascular System: Basic Science and Clinical Conditions, 2 edition. Edited by Horne T, Stader L. Toronto, Churchill Livingstone, 2010, pp 17-26

65. Brown HF, DiFrancesco D, Noble SJ: How does adrenaline accelerate the heart? Nature. 1979; %19;280: 235-6

66. DiFrancesco D: Characterization of single pacemaker channels in cardiac sino-atrial node cells. Nature. 1986; 324: 470-3

67. DiFrancesco D, Tortora P: Direct activation of cardiac pacemaker channels by intracellular cyclic AMP. Nature. 1991; 351: 145-7

68. Kamp TJ, Hell JW: Regulation of cardiac L-type calcium channels by protein kinase A and protein kinase C. Circ.Res. 2000; 87: 1095-102

69. Hulme JT, Westenbroek RE, Scheuer T, Catterall WA: Phosphorylation of serine 1928 in the distal C-terminal domain of cardiac CaV1.2 channels during beta1-adrenergic regulation. Proc.Natl.Acad.Sci.U.S.A. 2006; 103: 16574-9

70. Hulme JT, Lin TW, Westenbroek RE, Scheuer T, Catterall WA: Beta-adrenergic regulation requires direct anchoring of PKA to cardiac CaV1.2 channels via a leucine

Page 103: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

95

zipper interaction with A kinase-anchoring protein 15. Proc.Natl.Acad.Sci.U.S.A. 2003; 100: 13093-8

71. Zhou P, Zhao YT, Guo YB, Xu SM, Bai SH, Lakatta EG, Cheng H, Hao XM, Wang SQ: Beta-adrenergic signaling accelerates and synchronizes cardiac ryanodine receptor response to a single L-type Ca2+ channel. Proc.Natl.Acad.Sci.U.S.A. 2009; %20;106: 18028-33

72. Colyer J: Phosphorylation states of phospholamban. Ann.N.Y.Acad.Sci. 1998; 853:79-91.: 79-91

73. Zhong J, Hume JR, Keef KD: beta-Adrenergic receptor stimulation of L-type Ca2+ channels in rabbit portal vein myocytes involves both alphas and betagamma G protein subunits. J.Physiol. 2001; 531: 105-15

74. Scornik FS, Codina J, Birnbaumer L, Toro L: Modulation of coronary smooth muscle KCa channels by Gs alpha independent of phosphorylation by protein kinase A. Am.J.Physiol. 1993; 265: H1460-H1465

75. Hill MA, Zou H, Potocnik SJ, Meininger GA, Davis MJ: Invited review: arteriolar smooth muscle mechanotransduction: Ca(2+) signaling pathways underlying myogenic reactivity. J.Appl.Physiol. 2001; 91: 973-83

76. Yang Y, Shi Y, Guo S, Zhang S, Cui N, Shi W, Zhu D, Jiang C: PKA-dependent activation of the vascular smooth muscle isoform of KATP channels by vasoactive intestinal polypeptide and its effect on relaxation of the mesenteric resistance artery. Biochim.Biophys.Acta. 2008; 1778: 88-96

77. Wellman GC, Santana LF, Bonev AD, Nelson MT: Role of phospholamban in the modulation of arterial Ca(2+) sparks and Ca(2+)-activated K(+) channels by cAMP. Am.J.Physiol Cell Physiol. 2001; 281: C1029-C1037

78. Somlyo AP, Somlyo AV: Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev. 2003; 83: 1325-58

79. Raina H, Zacharia J, Li M, Wier WG: Activation by Ca2+/calmodulin of an exogenous myosin light chain kinase in mouse arteries. J.Physiol. 2009; 587: 2599-612

80. Adelstein RS, Conti MA, Pato MD: Regulation of myosin light chain kinase by reversible phosphorylation and calcium-calmodulin. Ann.N.Y.Acad.Sci. 1980; 356:142-50.: 142-50

81. Wooldridge AA, MacDonald JA, Erdodi F, Ma C, Borman MA, Hartshorne DJ, Haystead TA: Smooth muscle phosphatase is regulated in vivo by exclusion of phosphorylation of threonine 696 of MYPT1 by phosphorylation of Serine 695 in response to cyclic nucleotides. J.Biol.Chem. 2004; 279: 34496-504

Page 104: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

96

82. Lubomirov LT, Schubert R, Gagov HS, Duridanova DB, Pfitzer G: [Urocortin decreases phosphorylation of MYPT1 and increases the myosin phosphatase activity via elevation of the intracellular level of cAMP]. Biofizika. 2006; 51: 773-80

83. Azam MA, Yoshioka K, Ohkura S, Takuwa N, Sugimoto N, Sato K, Takuwa Y: Ca2+-independent, inhibitory effects of cyclic adenosine 5'-monophosphate on Ca2+ regulation of phosphoinositide 3-kinase C2alpha, Rho, and myosin phosphatase in vascular smooth muscle. J.Pharmacol.Exp.Ther. 2007; 320: 907-16

84. Al ZK, Bexis S, Docherty JR: Relaxations to beta-adrenoceptor subtype selective agonists in wild-type and NOS-3-KO mouse mesenteric arteries. Eur.J.Pharmacol. 2008; 587: 216-23

85. Hempelmann RG, Ziegler A: Endothelium-dependent noradrenaline-induced relaxation of rat isolated cerebral arteries: pharmacological characterization of receptor subtypes involved. Br.J.Pharmacol. 1993; 110: 1321-8

86. Ferro A, Coash M, Yamamoto T, Rob J, Ji Y, Queen L: Nitric oxide-dependent beta2-adrenergic dilatation of rat aorta is mediated through activation of both protein kinase A and Akt. Br.J.Pharmacol. 2004; 143: 397-403

87. Guimaraes S, Moura D: Vascular adrenoceptors: an update. Pharmacol.Rev. 2001; 53: 319-56

88. Ayajiki K, Toda N: Isolated bovine cerebral arteries from rostral and caudal regions: distinct responses to adrenoceptor agonists. Eur.J.Pharmacol. 1990; 191: 417-25

89. Ayajiki K, Toda N: Regional difference in the response mediated by beta 1-adrenoceptor subtype in bovine cerebral arteries. J.Cereb.Blood Flow Metab. 1992; 12: 507-13

90. Tsukahara T, Taniguchi T, Shimohama S, Fujiwara M, Handa H: Characterization of beta adrenergic receptors in human cerebral arteries and alteration of the receptors after subarachnoid hemorrhage. Stroke. 1986; 17: 202-7

91. Nakai K, Itakura T, Naka Y, Nakakita K, Kamei I, Imai H, Yokote H, Komai N: The distribution of adrenergic receptors in cerebral blood vessels: an autoradiographic study. Brain Res. 1986; 381: 148-52

92. Edvinsson L, Owman C, Sjoberg NO: Autonomic nerves, mast cells, and amine receptors in human brain vessels. A histochemical and pharmacological study. Brain Res. 1976; 115: 377-93

93. Miyamoto A, Ito K, Nishio A: Characterization of beta-adrenoceptors in pig basilar artery from functional and radioligand binding studies. Jpn.J.Pharmacol. 1993; 61: 93-9

94. Edvinsson L, Owman C: Pharmacological characterization of adrenergic alpha and beta receptors mediating the vasomotor responses of cerebral arteries in vitro. Circ.Res. 1974; 35: 835-49

Page 105: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

97

95. Gorman MW, Tune JD, Richmond KN, Feigl EO: Feedforward sympathetic coronary vasodilation in exercising dogs. J.Appl.Physiol. 2000; 89: 1892-902

96. Gao F, de Beer VJ, Hoekstra M, Xiao C, Duncker DJ, Merkus D: Both beta1- and beta2-adrenoceptors contribute to feedforward coronary resistance vessel dilation during exercise. Am.J.Physiol Heart Circ.Physiol. 2010; 298: H921-H929

97. Toda N, Okamura T: Beta adrenoceptor subtype in isolated human, monkey and dog epicardial coronary arteries. J.Pharmacol.Exp.Ther. 1990; 253: 518-24

98. Baron GD, Speden RN, Bohr DF: Beta-adrenergic receptors in coronary and skeletal muscle arteries. Am.J.Physiol. 1972; 223: 878-81

99. Yamada S, Kashiwabara T, Yamazawa T, Harada Y, Nakayama K: Demonstration of beta 1-adrenoceptor mediating relaxation of porcine coronary artery by radioligand binding and pharmacological methods. Life Sci. 1988; 43: 1999-2006

100. Nyborg NC, Mikkelsen EO: Characterization of beta-adrenoceptor subtype in isolated ring preparations of intramural rat coronary small arteries. J.Cardiovasc.Pharmacol. 1985; 7: 1113-7

101. Lui AH, McManus BM, Laher I: Endothelial and myogenic regulation of coronary artery tone in the mouse. Eur.J.Pharmacol. 2000; %20;410: 25-31

102. Martin WH, III, Murphree SS, Saffitz JE: Beta-adrenergic receptor distribution among muscle fiber types and resistance arterioles of white, red, and intermediate skeletal muscle. Circ.Res. 1989; 64: 1096-105

103. Chruscinski A, Brede ME, Meinel L, Lohse MJ, Kobilka BK, Hein L: Differential distribution of beta-adrenergic receptor subtypes in blood vessels of knockout mice lacking beta(1)- or beta(2)-adrenergic receptors. Mol.Pharmacol. 2001; 60: 955-62

104. Poole-Wilson PA, Swedberg K, Cleland JG, Di LA, Hanrath P, Komajda M, Lubsen J, Lutiger B, Metra M, Remme WJ, Torp-Pedersen C, Scherhag A, Skene A: Comparison of carvedilol and metoprolol on clinical outcomes in patients with chronic heart failure in the Carvedilol Or Metoprolol European Trial (COMET): randomised controlled trial. Lancet. 2003; 362: 7-13

105. Remme WJ, Cleland JG, Erhardt L, Spark P, Torp-Pedersen C, Metra M, Komajda M, Moullet C, Lukas MA, Poole-Wilson P, Di LA, Swedberg K: Effect of carvedilol and metoprolol on the mode of death in patients with heart failure. Eur.J.Heart Fail. 2007; 9: 1128-35

106. Remme WJ, Torp-Pedersen C, Cleland JG, Poole-Wilson PA, Metra M, Komajda M, Swedberg K, Di LA, Spark P, Scherhag A, Moullet C, Lukas MA: Carvedilol protects better against vascular events than metoprolol in heart failure: results from COMET. J.Am.Coll.Cardiol. 2007; 49: 963-71

Page 106: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

98

107. Beattie WS, Wijeysundera DN, Karkouti K, McCluskey S, Tait G, Mitsakakis N, Hare GM: Acute surgical anemia influences the cardioprotective effects of beta-blockade: a single-center, propensity-matched cohort study. Anesthesiology. 2010; 112: 25-33

108. Beattie WS, Karkouti K, Wijeysundera DN, Tait G: Risk associated with preoperative anemia in noncardiac surgery: a single-center cohort study. Anesthesiology. 2009; 110: 574-81

109. Reiter MJ: Cardiovascular drug class specificity: beta-blockers. Prog.Cardiovasc.Dis. 2004; 47: 11-33

110. Ragoonanan TE, Beattie WS, Mazer CD, Tsui AK, Leong-Poi H, Wilson DF, Tait G, Yu J, Liu E, Noronha M, Dattani ND, Mitsakakis N, Hare GM: Metoprolol reduces cerebral tissue oxygen tension after acute hemodilution in rats. Anesthesiology. 2009; 111: 988-1000

111. Hocht C, Di VC, Opezzo JA, Bramuglia GF, Taira CA: Pharmacokinetic-pharmacodynamic (PK-PD) modeling of cardiovascular effects of metoprolol in spontaneously hypertensive rats: a microdialysis study. Naunyn Schmiedebergs Arch.Pharmacol. 2006; 373: 310-8

112. van BJ, Trouwborst A, Schwarte L, Siegemund M, Ince C, Henny C: Intestinal and cerebral oxygenation during severe isovolemic hemodilution and subsequent hyperoxic ventilation in a pig model. Anesthesiology. 2002; 97: 660-70

113. van BJ, Siegemund M, Henny C, Ince C: Heart, kidney, and intestine have different tolerances for anemia. Transl.Res. 2008; 151: 110-7

114. van Klei WA, Bryson GL, Yang H, Forster AJ: Effect of beta-blocker prescription on the incidence of postoperative myocardial infarction after hip and knee arthroplasty. Anesthesiology. 2009; 111: 717-24

115. Ances BM: Coupling of changes in cerebral blood flow with neural activity: what must initially dip must come back up. J.Cereb.Blood Flow Metab. 2004; 24: 1-6

116. Vovenko EP, Chuikin AE: Oxygen tension in rat cerebral cortex microvessels in acute anemia. Neurosci.Behav.Physiol. 2008; 38: 493-500

117. Hare GM, Worrall JM, Baker AJ, Liu E, Sikich N, Mazer CD: Beta2 adrenergic antagonist inhibits cerebral cortical oxygen delivery after severe haemodilution in rats. Br.J.Anaesth. 2006; 97: 617-23

118. Habib RH, Zacharias A, Schwann TA, Riordan CJ, Durham SJ, Shah A: Adverse effects of low hematocrit during cardiopulmonary bypass in the adult: should current practice be changed? J.Thorac.Cardiovasc.Surg. 2003; 125: 1438-50

119. Karkouti K, Djaiani G, Borger MA, Beattie WS, Fedorko L, Wijeysundera D, Ivanov J, Karski J: Low hematocrit during cardiopulmonary bypass is associated with increased risk of perioperative stroke in cardiac surgery. Ann.Thorac.Surg. 2005; 80: 1381-7

Page 107: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

99

120. Fossum E, Moan A, Kjeldsen SE, Devereux RB, Julius S, Snapinn SM, Edelman JM, de FU, Fyhrquist F, Ibsen H, Kristianson K, Lederballe-Pedersen O, Lindholm LH, Nieminen MS, Omvik P, Oparil S, Wedel H, Dahlof B: The effect of losartan versus atenolol on cardiovascular morbidity and mortality in patients with hypertension taking aspirin: the Losartan Intervention for Endpoint Reduction in hypertension (LIFE) study. J.Am.Coll.Cardiol. 2005; 46: 770-5

121. Shin J, Johnson JA: Pharmacogenetics of beta-blockers. Pharmacotherapy. 2007; 27: 874-87

122. Christensen KL, Mulvany MJ: Location of resistance arteries. J.Vasc.Res. 2001; 38: 1-12

123. Schubert R, Lidington D, Bolz SS: The emerging role of Ca2+ sensitivity regulation in promoting myogenic vasoconstriction. Cardiovasc.Res. 2008; 77: 8-18

124. Schiffrin EL, Hayoz D: How to assess vascular remodelling in small and medium-sized muscular arteries in humans. J.Hypertens. 1997; 15: 571-84

125. Noble A, Thomas A, Johnson R, Bass P: Large Blood Vessels, The Cardiovascular System: Basic Science and Clinical Conditions, 2 edition. Edited by Horne T, Stader L. Toronto, Churchill Livingstone, 2010, pp 85-96

126. Gericke A, Martinka P, Nazarenko I, Persson PB, Patzak A: Impact of alpha1-adrenoceptor expression on contractile properties of vascular smooth muscle cells. Am.J.Physiol Regul.Integr.Comp Physiol. 2007; 293: R1215-R1221

127. Noble A, Thomas A, Johnson R, Bass P: Resistance Blood Vessels, The Cardiovascular System: Basic Science and Clinical Conditions, 2 edition. Edited by Horne T, Stader L. Toronto, Churchill Livingstone, 2010, pp 97-112

128. Panerai RB: Cerebral autoregulation: from models to clinical applications. Cardiovasc.Eng. 2008; 8: 42-59

129. Schubert R, Lehmann G, Serebryakov VN, Mewes H, Hopp HH: cAMP-dependent protein kinase is in an active state in rat small arteries possessing a myogenic tone. Am.J.Physiol. 1999; 277: H1145-H1155

130. Matta BF, Heath KJ, Tipping K, Summors AC: Direct cerebral vasodilatory effects of sevoflurane and isoflurane. Anesthesiology. 1999; 91: 677-80

131. Stapleton MP: Sir James Black and propranolol. The role of the basic sciences in the history of cardiovascular pharmacology. Tex.Heart Inst.J. 1997; 24: 336-42

132. Parascandola J: Abel, Takamine, and the isolation of epinephrine. J.Allergy Clin.Immunol. 2010; 125: 514-7

133. Ahlquist R, .: A study of the adrenotropic receptors. Am.J.Physiol. 1948; 153: 586-600

Page 108: by Mostafa Hossam El Beheiry - University of Toronto T-Space · 2013-09-27 · Mostafa Hossam El Beheiry Master of Science, 2010 Department of Physiology, University of Toronto Background/Rationale:

100

134. Brodde OE, Bruck H, Leineweber K: Cardiac adrenoceptors: physiological and pathophysiological relevance. J.Pharmacol.Sci. 2006; 100: 323-37

135. Bai N, Moien-Afshari F, Washio H, Min A, Laher I: Pharmacology of the mouse-isolated cerebral artery. Vascul.Pharmacol. 2004; 41: 97-106

136. Gurdal H, Can A, Ugur M: The role of nitric oxide synthase in reduced vasocontractile responsiveness induced by prolonged alpha1-adrenergic receptor stimulation in rat thoracic aorta. Br.J.Pharmacol. 2005; 145: 203-10

137. Fujiwara T, Chiba S: Vascular alpha-1 antagonistic and agonistic effects of beta adrenoceptor agonists in rabbit common carotid arteries. J.Pharmacol.Exp.Ther. 1992; 263: 457-61