cerebrovascular effects of vasoactive drugs

295
Cerebrovascular effects of vasoactive drugs: In vitro, in vivo and clinical investigations Yohannes Ayele Mamo Submitted in total fulfilment of the requirements of the degree of Doctor of Philosophy June 2015 Department of Pharmacology and Therapeutics Faculty of Medicine, Dentistry and Health Sciences University of Melbourne Produced on archival quality paper

Upload: others

Post on 16-Apr-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cerebrovascular effects of vasoactive drugs

Cerebrovascular effects of vasoactive drugs:

In vitro, in vivo and clinical investigations

Yohannes Ayele Mamo

Submitted in total fulfilment of the requirements of

the degree of Doctor of Philosophy

June 2015

Department of Pharmacology and Therapeutics

Faculty of Medicine, Dentistry and Health Sciences

University of Melbourne

Produced on archival quality paper

Page 2: Cerebrovascular effects of vasoactive drugs
Page 3: Cerebrovascular effects of vasoactive drugs

i

Abstract

The brain receives a tightly controlled blood flow that maintains its metabolic

needs despite variations in arterial blood pressure. Various physiological factors

– neural, humoral and vascular – are involved in the regulation of cerebral blood

flow by controlling the tone of cerebral arteries and arterioles. This study

characterised the in vitro effects of common vasoactive agents in rat isolated

cerebral arteries, as well as the cerebrovascular responses to systemic

vasoactive drug administration using in vivo animal models and clinically in

patients. Variations were found in effects induced by a number of vasoactive

agents in arteries isolated from different regions of the rat cerebral circulation.

These were due to heterogeneous distribution of receptors mediating vascular

responses. Activation of 1-adrenoceptors that are located on smooth muscle

cells of the anterior and middle cerebral arteries, but absent in posterior

communicating and basilar arteries, induced vasoconstriction. Activation of -

adrenoceptors induced vasodilatation in all four arteries. In the middle cerebral

artery, β-adrenoceptors are located on smooth muscle, however they are

located on endothelium in the basilar artery. The -adrenoceptor-mediated

relaxation in basilar artery involved release of nitric oxide (NO) and/or

induction of endothelial-derived hyperpolarisation (EDH) depending on which

agonist is used. Noradrenaline and isoprenaline favoured release of NO and

induction of EDH, respectively, in rat basilar artery. Activation of endothelin

receptors induced similar effects in all cerebral arteries tested. The endothelin-

1-induced contraction of cerebral arteries involved activation of both voltage-

operated calcium channels (VOCC) and non-VOCC. T-type VOCC are activated

by low endothelin-1 concentrations, while L-type were activated by high

concentrations of endothelin-1. In vivo investigation of cerebral arterial

reactivity using the rat cranial window technique showed that the

cerebrovascular effects induced by systemic administration of vasoactive

agents were predominantly autoregulatory responses to changes in arterial

blood pressure induced by the drugs. However, phenylephrine and vasopressin

may also have direct constrictor effects on cerebral arteries. The effects of

Page 4: Cerebrovascular effects of vasoactive drugs

ii

pre-emptive vasopressor administration on cerebral oxygen saturation during

anaesthesia in the ‘beach chair’ position was investigated in patients.

Noradrenaline showed a beneficial effect in preventing hypotension and

cerebral oxygen desaturation. In contrast, vasopressin did not prevent

hypotension and exacerbated cerebral desaturation. This study has shown that

regulation of cerebrovascular tone involves a complex integration between the

direct effects of vasoactive mediators on cerebral arteries and indirect

responses to changes in peripheral blood pressure. Thus, it is a matter of utmost

importance to understand the mechanisms mediating effects of vasoactive

drugs if they are to be used for neuroprotection during the management of

cerebrovascular disorders.

Page 5: Cerebrovascular effects of vasoactive drugs

iii

Declaration

This is to certify that:

The thesis comprises only my original work towards the PhD except

where indicated in the statement of contribution of others.

Due acknowledgement has been made in the text to all other

materials used.

The thesis is less than 100,000 words in length, exclusive of tables,

maps, bibliographies and appendices.

Yohannes Ayele Mamo (B.Pharm, M.Sc)

June 2015

Page 6: Cerebrovascular effects of vasoactive drugs

iv

Statement of contribution of others

Chapter 6: Experiments involving in vivo testing of vasoactive drugs in rats were

conducted in the Ocular Physiology laboratory, Melbourne Brain Centre. Dr Bang

Bui, Dr Zheng He and Mr Shajan Velaedan assisted in surgical preparation, drug

infusion and imaging. All the analyses were carried out by Yohannes Mamo.

Chapter 7: Experiments involving clinical testing of vasopressor drugs in

consented patients were conducted in The Avenue Hospital (Windsor,

Melbourne, VIC 3181, Australia) and the data presented in this chapter

represents a subset analysis of 24 patients recruited to specifically investigate

noradrenaline, vasopressin and the control (saline) group. This is a component

of a larger study (ANZCTR No 12610001075077). Associate Professor Paul

Soeding, Mr Grey Hoy (Orthopaedic Surgeon) and Mr Martin Spurgeon (Nurse)

have been involved in conducting the experiments. Data analyses were carried

out by Yohannes Mamo and A/Prof Paul Soeding.

Page 7: Cerebrovascular effects of vasoactive drugs

v

Acknowledgements

I must express a profound gratitude to my supervisors A/Prof Christine Wright,

A/Prof James Ziogas and A/Prof Paul Soeding for the unreserved support they

provided during my entire candidature. It is difficult to overstate Christine’s

generous support and guidance from the beginning to the end of my time in her

lab. It would have been impossible to complete my PhD research without her

fervent and persistent supervision. Her insightful comments and constructive

criticisms at different stages of my research were thought-provoking and helped

me focus my ideas and pay attention to the details. I am grateful to her for

bigheartedly holding me up to a high research standard and enforcing strict

validations for each result, and thus teaching me how to do a real research with

integrity. I am thankful to Jimmy for sharing his knowledge and experience.

The many valuable discussions I had with him helped me understand my

research area better and taught me how to question, organize thoughts and

express ideas. I am deeply grateful to Paul for exposing me to the clinical arena.

Those long discussions I had with him, along with his encouragement, have

assisted me to sort out the clinical study results. I am grateful to A/Prof Peter

Crack who, as a chair of my supervisory panel, has been presiding at several

progress meetings.

My sincere appreciation and gratitude goes to Prof James Angus, one of the

smartest pharmacologists I have ever known. Jim has always been there and

willing to share his knowledge and experience and provided his vivid comments

and suggestions. Jim’s unwaning passion for Science, broad spectrum of

knowledge and lateral thinking are truly inspirational. His support has

facilitated my study and helped me enrich research questions, design critical

experiments and define some intricate ideas.

I would like to acknowledge Dr Bang Bui, Dr Zheng He and Mr Shajan Velaedan

for their marvellous assistance in conducting in vivo experiments. Your hard

work spirts, hands-on experiments, as well as cheerful lab environment are

exceptional. It was a great experience working with them.

Page 8: Cerebrovascular effects of vasoactive drugs

vi

I would like to extend my gratitude to members of the Cardiovascular

Therapeutics Unit, past and present, for abundantly sharing their skills and

providing an all rounded support during my time in the lab. Mark, Linda,

Heather, Nits, Richard, Khammy and Ashenafi have been great colleagues and

friends whose presence made life much easier during those stressful PhD years.

I was lucky to be a part of this group and would like to thank these wonderful

people.

I would like to thank the Department of Pharmacology and Therapeutics for

providing a favourable research environment and sponsoring national and

international conferences. I thank Professor Daniel Hoyer, Mrs Fanoula

Mouratidis and all of the staff and fellow RHD students in the department for

their support throughout the years.

Many friends have helped me stay stable through these difficult years. Their

support and care helped me overcome setbacks and stay focused on my

graduate study. I greatly value their friendship and I deeply appreciate their

belief in me.

Most importantly, none of this would have been possible without the love and

care of my family. I would like to thank my wife Yeshimebet Desalegn -she has

always been there supporting me and stood by me through the good and bad

times. My lovely son Elnathan is the pride and joy of my life and has always

been a source of encouragement throughout.

Finally, I appreciate the RHD studentship offered by the Cardiovascular

Therapeutics Unit providing financial support towards living expenses and the

Melbourne Fee Remission Scholarship (MIFRS) awarded by the Faculty of

Medicine, Dentistry and Health Sciences, the University of Melbourne.

Page 9: Cerebrovascular effects of vasoactive drugs

vii

Communications

Publications

Mamo YA, Angus JA, Ziogas J, Soeding PF, Wright CE (2014). The role

of voltage-operated and non-voltage-operated calcium channels in

endothelin-induced vasoconstriction of rat cerebral arteries.

European Journal of Pharmacology: 742:65-73.

Mamo YM, Ziogas J, Soeding PF, Angus JA, Wright CE (2014).

Characterization of voltage-operated and non-voltage-operated

calcium channels in endothelin-induced vasoconstriction of rat

cerebral arteries. The FASEB Journal: 28 (1 Supplement), 841.6.

[Abstract]

Soeding PF, Currigan DA, Mamo YA (2015). Effect of interscalene

anaesthesia on cerebral oxygen saturation, Anaesthesia and Intensive

Care- In press [accepted in May 2015].

Soeding PF, Currigan DA, Mamo YA, Ziogas J, Hoy G (2015). Effect of

vasopressor infusion on cerebral oxygen saturation during beachchair

surgery. British Journal of Anaesthesia. Publication submitted on

16/06/2015.

National conference presentations

Mamo YA, Ziogas J, Soeding PF, Wright CE (2012). Poster: Endothelin

and endothelin receptor antagonism in rat isolated cerebral arteries:

relevance for subarachnoid haemorrhage, Joint ASCEPT-APSA

conference, 2-5 December 2012, Sydney.

Mamo YA, Ziogas J, Soeding PF, Wright CE (2013). Poster: Endothelin

causes vasoconstriction of rat cerebral arteries by differential

activation of voltage-operated and non-voltage operated calcium

channels. ASCEPT Annual Scientific Meeting, 1-4 December 2013,

Melbourne.

Mamo YA, Angus JA, Wright CE (2014). Major role of endothelial nitric

oxide release in the responses to noradrenaline in arteries isolated

Page 10: Cerebrovascular effects of vasoactive drugs

viii

from different regions of the rat cerebral circulation. ASCEPT-

MPGPCR joint scientific meeting, 7-11 December 2014, Melbourne.

International conference presentation

Mamo YA, Ziogas J, Soeding PF, Wright CE (2014). Poster: Endothelin

causes vasoconstriction of rat cerebral arteries by differential

activation of voltage-operated and non-voltage operated calcium

channels. Experimental Biology Annual Meeting, 26-30 April 2014, San

Diego.

Oral presentation

Mamo YA, Ziogas J, Soeding PF, Wright CE (2013). The roles of

voltage-operated and non-voltage-operated calcium channels in

endothelin-induced vasoconstriction of rat cerebral arteries. In:

Cardiovascular Research Domain Young Investigator Awards, MDHS,

University of Melbourne

Abstracts co-authored

Soeding P, Currigan D, Mamo Y, Hoy G (2014). The effect of

vasopressor therapy on cerebral oxygen saturation during beach chair

surgery. ANZCA Annual Scientific Meeting, Singapore.

Soeding P, Currigan D, Mamo Y, Hoy G (2014). The effect of

interscalene anaesthesia on cerebral oxygen saturation. ANZCA

Annual Scientific Meeting, Singapore.

Page 11: Cerebrovascular effects of vasoactive drugs

ix

Table of Contents

General introduction ......................................... 1

Introduction ..................................................................... 2

Anatomy of the cerebral circulation ........................................ 3

1.2.1. Organization of the cerebral circulation ................................ 3

1.2.2. The pial arteries and arterioles ........................................... 7

1.2.3. Parenchymal arterioles ..................................................... 8

1.2.4. The brain microcirculation ................................................ 8

1.2.5. Collateral vessels ............................................................ 9

The barriers of the brain ..................................................... 11

The neurovascular unit ....................................................... 12

Physiological regulation of cerebral blood flow .......................... 14

1.5.1. Cerebral autoregulation ................................................... 14

1.5.2. Neural control .............................................................. 15

1.5.3. Myogenic control ........................................................... 16

1.5.4. Endothelial control ........................................................ 17

i. Nitric oxide (NO) .............................................................. 18

ii. Prostacyclin (PGI2) ........................................................... 19

iii. Endothelium-derived hyperpolarisation (EDH) ......................... 19

1.5.5. Metabolic control .......................................................... 22

i. Hypoxia ......................................................................... 22

ii. Hypercapnia .................................................................. 22

Pathophysiology of the cerebral circulation .............................. 24

1.6.1. Cerebral ischaemia ........................................................ 24

1.6.2. Cerebral vasospasm ........................................................ 26

i. Pathogenesis of cerebral vasospasm ....................................... 26

ii. Role of inflammation in cerebral vasospasm ............................ 27

1.6.3. Migraine ..................................................................... 28

1.6.4. Alzheimer’s disease ........................................................ 28

1.6.5. Huntington’s disease ...................................................... 29

1.6.6. Seizures ...................................................................... 29

Mechanism of smooth muscle contraction in the cerebral circulation 31

1.7.1. Smooth muscle contraction ............................................... 31

Page 12: Cerebrovascular effects of vasoactive drugs

Table of content

x

1.7.2. Regulation of the intracellular free calcium concentration ......... 34

i. Calcium entry ................................................................. 34

ii. Calcium release from intracellular stores ............................... 35

1.7.3. Voltage-operated calcium channels (VOCC) ........................... 35

i. Structure of VOCC ............................................................ 35

ii. VOCC subtypes ............................................................... 36

iii. Function of VOCC ........................................................... 37

1.7.4. Role of calcium channels in basal tone of cerebral arteries ......... 39

1.7.5. Calcium channels involved in agonist-induced contraction of

cerebral arteries ........................................................... 40

Therapeutic options for the management of cerebral blood flow

disorders ........................................................................ 43

Objectives of the study....................................................... 45

Methods of in vitro small artery reactivity studies .... 47

Introduction .................................................................... 48

Physiological salt solutions................................................... 53

Protocols for in vitro vascular studies ..................................... 55

2.3.1. Preparation of Krebs’ PSS and dissolution of drugs ................... 55

2.3.2. Animals ...................................................................... 55

2.3.3. Extraction of the rat brain ................................................ 56

2.3.4. Dissection and setup of cerebral arteries .............................. 57

2.3.5. Normalization ............................................................... 58

2.3.6. Testing the effects of drugs .............................................. 61

2.3.7. Removal of endothelium .................................................. 62

Collection and analysis of data .............................................. 63

Effects of common vasoactive agents in rat isolated

cerebral arteries ............................................ 65

Introduction .................................................................... 66

3.1.1. Effects of endothelial factors on cerebrovascular tone .............. 67

3.1.2. The sympathetic nervous system ........................................ 67

3.1.3. The parasympathetic nervous system ................................... 68

3.1.4. 5-Hydroxytryptamine (5-HT) ............................................. 69

Page 13: Cerebrovascular effects of vasoactive drugs

Table of content

xi

3.1.5. Neuropeptides .............................................................. 69

3.1.6. Bradykinin ................................................................... 70

3.1.7. Vasopressin .................................................................. 70

3.1.8. Angiotensin .................................................................. 71

3.1.9. Soluble guanyly cyclase (sGC) stimulation/activation ................ 71

Materials and methods ....................................................... 74

3.2.1. Isolation of arteries ........................................................ 74

3.2.2. Experimental protocols ................................................... 74

3.2.3. Drugs and chemicals ....................................................... 74

Results .......................................................................... 75

3.3.1. Effects of noradrenaline in cerebral arteries .......................... 75

3.3.2. Contractile effects of endothelin-1 in rat cerebral arteries ......... 78

3.3.3. Contractile effects of 5-hydroxytryptamine (5-HT) in rat cerebral

arteries ...................................................................... 79

3.3.4. Contractile effects of vasopressin in rat cerebral arteries .......... 80

3.3.5. Effects of acetylcholine in rat cerebral arteries ...................... 81

3.3.6. Relaxant effects of bradykinin in rat cerebral arteries .............. 82

3.3.7. Relaxant effect of sodium nitroprusside in rat cerebral arteries ... 83

3.3.8. The effects of BAY 41-8543 in rat cerebral arteries .................. 84

Discussion ....................................................................... 86

Mechanisms involved in adrenoceptor-mediated

responses in rat cerebral arteries ........................ 91

Introduction .................................................................... 92

Materials and methods ....................................................... 95

4.2.1. Methods ...................................................................... 95

4.2.2. Experimental protocols ................................................... 95

4.2.3. Drugs and chemicals ....................................................... 95

Results .......................................................................... 96

4.3.1. Contractile effects of α1-adrenoceptor stimulation in cerebral

arteries ...................................................................... 96

4.3.2. Relaxant effects of β-adrenoceptor stimulation in cerebral

arteries ...................................................................... 96

Page 14: Cerebrovascular effects of vasoactive drugs

Table of content

xii

4.3.3. Effects of endothelium removal and inhibition of nitric oxide

production on isoprenaline-induced relaxation of cerebral

arteries ...................................................................... 99

4.3.4. Contractile effects of noradrenaline and the role of endothelium

and nitric oxide ........................................................... 101

4.3.5. Relaxant effects of noradrenaline in rat cerebral arteries and the

role of endothelium and nitric oxide .................................. 105

4.3.6. Roles of α- and β-adrenoceptors in the responses induced by

noradrenaline in rat cerebral arteries ................................ 110

4.3.7. Involvement of the endothelium-derived hyperpolarisation (EDH) in

β-adrenoceptor-mediated relaxation of cerebral arteries ......... 116

Discussion ..................................................................... 121

Effects of endothelin in the rat cerebral circulation and

the role of voltage-operated and non-voltage-operated

calcium channels .......................................... 129

Introduction .................................................................. 130

Materials and methods ..................................................... 134

5.2.1. Methods .................................................................... 134

5.2.2. Experimental protocols ................................................. 134

5.2.3. Drugs and chemicals ..................................................... 135

Results ........................................................................ 136

5.3.1. Effects of endothelin receptor stimulation in rat cerebral

arteries .................................................................... 136

5.3.2. Antagonism of endothelin receptors in rat cerebral arteries ...... 139

5.3.3. Effects of inhibiting nitric oxide production and endothelium

removal on the endothelin-1-induced contraction of cerebral

arteries .................................................................... 143

5.3.4. Contractile effects of endothelin in calcium-free physiological salt

solution .................................................................... 145

5.3.5. Effects of nifedipine pre-treatment on endothelin-1-induced

contraction ................................................................ 147

5.3.6. Effects of NNC 55-0396 pre-treatment on endothelin-1-induced

contraction ................................................................ 148

5.3.7. Effects of SK&F 96365 pre-treatment on endothelin-1-induced

contraction ................................................................ 148

Page 15: Cerebrovascular effects of vasoactive drugs

Table of content

xiii

5.3.8. Combined effects of nifedipine and SK&F 96365 on endothelin-1-

induced contraction ..................................................... 149

5.3.9. Relaxation of endothelin-1-induced contraction with calcium

channel blockers ......................................................... 151

Discussion ..................................................................... 157

Cerebrovascular effects of vasoactive substances in vivo

............................................................... 163

Introduction .................................................................. 164

Materials and methods ..................................................... 167

6.2.1. Anaesthesia and pre-surgical preparation ............................ 167

6.2.2. Cannulation of arteries and veins ..................................... 167

6.2.3. Cranial window preparation ............................................ 168

6.2.4. Imaging and image analysis ............................................. 169

6.2.5. Drug administration and haemodynamic monitoring................ 170

6.2.6. Experimental protocols ................................................. 171

6.2.7. Statistical analysis ....................................................... 172

Results ........................................................................ 173

6.3.1. Effects of intravenous vasoactive agents on blood pressure and

cerebral arterial diameter .............................................. 173

6.3.2. Intravenous versus intra-arterial effects of drug infusion on blood

pressure and cerebral arterial diameter.............................. 181

6.3.3. Time-course of effects of vasopressor agents on blood pressure and

cerebral artery diameter ............................................... 187

Discussion ..................................................................... 191

Effects of vasopressor agents on human cerebral oxygen

saturation during anaesthesia ........................... 197

Introduction .................................................................. 198

7.1.1. Background to clinical problem ........................................ 198

7.1.2. Principle of cerebral oximetry ......................................... 200

Materials and methods ..................................................... 204

7.2.1. Patient recruitment ..................................................... 204

7.2.2. Anaesthesia and patient preparation for experiments ............. 204

Page 16: Cerebrovascular effects of vasoactive drugs

Table of content

xiv

7.2.3. Experimental protocols ................................................. 205

7.2.4. Intervention ............................................................... 207

7.2.5. Statistical analysis ....................................................... 207

Results ........................................................................ 208

7.3.1. Patient characteristics and intra-operative parameters ........... 208

7.3.2. Effects of vasopressor agents on mean arterial blood pressure and

heart rate ................................................................. 210

7.3.3. Effects of vasopressor agents on cerebral oxygen saturation ..... 213

Discussion ..................................................................... 217

Summary and conclusions ................................ 223

References ............................................................... 229

Page 17: Cerebrovascular effects of vasoactive drugs

xv

List of figures

Figure 1-1: Organization of the human cerebral circulation. ..................... 4

Figure 1-2: Comparison of the human and rat cerebral circulations. ............ 6

Figure 1-3: The human cerebral venous system. .................................... 7

Figure 1-4: Architecture of the pial and cerebral microcirculation. ........... 10

Figure 1-5: Structural components of the blood brain barrier. ................. 13

Figure 1-6: Cerebral autoregulation and the vascular response to blood pressure

changes. ................................................................................. 15

Figure 1-7: Relaxation of vascular smooth muscle cells by endothelium-derived

factors. .................................................................................. 21

Figure 1-8: Mechanisms of smooth muscle contraction. ......................... 33

Figure 1-9: Structure of the voltage-operated calcium channel (Cav1). ....... 37

Figure 2-1: The first small artery wall tension measurement technique. ..... 50

Figure 2-2: An early version of wall tension measurement in intact cylindrical

segments of small resistance arteries. ............................................. 51

Figure 2-3: A wire myograph setup .................................................. 52

Figure 2-4: A photographic image of freshly isolated rat brain (ventral surface).

........................................................................................... 58

Figure 2-5: A representative trace demonstrating normalization of the basilar

artery. ................................................................................... 60

Figure 2-6: A screenshot display of a typical normalization window (DMT module

plugin) in LabChart7 (AD Instruments). ............................................. 61

Figure 3-1: Contractile effects of noradrenaline in rat cerebral arteries. .... 76

Figure 3-2: Relaxant effects of noradrenaline in rat cerebral arteries. ....... 77

Figure 3-3: Contractile effects of endothelin-1 in rat cerebral arteries. ...... 79

Figure 3-4: Contractile effects of 5-hydroxytryptamine (5-HT) in rat middle

cerebral and basilar arteries. ........................................................ 80

Figure 3-5: Contractile effects of vasopressin in rat middle cerebral and basilar

arteries. ................................................................................. 81

Page 18: Cerebrovascular effects of vasoactive drugs

List of figures

xvi

Figure 3-6: Relaxant effects of acetylcholine in rat middle cerebral and basilar

arteries. ................................................................................. 82

Figure 3-7: Relaxant effects of bradykinin in rat middle cerebral and basilar

arteries. ................................................................................. 83

Figure 3-8: Relaxant effects of sodium nitroprusside in rat middle cerebral and

basilar arteries. ........................................................................ 84

Figure 3-9: Relaxant effects of BAY 41-8543 in rat cerebral arteries. ......... 85

Figure 4-1: Effects of α1-adrenoceptor stimulation in rat cerebral arteries. . 97

Figure 4-2: Relaxant effects of β-adrenoceptor stimulation in cerebral arteries

........................................................................................... 98

Figure 4-3: Effects of endothelium removal on isoprenaline-induced relaxation

of cerebral arteries. .................................................................. 100

Figure 4-4: Effects of inhibition of nitric oxide production on isoprenaline-

induced relaxation of cerebral arteries. .......................................... 101

Figure 4-5: Contractile effects of noradrenaline in rat cerebral arteries .... 103

Figure 4-6: Effect of endothelium removal on the noradrenaline-induced

contraction of middle cerebral artery. ............................................ 104

Figure 4-7: Effects of inhibition of nitric oxide production on noradrenaline-

induced contraction of cerebral arteries. ......................................... 105

Figure 4-8: Relaxant effects of noradrenaline in rat cerebral arteries. ...... 107

Figure 4-9: Effects of endothelium removal on noradrenaline-induced relaxation

of rat basilar artery. ................................................................. 108

Figure 4-10: Effects of inhibition of nitric oxide production on the

noradrenaline-induced contraction of cerebral arteries. ....................... 109

Figure 4-11: Effects of adrenoceptor antagonism on the noradrenaline-induced

contraction in middle cerebral artery. ............................................ 112

Figure 4-12: Effects of β-adrenoceptor antagonism on the noradrenaline-

induced relaxation of cerebral arteries. .......................................... 113

Figure 4-13: The roles of β1- and β2-adrenoceptors in noradrenaline-induced

relaxation of basilar arteries. ....................................................... 114

Page 19: Cerebrovascular effects of vasoactive drugs

List of figures

xvii

Figure 4-14: Relaxant effects of adrenaline and formoterol in rat basilar

arteries. ................................................................................ 115

Figure 4-15: Relaxant effects of clonidine in rat cerebral arteries. .......... 116

Figure 4-16: Relaxant effects of isoprenaline in cerebral arteries pre-contracted

with K+ .................................................................................. 118

Figure 4-17: Effects of apamin and charybdotoxin combination on the relaxation

induced by noradrenaline and isoprenaline in rat basilar arteries. ........... 119

Figure 4-18: Effects of endothelial-derived EDH, NO and PGI2 inhibition on the

isoprenaline-induced relaxation of cerebral arteries. ........................... 120

Figure 4-19: Summary of the location and effects mediated by β-adrenoceptors

and effects of noradrenaline and isoprenaline in rat cerebral arteries. ..... 126

Figure 5-1: The endothelin pathway. .............................................. 132

Figure 5-2: Representative trace of the endothelin-induced contraction of

cerebral arteries. ..................................................................... 137

Figure 5-3: Contractile effects of endothelin-1 in rat cerebral arteries. ..... 138

Figure 5-4: Relaxant effects of sarafotoxin S6C in rat cerebral arteries. .... 139

Figure 5-5: Effects of bosentan on the endothelin-induced contraction of

cerebral arteries. ..................................................................... 141

Figure 5-6: Effects of inhibiting nitric oxide production on the endothelin-1-

induced contraction of cerebral arteries. ......................................... 144

Figure 5-7: Effects of endothelium removal on endothelin-1-induced

contraction of cerebral arteries. ................................................... 145

Figure 5-8: The contractile effects of endothelin-1 in calcium-free medium.

.......................................................................................... 147

Figure 5-9: Effect of calcium channel inhibition on endothelin-induced

contraction of middle cerebral arteries. .......................................... 150

Figure 5-10: Comparison of the effects of calcium channel antagonists on

endothelin-1-induced contraction of cerebral arteries. ........................ 151

Figure 5-11: Relaxant effects of calcium channel antagonists in cerebral arteries

pre-contracted with endothelin-1. ................................................. 155

Page 20: Cerebrovascular effects of vasoactive drugs

List of figures

xviii

Figure 5-12: Relaxation of endothelin-1 pre-contracted cerebral arteries with

combinations of calcium channel antagonists .................................... 156

Figure 5-13: A schematic representation of the contribution of different calcium

channels in the contractile response to endothelin-1. .......................... 162

Figure 6-1: Major physiological factors affecting cerebral blood flow and the

effects of vasoactive drugs. ......................................................... 165

Figure 6-2: Carotid artery cannulation. ........................................... 168

Figure 6-3: Cranial window preparation and imaging. .......................... 170

Figure 6-4: Representative images of cerebral arteries illustrating the effects

of vasoactive drug infusion .......................................................... 175

Figure 6-5: Effects of saline on blood pressure, heart rate and cerebral artery

outer diameter. ....................................................................... 176

Figure 6-6: Effects of phenylephrine on blood pressure, heart rate and cerebral

artery outer diameter. ............................................................... 177

Figure 6-7: Effects of vasopressin on blood pressure, heart rate and cerebral

artery outer diameter. ............................................................... 178

Figure 6-8: Effects of prenalterol on blood pressure, heart rate and cerebral

artery outer diameter. ............................................................... 179

Figure 6-9: Effects of formoterol on blood pressure, heart rate and cerebral

artery outer diameter. ............................................................... 180

Figure 6-10: Intravenous versus intra-arterial effects of phenylephrine on blood

pressure and cerebral artery outer diameter. .................................... 184

Figure 6-11: Intravenous versus intra-arterial effects of vasopressin on blood

pressure and cerebral artery outer diameter. .................................... 185

Figure 6-12: Intravenous versus intra-arterial effects of formoterol on blood

pressure and cerebral artery outer diameter. .................................... 186

Figure 6-13: The time-course of effects of intravenous and intra-arterial

phenylephrine on blood pressure and cerebral artery outer diameter. ...... 188

Figure 6-14: The time-course of effects of intravenous and intra-arterial

vasopressin on blood pressure and cerebral artery outer diameter. .......... 189

Figure 6-15: The time-course of effects of intravenous and intra-arterial

formoterol on blood pressure and cerebral artery outer diameter............ 190

Page 21: Cerebrovascular effects of vasoactive drugs

List of figures

xix

Figure 7-1: Application of near infrared spectroscopy (NIRS) for monitoring

regional cerebral oxygen saturation. .............................................. 203

Figure 7-2: Study protocol for patients randomized to receive either saline,

noradrenaline or vasopressin infusion. ............................................ 206

Figure 7-3: A representative measurement of haemodynamic and anaesthesia

parameters recorded in a study patient. .......................................... 209

Figure 7-4: Effects of saline infusion on MAP and HR and ScO2 in anaesthetised

patients in supine and beach chair positions. .................................... 214

Figure 7-5: Effects of noradrenaline on MAP, HR and ScO2 in supine and beach

chair positions during anaesthesia. ................................................ 215

Figure 7-6: Effects of vasopressin on MAP, HR and ScO2 in supine and beach

chair positions during anaesthesia. ................................................ 216

Page 22: Cerebrovascular effects of vasoactive drugs

xx

List of tables

Table 1-1: Classification of the voltage-operated calcium channels, their

location, specific inhibitors and physiological functions. ........................ 38

Table 2-1: The composition and application of common physiological salt

solutions used in isolated tissue experiments. .................................... 54

Table 2-2: The composition of Krebs’ physiological salt solution used in this

study ..................................................................................... 56

Table 3-1: Summary of the common mediators released by extrinsic and

intrinsic perivascular nerves, their respective receptors and the responses

elicited in cerebral arteries. ......................................................... 73

Table 5-1: Comparison of the pEC50 and Emax values of ET-1 and the fold shift

caused by bosentan pre-treatment in rat cerebral and mesenteric arteries 142

Table 5-2: Endothelin-1 pEC50 values in rat isolated cerebral arteries in the

presence of various calcium channel antagonists ................................ 153

Table 5-3: The effects of various calcium channel antagonist pre-treatments on

the Emax of endothelin-1 in rat isolated cerebral arteries ...................... 154

Table 6-1: Baseline values of rat blood pressure, heart rate and cerebral artery

diameter before intravenous administration of the drugs. ..................... 174

Table 6-2: Baseline rat blood pressure and heart rate before intra-femoral vein

or intra-carotid artery administration of the drugs .............................. 182

Table 7-1: Characteristics of the patients randomly assigned to saline,

noradrenaline or vasopressin infusion groups ..................................... 209

Table 7-2: Intraoperative details of the patients assigned to saline,

noradrenaline and vasopressin groups ............................................. 210

Table 7-3: Summary of MAP, heart rate and ScO2 before and after anaesthesia

in the supine and beach chair positions. .......................................... 212

Page 23: Cerebrovascular effects of vasoactive drugs

xxi

List of abbreviations

5-HT 5-Hydroxytryptamine

ABC ATP-binding cassette

Aβ Amyloid-β peptide

BIS Bispectral index value

BKCa Large-conductance calcium-activated potassium channels

CBF Cerebral blood flow

CGRP Calcitonin-gene related peptide

COX-1, 2 Cyclooxygenase-1, 2

DAG Diacylglycerol

EC50 Concentration of agonist that evokes a half-maximal response

EDH Endothelium-derived hyperpolarisation

EDTA Ethylenediaminetetraamino acetic acid

EET Epoxyeicosatrienoic acid

Emax Maximum response (effects)

eNOS Endothelial NOS

ET-1 Endothelin-1

ETCO2 End tidal carbon dioxide concentration

ETSEVO End tidal sevoflurane concentration

HR Heart rate

HVA High voltage-activated

ICP Intracranial pressure

IKCa Intermediate-conductance calcium-activated potassium

channels

iNOS Inducible NOS

IP3R Inositol 1, 4, 5- triphosphate receptor

KATP ATP-dependent K+ channels

KPSS PSS containing 124 mm potassium

Kv Voltage-dependent potassium channels

L-NAME N-nitro-L-arginine methyl ester

LVA Low voltage-activated

Page 24: Cerebrovascular effects of vasoactive drugs

List of abbreviations

xxii

MAP Mean arterial pressure

MDR Multidrug resistance

MLC Myosin light chain

MLCK Myosin light chain kinase

NIRS Near-infrared spectroscopy

nNOS Neuronal NOS

NO Nitric oxide

NOS Nitric oxide synthase

Paw Airway pressure

pEC50 -Log10ec50

PGI2 Prostacyclin

P-gp P-glycoprotein

PKC Protein kinase C

PLC-β Phospholipase C-β isoform

PRP Transient receptor potential

PSS Physiological salt solution

ROCC Receptor-operated calcium channels

ScO2 Cerebral oxygen saturation

sGC Soluble guanylyl cyclase

SKCa Small-conductance calcium-activated potassium channels

SOCC Store-operated calcium channels

SR Sarcoplasmic reticulum

TNF-α Tumour necrosis factor-α

TRPC1 Canonical transient receptor potential type-1

TXA2 Thromboxane A2

VOCC Voltage-operated calcium channels

Page 25: Cerebrovascular effects of vasoactive drugs

1

General introduction

Page 26: Cerebrovascular effects of vasoactive drugs

General introduction

2

Introduction

The brain is one of the most perfused organs receiving about 15-20% of the total

cardiac output and 20% of the available oxygen supply since it relies on

oxidative metabolism for its high metabolic needs. Glucose is the only substrate

for energy metabolism of the brain unlike other tissues (Cipolla, 2009). It

maintains a relatively constant blood flow over a wide range of arterial pressure

not only to provide optimum supply of oxygen and nutrients but also to protect

itself from damage due to overflow as the brain tissue cannot expand due to

the skull enclosure. The tone of cerebral arteries is controlled by concerted

action of various neural and endothelial mediators, as well as factors circulating

in blood. In addition, the water and solute transport into the brain is regulated

by the blood-brain barrier (Strange, 1992). Diseases such as stroke dysregulate

the normal function of cerebral arteries, such that cerebral blood flow is

obstructed consequently leading to cerebral ischaemia. In this chapter, the

organization of cerebral circulation, physiological regulation of cerebral blood

flow, factors involved in controlling cerebrovascular tone, pharmacological

profile of cerebral vasculature and diseases affecting cerebral blood flow and

the available treatment options will be reviewed. A detailed discussion of some

mediators and pharmacological agents is presented in Chapter 3.

Page 27: Cerebrovascular effects of vasoactive drugs

General introduction

3

Anatomy of the cerebral circulation

1.2.1. Organization of the cerebral circulation

The brain receives its arterial blood supply through two pairs of major arteries

- the right and left internal carotid and the right and left vertebral arteries, as

shown in Figure 1-1 (Day, 1987). The common carotid arteries originate from

the aortic arch within the chest and travel to the neck before bifurcating into

internal and external carotid arteries at the C3-C4 level of the vertebrae. The

external carotid artery then branches into several arteries that supply blood to

extracranial tissues as well as the basal and lateral brain surfaces. The internal

carotid arteries primarily supply the cerebral hemispheres and the eyes. Each

internal carotid artery contributes approximately 40% of the cerebral blood

flow (Edvinsson et al., 1993), the remainder being supplied by the two vertebral

arteries that distally fuse to form the basilar artery. Branches of the basilar

artery provide blood to the brainstem, cerebellum and cerebral cortex.

Page 28: Cerebrovascular effects of vasoactive drugs

General introduction

4

Figure 1-1: Organization of the human cerebral circulation.

The brain receives its blood supply through the right and left internal carotid as well

as a pair of vertebral arteries. The arteries ultimately join the Circle of Willis, an

anatomical anastomosis. Different arteries that branch off the Circle of Willis to

distribute blood supply to the two cerebral hemispheres are shown (University of

Miami Health System http://surgery.med.miami.edu/images/Circulation_of_

brain.gif).

The basilar artery branches into the right and left posterior cerebral arteries

that join the two ipsilateral internal carotid arteries via the posterior

communicating arteries to form the Circle of Willis, a ring of arteries

surrounding the pituitary stalk. Three pairs of main arteries - anterior, middle,

Page 29: Cerebrovascular effects of vasoactive drugs

General introduction

5

and posterior cerebral arteries - originate from the Circle of Willis and supply

the two hemispheres (Figure 1-1 and Figure 1-2). In humans, the proximal

portion of the anterior cerebral artery (A1) originates from the internal carotid

artery and passes over the optic chiasm connecting with its contralateral artery

through the anterior communicating artery that stabilizes blood flow between

anterior regions of the brain hemispheres. The two distal segments of the

anterior cerebral arteries (A2) enter the longitudinal fissure and run between

the hemispheres and pass in front of the corpus callosum. Several arteries

branch out of the cerebral artery to supply anterior regions of the brain (Day,

1987). The middle cerebral arteries originate from the internal carotid artery

at the medial end of the Sylvian fissure and supply the lateral cerebral

hemispheres. Abundant perforating vessels originate along the length of the

middle cerebral artery and supply blood to the brain (Day, 1987). Anatomical

variations in the Circle of Willis, often lack of anterior and posterior cerebral

arteries, are common in humans (Eftekhar et al., 2006; Hashemi et al., 2013).

This may potentially contribute to asymmetric flow rates in bilateral arteries

and can cause certain clinical consequences including ischaemic stroke (Tanaka

et al., 2006; Kardile et al., 2013). The anatomical organizations of the cerebral

circulation are generally similar in human and the rat. For this reason, the rat

is usually considered a suitable model to study cerebrovascular diseases. Rat

isolated cerebral arteries are also widely used to investigate the in vitro effects

of pharmacological agents on cerebral arteries. Nevertheless, some differences

are noted between the human and rat cerebral circulations in specific arteries

as shown in Figure 1-2. For example, the internal carotid artery continues to

form the middle cerebral artery in the human, but integrates with the Circle of

Willis in the rat cerebral circulation. Further, the anterior communicating

artery is present in human but absent in the rat cerebral circulation (Lee, 1995).

Each artery progressively divides into smaller arteries that run on the surface

(called pial arteries) until they penetrate into the brain tissue to provide blood

to the corresponding regions of the cerebral cortex. The penetrating arteries

become parenchymal arterioles and finally form capillaries (Lee, 1995).

Page 30: Cerebrovascular effects of vasoactive drugs

General introduction

6

Figure 1-2: Comparison of the human and rat cerebral circulations.

The diagram demonstrates the organization of the human (A) and rat (B) cerebral

arterial circulation. The major cerebral arteries constituting the Circle of Willis are

found both in the human and the rat, but some differences are also shown. In the

human, the internal carotid artery continues to form the middle cerebral artery

while in the rat, the internal carotid artery integrates with the Circle of Willis.

Further, variations in anterior and posterior communicating arteries are noted

between the human and rat cerebral circulations (Lee, 1995).

Unlike the arteries, the cerebral venous system is made up of valveless veins

and sinuses. The veins draining the cerebral hemispheres are subdivided into

outer or superficial cortical veins that are located on the surface of the cortex

and the central veins that are located deep inside the brain. The superficial

veins drain the cerebral cortex and subcortical white matter, while the deep or

central veins, consisting of subependymal veins, internal cerebral veins, basal

vein, and the great vein of Galen, drain the brain’s interior, including the deep

white and grey matter surrounding the lateral and third ventricles. The

superficial and cortical veins finally anastomose and empty into the superior

sagittal sinus. The superior sagittal sinus is then directed via a confluence of

Page 31: Cerebrovascular effects of vasoactive drugs

General introduction

7

sinuses toward the sigmoid sinuses and jugular veins (Capra and Kapp, 1987;

Schaller, 2004; Kilic and Akakin, 2008) (Figure 1-3).

Figure 1-3: The human cerebral venous system.

The major superficial and deep lying cerebral veins as well as the common sinuses

are shown. The veins ultimately join together and form the sigmoid sinus which

descends down to form the jugular vein. The figure also shows various smaller veins

and sinuses draining specific regions of the brain (obtained from

http://imueos.wordpress.com/2010/10/10/base-of-the-skull/).

1.2.2. The pial arteries and arterioles

Intracranial arteries lying on the surface of the brain within the subarachnoid

or subpial space are called pial arteries. They are surrounded by cerebrospinal

fluid in the pia–arachnoid space (Zhang et al., 1990). The cerebral pial arteries

are extensively branched and are characterised by a network of collaterals,

branches that interconnect different arteries. When a certain artery is

occluded, the collaterals will provide an alternate supply of blood to the tissues

Page 32: Cerebrovascular effects of vasoactive drugs

General introduction

8

through another artery as discussed below (Coyle, 1994). The pial arteries

receive abundant peripheral innervations (extrinsic innervations) that release

different neurotransmitters (discussed in subsequent sections and in Chapter

3). The peripheral innervations are progressively lost as the arteries enter into

the brain parenchymal tissue (Figure 1-4) (Hamel, 2006). Remarkably, these

arteries contribute to about 50% of the cerebral vascular resistance (Cipolla et

al., 2014).

1.2.3. Parenchymal arterioles

The deep penetrating arteries become parenchymal arterioles once they enter

the brain tissue and become almost completely surrounded by astrocytes,

pericytes and neurons (Figure 1-4). They are usually long unbranched arteries

that connect the pial arteries to the capillaries (Cipolla et al., 2014).

Parenchymal arteries are structurally distinct from pial arteries and are usually

made up of single layer of smooth muscle cells. Parenchymal arterioles are

intrinsically innervated by afferent nerves from subcortical nuclei and the local

cortical interneurons (Hamel, 2006).

1.2.4. The brain microcirculation

The brain microcirculation consists of extensive capillary networks that are

made up of endothelium surrounded by basal lamina and pericytes, but lack

smooth muscle cells. The basal lamina is continuously covered by astrocytic

end-feet and sparsely by the neurons. The unique tight junctions of endothelial

cells, along with pericytes, astrocytes and the basal membrane, play a barrier

function (called the Blood-Brain barrier; discussed in section 1.3) that prevents

free movement of solutes between blood and brain tissues (Abott et al., 1990;

Cipolla, 2009). The tripartite association between neuron, astrocyte and

cerebral microvessel is usually referred to as the neurovascular unit (Figure

1-4).

Page 33: Cerebrovascular effects of vasoactive drugs

General introduction

9

1.2.5. Collateral vessels

The collateral circulation of the brain is a network of arteries and arterioles

that maintains stable cerebral blood flow when the primary routes are

obstructed or compromised. The Circle of Willis, an anastomotic loop with low

resistance, theoretically allows communication between the anterior and

posterior circuits. Under normal conditions, the Circle of Willis does not

function as a redistributor of cerebral blood flow, however, when the large

arteries supplying the brain are occluded, it allows collateral support to the

obstructed side depending on the anatomical ‘completeness’ of the

anastomosis (Day, 1987). The networks of pial arteries furnish secondary

collateral redistribution of blood flow when there is an occlusion of arteries

distal to the Circle of Willis. The penetrating arterioles are mostly unbranched

and lack collateral connections (Nishimura et al., 2007). Thus, occlusion of

these vessels results in infarction of the surrounding tissues.

Page 34: Cerebrovascular effects of vasoactive drugs

General introduction

10

Figure 1-4: Architecture of the pial and cerebral microcirculation.

The surface pial arteries receive extrinsic innervations: sympathetic nerves

originating from superior cervical ganglia (SCG), parasympathetic nerves originating

from the sphenopalatine ganglion (SPG) and the otic ganglion (OG), as well as sensory

neurons from the trigeminal ganglion (TG). Several neurotransmitters released by

the extrinsic and intrinsic nerves are shown. The penetrating arterioles and

capillaries are extensively surrounded by intrinsic nerves and astrocytic endfeet. The

microcirculation also receives neural projections from subcortical areas such as the

locus coeruleus, raphe nucleus, basal forebrain and the thalamus. The inset shows a

neurovascular unit (obtained from Hamel (2006)).

Page 35: Cerebrovascular effects of vasoactive drugs

General introduction

11

The barriers of the brain

There are three main interfaces that physically isolate the brain from other

compartments of the body - the cerebrospinal fluid-brain, blood-brain and

cerebrospinal fluid-blood interfaces. These interfaces are the main barriers

that provide protective functions to the central nervous system (Abbott et al.,

2006). The cerebrospinal fluid is formed by the choroid plexus and its contact

with the blood is prevented by the tight junction of ependymal cells lining the

plexus. The cerebrospinal fluid-brain barrier is provided by neuroependymal

cells interconnected by tight junctions in early life. However, the barrier is

gradually lost during adult age due to morphological changes in the ependymal

cells (Cipolla, 2009).

The blood-brain barrier is the main barrier that protects the neural tissue from

contamination of blood-borne substances. The blood-brain barrier is present in

pial arteries and arterioles, veins and capillaries, but is absent in some parts of

the brain such as the area postrema, median eminence, neurohypophysis,

pineal gland, subfornical organ and lamina terminalis (Ganong, 2000).

Structurally, the blood-brain barrier is made by endothelial cells connected

paracellularly via extensive tight junctions and adherens junctions (Figure 1-5)

(Ballabh et al., 2004). The blood-brain barrier is a selective barrier that allows

transcellular passage of small lipid soluble molecules (<400 Da) only. Larger

molecules such as glucose, amino acids, nucleotides and vitamins are

transported by specific transporters located either on the apical or basolateral

membrane of the endothelium. There are also active efflux mechanisms

including ATP-binding cassette (ABC) and multidrug resistance (MDR) P-

glycoproteins (P-gp) that remove metabolites and drugs from the brain (Cipolla,

2009). The permeability of the tight junctions can be modulated through actin

stress fibres in the cytoplasm of the endothelial cells (Wolburg et al., 1994).

Certain agonists (e.g. cAMP) decrease permeability of the tight junction by

relaxing these fibres, while other substances (e.g. protein kinase C and vascular

endothelial growth factors) increase permeability by promoting contraction of

the fibres (Abbott et al., 2006; Cipolla, 2009).

Page 36: Cerebrovascular effects of vasoactive drugs

General introduction

12

The neurovascular unit

The neurovascular anatomical unit comprises of endothelial cells, basal lamina,

pericytes, astrocytes and the neurons. Functional interaction between these

components interfaces neuronal activity with blood flow in order to

accommodate for the metabolic demands and provide means for removal of the

end-products (Merlini et al., 2012). Changes in localized cerebral blood flow in

response to increased metabolic need are known as neurovascular coupling or

functional hyperaemia. Increased metabolic activities lead to release of

neurotransmitters which may directly act on the vascular cells. Since the

surface of capillaries is extensively covered by astrocytes, the neurons may

activate the astrocytes which in turn alter the vascular tone (Petzold and

Murthy, 2011). The astrocytes release factors that regulate the function of

endothelial cells. In addition astrocytes provide a cellular link that transmits

neural signals to endothelial cells (Figure 1-5).

Page 37: Cerebrovascular effects of vasoactive drugs

General introduction

13

Figure 1-5: Structural components of the blood brain barrier.

The endothelial cells are encased with basal lamina, pericytes and astrocytic

endfeet. The neurons and microglia have sparse direct contact with the capillaries

- the astrocytes provide cellular links between the neurons and endothelial cells.

a) Example of the transporters expressed by endothelial cells – glucose transporter

1 (GLUT1), L-amino acid transporter 1 (LAT1), excitatory amino acid transporter 1-

3 (EAAT1-3) and P-glycoprotein (Pgp); and b) astrocyte-endothelial interactions -

astrocytes release various factors that activate specific receptors located on

endothelial cells. 5-HT, 5-hydroxytryptamine; ANG1, angiopoetin 1; bFGF, basic

fibroblast growth factor; ET1, endothelin; GDNF, glial cell line-derived

neurotrophic factor; LIF, leukaemia inhibitory factor; P2Y2, purinergic receptor;

TGFβ, transforming growth factor-β; TIE2, endothelium-specific receptor tyrosine

kinase 2. The tight junction between individual endothelial cells is shown (obtained

from Abbott et al. (2006)).

Page 38: Cerebrovascular effects of vasoactive drugs

General introduction

14

Physiological regulation of cerebral blood flow

1.5.1. Cerebral autoregulation

Cerebral autoregulation is defined as an inherent ability of the brain to

maintain the blood flow relatively constant over a wide range of peripheral

arterial blood pressure (about 50-150 mmHg) (Paulson et al., 1990; van Beek et

al., 2008). It allows constant blood flow that matches with the metabolic needs

of the brain in the face of fluctuation in perfusion pressure (arterial blood

pressure minus intracranial pressure). When the arterial blood pressure falls

below the lower limit, cerebral blood flow decreases since the vasodilatation

reaches the maximum capacity and the vessels collapse. Equally, as the blood

pressure exceeds the upper limit, the arteries are forcefully dilated by the

pressure, a condition called ‘breakthrough of autoregulation’, and fail to

control the cerebral blood flow (Figure 1-6). Thus, the cerebral blood flow

increases/decreases linearly with changes in peripheral arterial blood pressure

outside the autoregulatory range (Paulson et al., 1990; Cipolla, 2009). The

mechanism underlying cerebral autoregulation involves interaction of multiple

components including neurogenic, myogenic and metabolic factors (Busija,

1993).

Cerebral autoregulation can be disrupted by diseases such as chronic

hypertension, long-lasting diabetes, cerebrovascular diseases or any factor that

causes excessive vasoconstriction or vasodilatation. Hypertension shifts both

the lower and upper limits towards higher pressure (called hypertensive

adaptation)(Strandgaard et al., 1973). In chronic diabetic patients, the normal

autoregulatory plateau is absent so that the cerebral blood flow depends partly

on the peripheral arterial blood pressure (Edvinsson et al., 1993). Acute

cerebrovascular ischaemia is another common cause of loss of autoregulation,

known as vasomotor paralysis. The autoregulatory function of the blood vessels

is lost after ischaemic stroke in areas distal to the occlusion (Eames et al.,

2002; Immink et al., 2005). A more detailed discussion of cerebral

autoregulation is presented in Chapter 6 and Chapter 7.

Page 39: Cerebrovascular effects of vasoactive drugs

General introduction

15

Figure 1-6: Cerebral autoregulation and the vascular response to blood

pressure changes.

The vasodilator or vasoconstrictor responses of cerebral arteries are responsible for

maintenance of a constant cerebral blood flow (CBF) despite fluctuations in blood

pressure within the autoregulatory range (50-150 mmHg). Below the lower

autoregulatory limit, impaired dilation of the arteries will lead to cerebral

ischaemia, whereas above the autoregulatory limit, forceful dilation of the arteries

causes uncontrolled cerebral blood flow that results in vasogenic oedema and brain

damage (obtained from Pires et al. (2013)).

1.5.2. Neural control

The cerebral circulation has been known to receive abundant peripheral

innervation, called extrinsic innervation. The sympathetic, parasympathetic

and sensory innervations of the cerebral circulation originate mainly from

superior cervical, trigeminal and sphenopalatine ganglia, respectively (Lee et

al., 1976; Duckles, 1980; Paulson et al., 1990; Lincoln, 1995; Mitsis et al., 2009;

ter Laan et al., 2013). In addition, cerebral arteries receive neural projections

from various subcortical nuclei (called intrinsic innervations) such as the

Page 40: Cerebrovascular effects of vasoactive drugs

General introduction

16

thalamus, locus coeruleus, raphe nucleus and basal forebrain (Raichle et al.,

1975; Micieli et al., 1994; Hamel, 2006). However, the role of the various neural

systems in the regulation of cerebral blood flow has been controversial. For

example, several studies have shown that the autonomic nervous system plays

a role in the beat-to-beat regulation of cerebral blood flow in humans (Zhang

et al., 2002), but others reported contradictory results (Heistad and Marcus,

1978; Tuor, 1992; Roatta et al., 1998; Dunatov et al., 2011). The potential role

of perivascular nerves in cerebral autoregulation may be changed during

disease conditions such as subarachnoid haemorrhage (Tsukahara et al., 1986a;

Tsukahara et al., 1988; Budohoski et al., 2013). In line with this, sympathetic

activity has been greatly increased in patients with subarachnoid haemorrhage

as manifested by persistent elevation of the noradrenaline concentration in

plasma (Naredi et al., 2000). The effects of the nerves releasing common

neurotransmitters on cerebral arteries are discussed in Chapter 3.

1.5.3. Myogenic control

An intrinsic property of vascular smooth muscle cells to respond to changes in

mechanical stretch or intravascular pressure is known as the myogenic

response. Some arteries constrict in response to increased transmural pressure

or dilate when the pressure is decreased or removed. The myogenic response is

more prominent in small cerebral arteries and arterioles than other vascular

beds in several species (Wallis et al., 1996). The myogenic response is important

for maintenance of arterial resistance and perfusion of tissues when the arterial

blood pressure decreases (Davis, 2012). In addition, it protects smaller

arterioles and capillaries from physical damage under conditions of increased

perfusion pressure (Paulson et al., 1990). The mechanism by which wall tension

or pressure on the vessels elicits the myogenic response is not clearly

understood. Multiple ion channels, including the calcium, potassium and

chloride channels, may be activated when the smooth muscle cells are

stretched (Bulley et al., 2012). Potassium channels the ATP-sensitive K+(KATP),

large-conductance Ca2+-activated K+(BKCa), voltage-activated K+(KV) and inward

rectifier K+(KIR) channels are particularly important since the electrochemical

Page 41: Cerebrovascular effects of vasoactive drugs

General introduction

17

gradient of K+ ion plays critical role in the regulation of membrane potential

and vascular tone (Jackson, 2000). Transient receptor potential (TRP) channels,

which are non-selective cation channels, also play a significant role in myogenic

activation of vascular smooth muscle contraction (Earley and Brayden, 2010).

Multiple TRP channels may be involved in the mechanotransduction of myogenic

tone in cerebral arteries (Kim et al., 2013). The myogenic response is associated

with membrane depolarization that leads to increased calcium influx through

the voltage-operated calcium channels (VOCC). It has been indicated that both

L- and T-type VOCC may also contribute to the genesis of the myogenic response

(Abd El-Rahman et al., 2013).

The myogenic response can be deregulated during cerebral ischaemia and

reperfusion (Palomares and Cipolla, 2013). Multiple mechanisms are involved in

the ischaemia-induced loss of the myogenic response. First, increased

generation of peroxynitrite can lead to endothelial damage which in turn

affects release of vasoactive substances such as nitric oxide or endothelin-1

(ET-1). The increased level of vasoactive mediators can directly affect the

myogenic tone in cerebral arteries and arterioles. Second, ischaemia may alter

the response of vascular smooth muscle cells to an increased pressure by

causing certain structural changes such as depolymerisation of the actin

cytoskeleton (Cipolla et al., 2001).

1.5.4. Endothelial control

In addition to its barrier function of protecting the brain tissues from blood-

borne substances, the cerebrovascular endothelium plays a critical role in

regulating cerebral blood flow. The vascular endothelium releases several

vasoactive mediators that can directly affect the vascular tone. Both

vasoconstrictor agents, such as ET-1 and thromboxane A2 (TXA2), and

vasodilator agents, such as nitric oxide (NO), prostacyclin (PGI2) and

endothelium-derived hyperpolarisation (EDH), are released by the

endothelium. More substances with vasodilator properties including carbon

monoxide, hydrogen peroxide, hydrogen sulphide, potassium ion (K+) and

Page 42: Cerebrovascular effects of vasoactive drugs

General introduction

18

methane have been reported to be released by the vascular endothelium (Qi et

al., 2011) (Figure 1-7).

i. Nitric oxide (NO)

NO is one of the most extensively investigated relaxing factors released by the

vascular endothelium. It is constitutively synthesized from L-arginine by

enzymatic action of nitric oxide synthase (NOS) (Tousoulis et al., 2012). There

are three isoforms of NOS - endothelial NOS (eNOS), neuronal NOS (nNOS) and

inducible NOS (iNOS). Both eNOS and nNOS are constitutively expressed and

well characterised in the cardiovascular and nervous systems. iNOS, however,

is activated only during inflammation or after stimulation of the immune system

(Arnold et al., 1977; Ignarro et al., 1987; Bryan et al., 2009).

Several vasoactive substances released from nerve terminals, such as

acetylcholine, substance P and calcitonin-gene related peptide (CGRP) can

induce endothelial production of NO. The NO produced in endothelial cells can

freely diffuse into smooth muscle cells where it increases the level of cGMP by

activating the soluble guanylyl cyclase enzyme (sGC). cGMP then triggers a

cascade of intracellular events that culminate in vascular smooth muscle

relaxation. Furthermore, endothelial nitric oxide causes a number of other

cellular effects including smooth muscle proliferation, platelet aggregation,

and leukocyte recruitment (Zhong et al., 2011).

The basal production of NO contributes to the physiological regulation of

cerebral blood flow by inhibiting the resting tone or decreasing cerebral

vascular resistance. This maintains the local blood flow to the brain tissues

(Faraci, 1994). It has been suggested that NO also contributes to maintenance

of the cerebral blood flow by extending the lower limit of cerebral

autoregulation. Inhibition of NOS depresses the cerebral blood flow responses

to hypotension (vertical autoregulation) in rats (Stephen, 2003), while the

autoregulation curve was shifted to the right in eNOS knock-out mice (Atochin

and Huang, 2011).

Page 43: Cerebrovascular effects of vasoactive drugs

General introduction

19

Endothelial impairment or decreased nitric oxide production can result in

serious impairment of the cerebral circulation. The production and signalling of

NO can be significantly affected by cerebral ischaemia. Occlusion of the middle

cerebral artery caused transient increase followed by long-term deficiency of

NO leading to neurological damage (Terpolilli et al., 2012). Deficiency of NO

due to dysfunctional eNOS has also been implicated in the pathogenesis of

cerebral vasospasm following subarachnoid haemorrhage (Pluta, 2006). On the

other hand, overproduction of nitric oxide may result in the development or

exacerbation of neurological damage in Alzheimer’s disease, multiple sclerosis,

head trauma or stroke (Toda et al., 2009).

ii. Prostacyclin (PGI2)

Another vasoactive mediator produced by the vascular endothelium is PGI2.

Activation of phospholipase A2 leads to liberation of arachidonic acid from

membrane-bound lipids. Arachidonic acid is converted to prostaglandin H2

(PGH2) by catalytic action of cyclooxygenase-1 (COX-1) and COX-2 enzymes.

PGH2 is further metabolized by prostacyclin synthase enzyme to generate PGI2

(Marcus et al., 1980). PGI2 activates IP receptors to elicit relaxation of vascular

smooth muscle cells. PGI2 relaxes cerebral arteries but considerable regional

and interspecies variation of the response exist (Brown and Pickles, 1982; Uski

et al., 1983). PGI2 was found to be decreased in primate cerebral arteries after

subarachnoid haemorrhage leading to the suggestion that a deficiency in PGI2

may be implicated in the pathogenesis of cerebral vasospasm (Nosko et al.,

1988).

iii. Endothelium-derived hyperpolarisation (EDH)

Endothelium-derived hyperpolarisation (EDH) refers to the agonist-induced

endothelium-dependent vasodilator mechanism(s) excluding NO and the COX

products (Cipolla, 2009). The vascular smooth muscle cell hyperpolarisation

may be caused by several factors released from the endothelium as shown in.

Figure 1-7. EDH is generally characterised by increased potassium conductance

with subsequent hyperpolarisation, and thus relaxation, of vascular smooth

Page 44: Cerebrovascular effects of vasoactive drugs

General introduction

20

muscle cells. Certain agonists like acetylcholine can cause membrane

hyperpolarisation by increasing potassium efflux. The major potassium channels

involved in the hyperpolarisation are the small- (SKCa), intermediate- (IKCa) and

large-conductance calcium-activated potassium channels (BKCa) expressed in

endothelial cells (Marrelli et al., 2003; Ozkor and Quyyumi, 2011). Activation

of a receptor on endothelial cells by an agonist causes release of calcium from

the endoplasmic reticulum. An increase in intracellular calcium activates the

KCa in the proximity of the myoendothelial junction and triggers a complex

process including transmission of hyperpolarisation to the smooth muscle cells

via gap junctions. This process ultimately promotes the relaxation of smooth

muscle cells. Basal EDH is present in all penetrating cerebral arterioles and is

responsible for inhibition of the basal tone. This is typically manifested by

vasoconstriction induced in cerebral arteries when the SKCa and IKCa channels

are inhibited (Ledoux et al., 2006; Cipolla et al., 2009).

Epoxyeicosatrienoic acids (EETs), metabolites of endothelial cytochrome P450

epoxygenase, and H2O2 may also contribute to the EDH function (Campbell et

al., 1996; Fleming, 2001; Bellien et al., 2006; Fleming and Busse, 2006). In

addition, EDH may work in synergy with NO and PGI2 in regulating the vascular

tone. Thus, endothelium-dependent vasodilatation is maintained during states

of decreased NO availability such as hypertension, owing to the compensatory

increase of EDH activity (Taddei et al., 1999).

Page 45: Cerebrovascular effects of vasoactive drugs

General introduction

21

Figure 1-7: Relaxation of vascular smooth muscle cells by endothelium-

derived factors.

Activation of endothelial cells due to shear stress caused by blood flow or agonist-

induced stimulation of specific surface receptors leads to increased intracellular Ca2+

concentration resulting in activation of various enzymes such as endothelial nitric

oxide synthase (eNOS), cyclooxygenase (COX) or cytochrome P450 2C (CYP450 2c).

Activation of phospholipase A2 liberates membrane-bound arachidonic acid that is

converted to either prostacyclin (PGI2) or eicosatrienoic acids (EETs) by the catalytic

action of COX or CYP450 2c, respectively. eNOS and COX increase production of nitric

oxide (NO) and prostacyclin (PGI2) which cause smooth muscle relaxation by

elevating the level of cGMP or cAMP in the smooth muscle cells. Elevated cytosolic

Ca2+ concentration causes membrane depolarization that is conducted to smooth

muscle cells through the myoendothelial gap junction. Depolarization of smooth

muscle cells finally causes relaxation of blood vessels (Ozkor and Quyyumi, 2011)

.

Page 46: Cerebrovascular effects of vasoactive drugs

General introduction

22

1.5.5. Metabolic control

Metabolic demand due to increased neuronal activity of the brain is among the

driving factors that increase cerebral blood flow. A number of mediators are

proposed to play important roles in coupling neuronal activity to the cerebral

blood flow. These include CO2, H+, O2, adenosine and adenosine nucleotides, K+

and Ca2+ (Paulson et al., 1990). Both hypoxia and hypercapnia seem to play

critical roles in cerebral blood flow.

i. Hypoxia

It has been well known that hypoxia increases cerebral blood flow by inducing

a vasodilator response in cerebral arteries (Poulin and Robbins, 1998). However,

the cerebral blood flow does not respond to hypoxia until the tissue PO2 falls

below ~50 mmHg. Further decrease in PO2 can significantly increase the

cerebral blood flow (up to 400% of resting levels). Hypoxia causes cerebral

vasodilation through a number of mechanisms. Decrease in ATP levels due to

hypoxia can induce smooth muscle membrane depolarization by activating ATP-

dependent K+ channels (KATP). Hypoxia can also increase cerebral blood flow by

inducing release of vasodilator mediators. Acute hypoxia promotes upregulation

of eNOS thus inducing local release of NO (Min et al., 2006). Further, hypoxia

induces release of adenosine from endothelial cells (Coney and Marshall, 1998).

Prolonged hypoxia can increase cerebral tissue perfusion by increasing capillary

density in the brain (Harik et al., 1995; Cipolla, 2009).

ii. Hypercapnia

Hypercapnia significantly increases cerebral blood flow by dilating the cerebral

arteries and arterioles whereas hypocapnia causes the opposite effect (Kety

and Schmidt, 1948; Kontos et al., 1971). The vasodilatation induced by CO2 may

involve different mechanisms including effects of extracellular H+ or through

release of NO or the prostanoids (Cipolla, 2009). Increased extracellular H+

concentration, which is an initial step, may lead to membrane hyperpolarisation

through direct activation of the K+ channels or indirectly by activating the COX

Page 47: Cerebrovascular effects of vasoactive drugs

General introduction

23

and NOS enzymes and generating the prostanoids and NO, respectively. These

mechanisms ultimately relax the vascular smooth muscle cells by decreasing

the level of intracellular calcium (Brian, 1998).

Page 48: Cerebrovascular effects of vasoactive drugs

General introduction

24

Pathophysiology of the cerebral circulation

Cerebral blood flow is known to be affected, focally or globally, under several

disorders of the brain. Morbidities and mortalities associated with disease

states such as ischaemia and haemorrhagic stroke are caused by diminished or

complete obstruction of blood flow to certain regions of the brain. Several brain

diseases including neurodegenerative disorders and migraine headache are also

associated with abnormal cerebral blood flow. Alzheimer’s disease,

Huntington’s disease, seizures and migraine are among the pathologic

conditions wherein cerebral blood flow is manifestly disrupted. Thus, cerebral

vascular disorders need to be emphasised while dealing with these diseases.

1.6.1. Cerebral ischaemia

Cerebral ischaemia is a condition where the blood flow is insufficient to meet

its metabolic needs of the brain. It may be caused by obstruction or constriction

of the cerebral arteries. The poor oxygen supply can result in brain tissue

infarction. Decreases in oxygen and glucose delivery lead to decoupling of

oxidative phosphorylation and thus lessen ATP production and disruption of ion

transport systems. This causes elevation of intracellular K+ concentration due

to the failure of the Na-K pump (Na-K ATPase) and causes neuronal

depolarization that promotes Ca2+ influx through voltage-operated calcium

channels, subsequently leading to release of excitotoxic amino acids such as

glutamate. This condition finally leads to an irreversible neuronal damage or

death. In addition, elevated intracellular calcium can decrease cellular

integrity and survival by activating a multitude of enzymes that are involved in

apoptotic cell death (Turner et al., 2013). Chronic ischaemia can trigger further

damage by activating different inflammatory mechanisms. Initially, ischaemia

activates the microglia (inflammatory cells in the brain) followed by influx of

circulating inflammatory cells including granulocytes, leukocytes, T cells and

monocytes from the blood. Excessive release of cytokines and chemokines

causes oxidative stress and disruption of the blood-brain barrier further

propagating the inflammatory responses (Turner et al., 2013).

Page 49: Cerebrovascular effects of vasoactive drugs

General introduction

25

There is no clear understanding of the direct effects of acute ischaemia on

cerebral autoregulation. It has been suggested that, while ischaemia is caused

due to decreased blood flow below a critical level to an area of the brain, it

may also induce dysfunction of autoregulation in the surrounding areas.

However, there is insufficient evidence showing failure of the autoregulatory

capacity of the vessels in the surrounding infarction area (Jordan and Powers,

2012). On the other hand, ischaemia-reperfusion can cause a macro- or micro-

vascular dysregulation that may lead to exacerbation of brain injury.

Reperfusion after long lasting ischaemia (more than 15-30 minutes) can

promote loss of control on cerebral blood flow by impairing the various vascular

autoregulatory mechanisms. Decrease in myogenic tone was observed after 6-

12 hours of reperfusion. Occlusion of the arteries causes vascular paralysis or

dilatation of the arteries downstream by activating release of different

vasodilator mechanisms. These include myogenic vasodilatation (dilator

response due to drop in arterial wall pressure) and build-up of carbon dioxide

and lactic acidosis. Ischaemic reperfusion will then induce hyperaemia because

the ‘paralysed’ arteries will no more be able to regulate blood flow to the

ischaemic area. Exposure of the microcirculation to uncontrolled and excessive

pressure results in further damage of the neurons, disruption of the blood-brain

barrier and brain oedema. The mechanism underlying diminished tone of the

arteries after ischaemia-reperfusion has been suggested to involve oxidative

stress (due to generation of reactive oxygen and nitrogen species) and loss of

F-actin in vascular smooth muscle cells (Palomares and Cipolla, 2011).

Ischaemia-reperfusion may also alter the vasodilator function of NO. Even

though ischaemia induces sustained upregulation of eNOS, postischaemic

reperfusion actually diminishes NO-mediated vasodilatation. This could be due

to the uncoupling of eNOS that ends up in generation of superoxide anions

instead of NO. One reason for uncoupling of eNOS could be decreased

availability of substrate (L-arginine) and the cofactor tetrahydrobiopterin (BH4)

which have been depleted during ischaemia and oxidation or inactivation of the

cofactor during reperfusion. Ischaemia-reperfusion may also affect dilatation

of cerebral arteries by enhancing EDH responses via activation of SKCa and IKCa

Page 50: Cerebrovascular effects of vasoactive drugs

General introduction

26

channels in the parenchymal arterioles (Huk et al., 1997; Kidd et al., 2005;

Sanchez et al., 2006; Cipolla and Godfrey, 2010).

Furthermore, ischaemia-reperfusion may alter the effects of endothelin-1 in

the cerebral arteries. Not only has the concentration of endothelin-1 been

increased in plasma, cerebrospinal fluid and brain tissue after stroke, but also

antagonism of ETA receptors improved microvascular reperfusion after transient

middle cerebral artery occlusion (Dawson et al., 1999). Apart from being a

potent vasoconstrictor peptide, endothelin-1 induces superoxide production

and disrupts the integrity of the blood-brain barrier, thus promoting cerebral

oedema.

1.6.2. Cerebral vasospasm

Cerebral vasospasm is a narrowing of arteries usually occurring few days after

the rupture of aneurysm. It usually occurs in two phases. There is an acute

initial phase starting in 1 to 3 days of subarachnoid haemorrhage. The delayed

phase, occurring between 4 to 14 days following subarachnoid haemorrhage, is

responsible for most neurological morbidities and mortality (Tani, 2002).

Angiographic vasospasm is observed in up to 70% of subarachnoid haemorrhage

patients, while 20-30% of them manifest clinical signs and symptoms. Nearly

half of the patients with symptomatic vasospasm will develop infarctions that

may lead to permanent disability or death (Dumont et al., 2003).

i. Pathogenesis of cerebral vasospasm

The mechanism leading to development of vasospasm is still elusive. It has been

proposed that the blood products released when the aneurysm ruptures may

directly or indirectly play the main role in the pathogenesis of vasospasm. A

number of products released as a result of bleeding have vasoconstrictor

activity. Oxyhaemoglobin and fibrin degradation products are among these

spasmogens abundantly found in cerebrospinal fluid of subarachnoid

haemorrhage patients (Hirano and Hirano, 2010). In addition, endothelin,

metabolites of arachidonic acid, catecholamines and 5-hydroxytryptamine (5-

Page 51: Cerebrovascular effects of vasoactive drugs

General introduction

27

HT) are suggested to contribute to the development of cerebral vasospasm

given their potent vasoconstrictor capacity and local production (Edvinsson and

Povlsen, 2011). Endothelin is typically increased in cerebrospinal fluid of

subarachnoid haemorrhage patients and thus has been at the centre of interest

for researchers.

Phenotypic changes of several vasoconstrictor receptors in vascular smooth

muscle cells of subarachnoid haemorrhage animal models and human

subarachnoid haemorrhage patients have been reported. Edvinsson and Povlsen

(2011) have reviewed studies on plasticity of contractile receptors as a

mechanism underlying cerebral vasospasm. Generally, receptor proteins and

mRNA of endothelin type B receptors (ETB), angiotensin II type 1 receptors

(AT1), 5-HT1B, and thromboxane A2 (TP) receptors were found to be up-

regulated after experimental subarachnoid haemorrhage.

ii. Role of inflammation in cerebral vasospasm

A series of complex inflammatory processes are elicited by presence of blood

components in the cerebrospinal fluid following the rupture of an arterial

aneurysm. After bleeding, the red blood cells are lysed in the cerebrospinal

fluid and release haemoglobin which can trigger an inflammatory cascade. This

involves upregulation of cell adhesion molecules on leukocytes and endothelial

cells thereby recruiting macrophages and neutrophils to the subarachnoid space

which phagocytose the haemoglobin. After some time, the trapped

macrophages and neutrophils themselves undergo apoptosis and release their

intracellular contents consisting mainly of inflammatory cytokines, reactive

oxygen species and vasoconstrictor mediators such as endothelin-1 into the

cerebrospinal fluid. This ultimately ends up in propagated inflammation and

vasoconstriction (Pradilla et al., 2010).

Overproduction of inflammatory mediators could be among the major

mechanisms of underlying delayed cerebral vasospasm. In this regard the

expression of tumour necrosis factor-α (TNF-α) in blood and vascular smooth

muscle cells of a rat model were shown to be markedly increased suggesting a

Page 52: Cerebrovascular effects of vasoactive drugs

General introduction

28

role of proinflammatory cytokines in the genesis of the spasm after

subarachnoid haemorrhage (Vecchione et al., 2009; Maddahi and Edvinsson,

2010).

Fassbender et al. (2001) evaluated the release of proinflammatory cytokines

(IL-1β, Il-6 and TNF-α) in patients with subarachnoid haemorrhage.

Interestingly, the cerebrospinal fluid level of the cytokines showed significant

increments between days 5 and 9, but declined thereafter. There was a parallel

decrease in blood flow (demonstrated by increased cerebral blood flow

velocity) between days 7 and 11, only slightly lagging behind. In addition, the

concentration of IL-6 was found to be significantly elevated in the cerebrospinal

fluid of subarachnoid haemorrhage patients with poor clinical outcome.

1.6.3. Migraine

Migraine is a condition characterised by a recurrent moderate or severe

headache lasting for up to several days. The pathogenesis of migraine is partly

related to dilatation of both intracranial and extracranial arteries (Spierings,

2003; Toda et al., 2009). The vasodilatation may be linked to excessive

production and release of NO via stimulation of certain endothelial receptors.

For example, activation of 5-HT2A receptors enhances the production of nitric

oxide in the trigeminovascular pathway in the rat. In addition to its vasodilator

effect, nitric oxide may also sensitize the perivascular as well as central

trigeminal nociceptors (Srikiatkhachorn et al., 2002; Barbanti et al., 2014).

1.6.4. Alzheimer’s disease

The pathology of Alzheimer’s disease is predominantly caused by neurovascular

dysfunction wherein the availability of nitric oxide is decreased (Austin et al.,

2013). Chronic cerebral hypoperfusion seems to underlie the cognitive decline

associated with Alzheimer’s disease. The structure and function of the

endothelium may be altered due to generation of superoxide radicals as a result

of interaction with amyloid-β peptide (Aβ). Endothelium-dependent relaxation

was found to be profoundly decreased in transgenic mice overexpressing the

Page 53: Cerebrovascular effects of vasoactive drugs

General introduction

29

amyloid precursor protein (Khalil et al., 2002). The decrease in cerebral blood

flow may also be caused by cerebral thrombosis, atherosclerosis and cerebral

infarction or decreased cerebral arterial dilatation. Subsequently, there will be

reduced oxygen supply to an area in the brain leading to metabolic dysfunction,

increased superoxide production as well as Aβ deposition which may cause

progressive neurodegeneration and cognitive decline. In some patients the

expression of asymmetric dimethyl arginine, an endogenous NOS inhibitor, is

found to be increased (Toda et al., 2009; Hunter et al., 2012). Thus,

Alzheimer’s disease is thought to be associated with loss of functional

vasoregulation due to decreased production of NO.

1.6.5. Huntington’s disease

Dysfunctional cerebral blood flow has been described in Huntington’s disease,

a neurodegenerative disorder causing cognitive dysfunction, psychiatric

disturbance and movement disorders characterised by involuntary movements

as well as altered motor control (Chen et al., 2012). Decreased cerebral blood

flow may induce oxidative stress that causes neurodegeneration. The cause of

changes in blood flow is not clearly known but may involve overproduction of

nitric oxide. Increased nitric oxide production may lead to oxidative stress

through conversion into peroxynitrite (Aguilera et al., 2007; Toda et al., 2009).

1.6.6. Seizures

Increased cerebral blood flow and hyperbaric oxygen have been shown to

precede the clinical onset of seizures (Moseley et al., 2014). This may be

related to an elevated production of nitric oxide that increases cerebral blood

flow by dilating cerebral arteries several minutes before onset of the seizure.

Endothelial nitric oxide may play a key role in the development of hyperoxic

hyperaemia preceding oxygen seizures, while neuronal nitric oxide may

mediate toxic effects of hyperbaric oxygen primarily through its reaction with

superoxide to generate a stronger oxidant peroxinitrite (De Vasconcelos et al.,

2006; Toda et al., 2009). This assertion is supported by a previous finding that

Page 54: Cerebrovascular effects of vasoactive drugs

General introduction

30

local application of N-nitro-L-arginine, a nitric oxide synthase inhibitor,

abolished increases in rat cerebral blood flow due to an induced seizure by

topical bicuculline methiodide application via the cranial window (De

Vasconcelos et al., 1995).

Page 55: Cerebrovascular effects of vasoactive drugs

General introduction

31

Mechanism of smooth muscle contraction in the cerebral

circulation

1.7.1. Smooth muscle contraction

Vascular smooth muscle cells contract in response to mechanical, electrical or

chemical stimuli (Bayliss, 1902; Ehrreich and Clopper, 1970). Mechanical

stretching of the smooth muscle cells can induce contraction by stimulating

calcium channels. Smooth muscle cells also contract when electrically

stimulated or due to depolarization of the membrane. Chemical mediators are

the most common and physiologically relevant stimuli for smooth muscle

contraction. A number of endogenous ligands such as noradrenaline,

angiotensin II, vasopressin, endothelin-1 and thromboxane A2 affect the tone of

vascular smooth muscle cells. Binding of an agonist to a G-protein-coupled

receptor on smooth muscle cells stimulates the membrane-bound

phospholipase C-β isoform phospholipase (PLC-β) enzyme that catalyses

conversion of phosphatidylinositol 4,5-bisphosphate (PI2) into a soluble inositol

1,4,5-triphosphate (IP3) and a membrane-bound diacylglycerol (DAG)

(Alexander et al., 1985). IP3 stimulates its receptor located on the sarcoplasmic

reticulum (SR) and causes initial release of Ca2+, which then induces further

release of calcium (calcium-induced calcium release). The initial increase in

intracellular free calcium concentration [Ca2+]i may be responsible for the rapid

transient phase of smooth muscle contraction (Touyz and Schiffrin, 2000).

The sustained phase of smooth muscle contraction is associated with influx of

calcium from the extracellular space via the voltage-, ligand- or store-operated

calcium channels. DAG stimulates protein kinase C (PKC), a Ca2+- and a

phospholipid-dependent kinase that activates specific proteins involved in

smooth muscle contraction (Bell et al., 1986; Rasmussen et al., 1987). The free

cytosolic Ca2+ binds to calmodulin (CAM) to form a Ca2+-CAM complex that

activates myosin light chain kinase (MLCK). MLCK catalyses phosphorylation of

MLC, a 20 kD regulatory subunit of myosin. MLC phosphorylation increases

myosin ATPase activity and causes smooth muscle contraction by promoting

Page 56: Cerebrovascular effects of vasoactive drugs

General introduction

32

cross-bridging of myosin with actin filaments (Khalil, 2010). Furthermore,

activation of the Rho kinase pathway promotes sustained contraction of smooth

muscle cells by inducing calcium sensitization through inhibition of myosin

phosphatase activity (Fukata et al., 2001) (Figure 1-8).

Page 57: Cerebrovascular effects of vasoactive drugs

General introduction

33

Figure 1-8: Mechanisms of smooth muscle contraction.

Binding of an agonist (A) to a receptor (R) stimulates a membrane bound

phospholipase β enzyme (PLCβ) which converts phosphatidylinositol 4,5-

bisphosphate (PI2) into inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG). IP3

stimulates its receptor on the sarcoplasmic reticulum (SR) and causes release of Ca2+.

The free cytosolic Ca2+ binds to calmodulin (CAM) and thus activates MLC kinase

(MLCK) and causes MLC phosphorylation. DAG activates protein kinase C (PKC) which

phosphorylates CPI-17, and inhibits MLC phosphatase and thereby increases the

sensitivity of myofilament to Ca2+. PKC also phosphorylates calponin (Cap) and

promotes binding of actin to myosin. PKC may activate a mitogen-activated protein

kinase (MAPK) pathway leading to phosphorylation of the actin-binding protein

caldesmon (CaD). Activation of the RhoA/Rho-kinase pathway promotes contraction

of smooth muscle cells by inhibiting MLC phosphatase. G, heterotrimeric G protein;

Page 58: Cerebrovascular effects of vasoactive drugs

General introduction

34

PC, phosphatidylcholine; PS, phosphatidylserine; PE, phosphatidylethanolamine; AA,

arachidonic acid (from Khalil (2010)).

1.7.2. Regulation of the intracellular free calcium

concentration

Calcium (Ca2+) is an ubiquitous intracellular messenger that is involved in the

regulation of various cellular processes such as muscle contraction,

neurotransmission, secretion, gene transcription and cell proliferation

(Bootman et al., 2001). Calcium transport across membranes is the basic

homeostatic process by which eukaryotic cells maintain these functions. The

concentration of calcium in the cytoplasm is normally about four orders of

magnitude lower than that of the extracellular space in the resting state; this

gradient is maintained by different Ca2+ transporting systems. Intracellular Ca2+

needed for such activities as vascular smooth muscle contraction is provided by

the balance between entry from the extracellular space or release from internal

stores and extrusion via the plasmalemmal Ca2+-adenosine triphosphatase (Ca2+-

ATPase) and the Na+–Ca2+ exchanger, or uptake into intracellular stores (Alborch

et al., 1995; Nikitina et al., 2007).

i. Calcium entry

Cells use several different types of Ca2+ influx channels, which can be grouped

on the basis of their activation mechanisms: voltage-operated calcium channels

(VOCC), receptor-operated calcium channels (ROCC; also called ligand-gated

calcium channels), mechanically-activated calcium channels and store-

operated calcium channels (SOCC) (Bootman et al., 2001).

Voltage-operated calcium channels are mostly activated by depolarization of

the plasma membrane. They are employed by excitable cells such as muscle

and neural cells. ROCC are activated by the binding of agonists (such as

acetylcholine, serotonin, ATP or glutamate) to the extracellular domain of the

channels, while mechanically-operated channels open in response to cell

deformation or stress. SOCC are activated when the calcium in the intracellular

Page 59: Cerebrovascular effects of vasoactive drugs

General introduction

35

stores is depleted. ROCC include the transient receptor potential channels that

are ubiquitously expressed in almost all mammalian tissues. In addition, certain

unidentified channels may be activated by intracellular messengers such as DAG

or arachidonic acid (Berridge et al., 2000).

ii. Calcium release from intracellular stores

Calcium can be released from the endoplasmic or sarcoplasmic reticulum

through two classical channels – inositol 1, 4, 5- triphosphate receptors (IP3Rs)

and ryanodine receptors (RyRs) (Tykocki and Watts, 2010). IP3 can directly

activate IP3R and release Ca2+ from SR but DAG inhibits Ca2+ release through

activation of PKC. RyRs are activated by local increase of calcium in the

cytoplasm and furnish additional release of Ca2+ from SR amplifying the small

Ca2+ signal produced by VOCCs or IP3-mediated Ca2+ release. RyRs are also

involved in the termination of calcium influx through VOCC; i.e., localized

release of Ca2+ through RyRs, called ‘calcium spark’, activates calcium-sensitive

potassium channels, thus causing hyperpolarisation of the plasma membrane

which closes the VOCC (Berridge et al., 2000).

1.7.3. Voltage-operated calcium channels (VOCC)

i. Structure of VOCC

The voltage-operated Ca2+ channels comprise a family of structurally-related

proteins coupling electrical signals on the cell surface to intracellular events

like muscle contraction, secretion, neurotransmission and gene expression

(Catterall et al., 2005). Different types of VOCC have been identified based on

the properties of the Ca2+ current, pharmacological profile and structure of the

protein subunits constituting the channels (Catterall, 2000).

The voltage–operated calcium channels described to date are made up or five

subunits, namely α1, α2, , β and subunits. The α1 subunit has four homologous

domains (I-IV) with six transmembrane segments (S1-S6) in each domain as

shown in Figure 1-9. The S4 segment in each domain contains the voltage sensor

Page 60: Cerebrovascular effects of vasoactive drugs

General introduction

36

while the pore loop between S5 and S6 determines ion conductance and

selectivity (Catterall et al., 2005). The α1 subunit can form a functional channel

by itself; hence the other subunits are often called auxiliary or ancillary

subunits. However they can greatly modulate the electrophysiological and

pharmacological properties of the channels (Bergsman et al., 2000). The

diversity of VOCC is mostly related to the variation of the α1 subunits.

ii. VOCC subtypes

Based on the degree of depolarization required for their gating, VOCC can be

categorized into two major classes – low voltage-activated (LVA) and high

voltage-activated (HVA) calcium channels (Bergsman et al., 2000). Alphabetical

nomenclature had been used to describe the calcium currents transmitted by

these channels. Thus, VOCC were classified as T-, L-, N-, P/Q and R-type.

Currently, the α1 subunits of various VOCCs have been cloned and characterised

structurally and their familial relationship revealed. Based on their sequence

homology, ten α1 subunits have been identified and designated a new system

of nomenclature. Table 1-1 summarizes the new channel nomenclature along

with the alphabetical designation of the respective type of Ca2+currents.

Page 61: Cerebrovascular effects of vasoactive drugs

General introduction

37

Figure 1-9: Structure of the voltage-operated calcium channel (Cav1).

The different subunits constituting the Cav1 calcium channel (α1, α2, β, and ) and

the different domains of α1 subunit (I-IV) are shown (from Catterall et al. (2005).

The L- (Cav1.1-1.4), P/Q- (Cav2.1), N- (Cav2.2), and R-type (Cav2.3) VOCC are

activated at less negative voltages and thus are classified as HVA, while T-type

(Cav3.1-3.3) calcium channels are categorized as LVA since they are activated

at lower depolarization potentials (Nikitina et al., 2007; Kuo et al., 2011).

iii. Function of VOCC

Voltage-dependent calcium channels are ubiquitously distributed throughout

many different tissues and involved in a variety of functions (Bergsman et al.,

2000). They are mainly responsible for such functions as coupling of excitation

and contraction, rhythmic activities, excitation-secretion coupling, synaptic

nerve transmission, and proliferation as shown in Table 1-1 (Leanne, 2006).

Page 62: Cerebrovascular effects of vasoactive drugs

38

Table 1-1: Classification of the voltage-operated calcium channels, their location, specific inhibitors and physiological

functions.

Name Current Localization Selective antagonist Function

Cav1.1 L Skeletal muscle; transverse tubules Dihydropyridines, phenylalkylamines and benzothiazepines

Excitation-contraction coupling

Cav1.2 L Cardiomyoctes; smooth muscles; endocrine cells; neuronal cell bodies; proximal dendrites

Dihydropyridines, phenylalkylamines and benzothiazepines

Excitation-contraction coupling; hormone release; regulation of transcription; synaptic integration

Cav1.3 L Endocrine cells; neuronal cell bodies and dendrites; cardiac atrial myocytes and pacemaker cells; cochlear hair cells

Dihydropyridines, phenylalkylamines and benzothiazepines

Hormone release; regulation of transcription; synaptic regulation; cardiac pacemaking; hearing; neurotransmitter release from sensory cells

Cav1.4 L Retinal rod and bipolar cells, spinal cord, adrenal gland and mast cells

Dihydropyridines, phenylalkylamines and benzothiazepines

Neurotransmitter release, photoreceptors

Cav2.1 P/Q Nerve terminal and dendrites; neuroendocrine cells

-Agatoxin IVA Neurotransmitter release; dendritic Ca2+ transients; hormone release

Cav2.2 N Nerve terminals and dendrites; neuroendocrine cells

-Conotoxin GVIA Neurotransmitter release; dendritic Ca2+ transients; hormone release

Cav2.3 R Neuronal cell bodies and dendrites SNX-482 Repetitive firing; dendritic calcium transients

Cav3.1 T Neuronal cell bodies and dendrites; cardiac and smooth muscle myocytes

Kurotoxin, NNC55-0396 and mibefradil

Pacemaking; repetitive firing

Cav3.2 T Neuronal cell bodies and dendrites; cardiac and smooth muscle myocytes

Kurotoxin, NNC55-0396 and mibefradil

Pacemaking; repetitive firing

Cav3.3 T Neuronal cell bodies and dendrites Kurotoxin, NNC55-0396 and mibefradil

Pacemaking; repetitive firing

Adapted from Catterall et al. (2005).

Page 63: Cerebrovascular effects of vasoactive drugs

General introduction

39

L-type calcium channels play a central role in excitation-contraction coupling

in skeletal muscle, cardiac muscle and other smooth muscles. Other types of

VOCC may also play supportive roles during muscle contraction.

The L-type calcium channels of skeletal muscle, localized in the transverse

tubules system, are not directly required for contraction although blockade

thereof completely inhibits contraction. Rather, they serve as voltage sensors

for release of calcium from internal stores (Näbauer et al., 1989). Calcium

entry, however, is essential for contraction of cardiac and smooth muscles

unlike skeletal muscle (Davis and Hill, 1999). In contrast to striated muscles

(i.e., skeletal and cardiac), smooth muscles have a tendency to undergo a

biphasic contraction - fast tonic contraction mediated by L-type VOCC followed

by a slow phasic response, possibly mediated by T-type VOCCs (Leanne, 2006).

There is a growing body of evidence supporting the vasoconstrictor role of T-

type channels in renal and cerebral circulations (Navarro-Gonzalez et al., 2009;

Kuo et al., 2011). In cardiac cells, T-type channels furnish the major proportion

of current required for pacemaker depolarization in the sino-atrial node

(Bergsman et al., 2000).

1.7.4. Role of calcium channels in basal tone of cerebral

arteries

As in other peripheral blood vessels, calcium influx through VOCCs is the main

pathway for electromechanical coupling in cerebral arteries (Nikitina et al.,

2007; Kuo et al., 2011). The dynamic range of global cytosolic calcium ([Ca2+]i)

mediating contraction in cerebral arteries is however quite narrow (Wellman,

2006). As a result, cerebral arteries typically display spontaneous submaximal

constriction depending on the level of intraluminal pressure or isometric

tension. This phenomenon, termed myogenic tone, is an essential mechanism

in regulation of local blood flow and tissue perfusion in the cerebral vasculature

(Davis and Hill, 1999).

Page 64: Cerebrovascular effects of vasoactive drugs

General introduction

40

Basal uptake of Ca2+ in the resting state varies in cerebral arteries from

different species as measured using a 45Ca2+ isotope. The resting state of Ca2+

influx in dog basilar artery was found to be higher than that in the corresponding

mesenteric artery and inhibited by nifedipine (Asano et al., 1993). This shows

that the basilar artery may be more depolarized than the mesenteric artery in

the resting state. Voltage clamp studies also demonstrated that L-type calcium

channels have significant open-state probability around the normal resting

potential in canine basilar artery myocytes (Langton and Standen, 1993). On

the other hand, verapamil or nifedipine did not affect the basal uptake in

bovine isolated middle cerebral arteries, but significantly blocked the 45Ca2+

uptake induced by potassium or serotonin (Wendling and Harakal, 1987). This

indicates involvement of non-L-type VOCC in basal tone while L-type VOCCs are

mainly responsible for contraction induced by membrane depolarization.

1.7.5. Calcium channels involved in agonist-induced

contraction of cerebral arteries

Agonist-induced contraction of smooth muscle cells is dependent on Ca2+ influx

from the extracellular space. 5-HT, prostanoids (prostaglandin F2α and

thromboxane A2) and endothelins are generally known to increase Ca2+ influx

through the VOCCs or ROCC independent of depolarization or release from

intracellular stores (Salom et al., 1991; Wendling and Harakal, 1991; Tejerina

et al., 1993). However there is variation in the extent of Ca2+ influx induced by

the agonists in different species or different arterial beds within the same

species.

Despite the acceptance of L-type calcium channels as mediators of agonist-

induced cerebral artery contraction, nifedipine did not completely reverse this

in basilar arteries from rats, dogs and rabbits (Navarro-Gonzalez et al., 2009).

This indicates that other types of calcium channels may be involved in agonist-

induced vascular contraction or resting basal tone of cerebral arteries. It may

also support the suggestion of the existence of receptor-operated calcium

Page 65: Cerebrovascular effects of vasoactive drugs

General introduction

41

channels in addition to the conventional VOCCs in the cerebral circulation

(Alborch et al., 1995).

The role of T-type VOCCs in contraction of vascular smooth muscle has been

controversial mainly because these channels transmit tiny and transient, rather

than persistent, Ca2+ currents and are more sensitive to depolarization. The

window current, defined as the voltage range over which the channel has the

capacity to provide persistent calcium influx, for T-type VOCC ranges from -65

to -45 mV (-30 to 0 mV for L-type VOCC) (Kuo et al., 2011). There is strong

evidence from electrophysiological and molecular studies supporting the

existence of T-type VOCCs in cerebral arteries. To illustrate, T-type VOCCs

(along with N-type VOCC) were identified using patch clamp studies in vascular

smooth muscle cells isolated from dog basilar arteries (Navarro-Gonzalez et al.,

2009).

Expression of T-type VOCCs in renal (Poulsen et al., 2011), coronary (Küng et

al., 1995) and mesenteric arteries (Gustafsson et al., 2001) has been previously

described. Similarly, Kuo et al. (2010) found expression of mRNA and proteins

of T-type (Cav3.1 and Cav3.2) and L-type (Cav1.2) in smooth muscle cells of

adult rat basilar and middle cerebral arteries and their branches. They also

reported that about 20% of the Ca2+ current in smooth muscle cells of major

cerebral arteries and about 45% of the current in smooth muscle cells of their

branches were constituted by T-channels.

The HVA and LVA calcium channels might be responsible for different functions

in cerebral arteries - L-type VOCC playing a major role in vasomotion and

essentially responsible for contraction, while T-type VOCC control basal

vascular tone (Navarro-Gonzalez et al., 2009). Incubation of basilar arteries

with nifedipine abolishes vasomotion without affecting the vascular tone.

Application of depolarizing current in the presence of nifedipine, however,

caused immediate vasorelaxation. On the other hand, three T-channel

antagonists (mibefradil, pimozide and flunarizine), unlike nifedipine, relaxed

the basal tone of basilar arteries, further supporting the role of T-channels in

Page 66: Cerebrovascular effects of vasoactive drugs

General introduction

42

maintaining resting tone of the cerebral arteries. In addition, vasoconstriction

induced by 40 mM KCl in the presence of an L-channel blocker was significantly

inhibited by mibefradil.

Electrophysiological and molecular studies previously revealed the presence of

mRNA and protein for L-(Cav1.2 and Cav1.3), N- (Cav2.2) and T-type (Cav3.1 and

Cav3.3) α1 Ca2+ channel subunits (Nikitina et al., 2007) in canine basilar arteries.

Nimodipine and mibefradil relaxed intact arteries under isometric tension while

-agatoxin IVA, -conotoxin GVIA or SNX-482 did not relax these arteries. This

confirms a functional role of L- and T-type, not N-type, VOCC in canine basilar

arteries.

Page 67: Cerebrovascular effects of vasoactive drugs

General introduction

43

Therapeutic options for the management of cerebral

blood flow disorders

Various therapeutic agents have been tested as treatment modalities to prevent

or minimize the neurological morbidities and mortalities associated with the

disorders of the cerebral blood flow. Delayed cerebral vasospasm is among the

most widely investigated as far as pharmacotherapy of the cerebral circulation

disorders is concerned.

Treatment of cerebral vasospasm aims to reduce neurological damages due to

ischaemia. The commonly employed treatment modalities include

haemodynamic management, endovascular techniques and oral nimodipine

pharmacotherapy (Rahimi et al., 2006; Al-Tamimi et al., 2010). Haemodynamic

management constitutes the triple H therapy (induced hypertension,

haemodilution and hypervolaemia). It was designed to improve cerebral

perfusion by increasing blood pressure or cerebral perfusion pressure and

volume expansion. The explanation for haemodynamic management is that

vasospasm impairs cerebral autoregulation, thus increasing perfusion pressure

will passively increase cerebral blood flow according to the Hagen-Poiseuille

law. Although triple H therapy improves cerebral perfusion its benefit in

decreasing neurological deficits and mortality is not evident (Castanares-

Zapatero and Hantson, 2011). Further, triple H therapy is associated with other

morbidities such as fluid overload, pulmonary oedema, hyponatremia,

congestive heart failure and myocardial infarction (Al-Tamimi et al., 2010).

Nimodipine is the only L-type calcium channel blocker in clinical use for

cerebral vasospasm to date offering a modest clinical outcome with negligible

effect on the angiographic vasospasm (Pickard et al., 1989). Whether the

efficacy of nimodipine in preventing secondary ischaemia is due to its ability to

cross the blood brain barrier or its effect on other calcium channels is not

clearly known. Nimodipine may protect neuronal death by decreasing calcium

influx (Al-Tamimi et al., 2010). Nonetheless, the possibility that nimodipine

may dilate the penetrating arteries and arterioles rather than the pial arteries

Page 68: Cerebrovascular effects of vasoactive drugs

General introduction

44

cannot be ruled out. Despite the improvement in cerebral vasospasm

demonstrated by several other drugs tested thus far, most did not modify

clinical outcomes. Among the commonly studied agents are neuroprotective

agents (such as corticosteroids), statins, magnesium sulphate, nitric oxide

donors and endothelin antagonists (Castanares-Zapatero and Hantson, 2011).

Recently, clazosentan (selective ETA antagonist), which has been in a phase III

clinical trial (CONSCIOUS II and III), decreased postaneurysmal subarachnoid

haemorrhage vasospasm-related morbidity and mortality, but did not improve

outcome (extended Glasgow Outcome Scale) (Macdonald et al., 2012).

Limited progress has been made in the pharmacological treatment of ischaemic

stroke as well. The current approach in the management of acute ischaemic

stroke mainly employs an anticoagulant or a thrombolytic agent. As such,

aspirin and recombinant tissue plasminogen activator are among the few

therapeutic agents with demonstrated efficacy in improving clinical outcomes

(Ducruet et al., 2009). Statins are also found to be fairly effective in prevention

of ischaemic stroke, a benefit independent of their inhibition of cholesterol

synthesis. Statins prevent brain tissue damage by maintaining cerebral blood

flow through increasing nitric oxide production (Terpolilli et al., 2012). While

various agents with thrombolytic activity and neuroprotective benefits are

currently being investigated for cerebral vasospasm (Kikuchi et al., 2014;

Logallo et al., 2014), the quest for effective therapeutic agents is unanswered.

Understanding the responses of cerebral circulation to pharmacological

stimulation and the mechanisms involves is vital to for the development of

novel drugs that can effectively be used for prevention and treatment of the

cerebrovascular disorders. Thus, the current study attempts to analyse the

responses of cerebral circulation to activations of different pharmacological

targets.

Page 69: Cerebrovascular effects of vasoactive drugs

General introduction

45

Objectives of the study

The goal of this study is to investigate the responses of cerebral arteries to

pharmacological stimulation in isolated arteries, animal models and human

subjects. Specifically, the study aims to analyse the:

Reactivity of cerebral arteries isolated from different regions of the

rat cerebral circulation;

Mechanisms involved in adrenoceptor-mediated responses in rat

isolated cerebral arteries;

Effects of endothelin-1 on rat cerebral arteries and the roles of

voltage-operated and non-voltage-operated calcium channels in

mediating the responses;

In vivo responses of cerebral arteries to systemically-administered

vasoactive agent in the rat; and

Effects of vasopressor administration on cerebral oxygen saturation

during anaesthesia in supine and beach chair positions in patients.

Page 70: Cerebrovascular effects of vasoactive drugs
Page 71: Cerebrovascular effects of vasoactive drugs

47

Methods of in vitro small artery

reactivity studies

Page 72: Cerebrovascular effects of vasoactive drugs

Methods

48

Introduction

Most of the knowledge regarding effects of vasoactive agents on blood vessels

and their targets has been generated in vitro in isolated tissue preparations, a

technique more than a hundred years old. Various techniques have been

employed to characterise the responses of large arteries isolated from various

species in an artificially controlled condition without the influence of

homeostatic mechanisms such as autonomic nervous system (Moulds, 1983;

Spiers and Padmanabhan, 2005). In most cases, the active force induced in the

arteries is measured where the vessel diameter is held constant (isometric) or

change in diameter is measured under constant pressure (isobaric) or constant

tension (isotonic). In vitro studies are the most reliable methods to accurately

generate agonist concentration-response curves to various vasoactive

substances in blood vessels. Although attempts have been made to simulate the

in vivo parameters (temperature, electrolyte composition, oxygen, pH and

transmural pressure), it is impossible to mimic the exact physiological

conditions (Yu et al., 2003).

Previous studies have been commonly conducted in spiral strips or ring

preparations of large arteries of humans and animals set up in organ baths.

Following the understanding of the major contribution of small resistance

arteries to total peripheral resistance, however, there has been a growing

interest in investigating the contractility of these blood vessels in normal and

various disease states. Several investigators attempted to isolate small arteries

and characterise the responses using their own custom-made devices. For

example, the variation in pharmacological response between different arterial

beds was investigated by Bohr et al. (1961) in smooth muscle preparations of

small resistance arteries isolated from various tissues. Rabbit or dog small

resistance arteries (internal diameter 200-300 µm) isolated from mesentery,

kidney, lung and brain were sliced into helical strips and mounted on a

displacement transducer that was able to record the phasic change in tension.

The contractile responses elicited by different vasoactive agents in arteries

isolated from different organs were then compared (Figure 2-1).

Page 73: Cerebrovascular effects of vasoactive drugs

Methods

49

While this endeavour had advanced technical capability to characterise

pharmacological agents in small resistance arteries in vitro, there were still

concerns about the structural and functional integrity of the vessels. Bevan and

Osher (1972) later described an improved method to measure the wall tension

in intact cylindrical segments of small arteries. The vessels were dissected and

mounted either on a metallic rod and a thin platinum wire (for arteries with

internal diameter >200 µm) or on a pair of thin platinum wires (for smaller

arteries) that were connected to tension gauge recorder (Figure 2-2). This

method allowed continuous measurement of the response in isolated small

resistance arteries with accuracy. To date, more advanced myographs with

better sensitivity are commercially available.

Myography is an in vitro method widely used to characterise functional

responses of isolated small resistance arteries of 100-500 µm internal diameter.

There are two types of myography techniques – wire and pressure myography.

Wire myography is based on measurement of the contraction force under

isometric conditions, while pressure myography mostly measures diameter of

the pressurized vessels under isobaric conditions. Both wire and pressure

myographs are widely used in in vitro investigation responses in small vessels,

but some arteries showed differences in the pressure-diameter relationship

(Lew and Angus, 1992). The response to certain vasoactive mediators may also

vary when the arteries are mounted in the wire or pressure myograph. To

illustrate, pressurised rat mesenteric arteries were found to be 5-6 fold more

sensitive to noradrenaline and phenylephrine than wire-mounted vessels (Buus

et al., 1994). Various versions of the wire myograph have been introduced since

it was initially developed about four decades ago by Mulvany and Halpern

(1976).The four-channel wire myograph (model 620M; Danish Myo Technology;

Aarhus, Denmark) is now widely used for in vitro characterisation of isolated

small resistance arteries because it allows a simultaneous investigation of four

arteries in separate baths (Figure 2-3). The complete myography set up consists

of a myograph with isometric transducers, a data converter interface

(PowerLab) and a computer with software that helps to record the contractile

force and analyse data.

Page 74: Cerebrovascular effects of vasoactive drugs

Methods

50

Figure 2-1: The first small artery wall tension measurement technique.

A. Preparation of arterial strips and mounting on a displacement transducer and B.

a typical recording comparing the contractile responses induced by vasoactive agents

in the mesenteric and renal arteries (from Bohr et al. (1961)).

Page 75: Cerebrovascular effects of vasoactive drugs

Methods

51

Figure 2-2: An early version of wall tension measurement in intact

cylindrical segments of small resistance arteries.

A. An artery (200 µm i.d.) connected to Statham strain gauge with a stainless steel

rod (1.5 cm long; 200 µm i.d.) and a platinum wire (100 µm i.d.); B. A small artery

connected to two bridges of supporting plates, one connected to the strain gauge,

with a pair of thin platinum wires (25 µm i.d.); and C. A typical trace of spontaneous

rhythmic activity of a small mesenteric artery (200 µm i.d.) (slightly modified from

Bevan and Osher (1972)).

Page 76: Cerebrovascular effects of vasoactive drugs

Methods

52

Figure 2-3: A wire myograph setup

The setup consists of a 4-channel wire myograph (620M; Danish Myo Technology;

Aarhus, Denmark) connected to a PowerLab (AD Instruments, Pty. Ltd, Bella Vista,

NSW, Australia) and a computer. A segment of a small artery is set up on a pair of

jaws with the help of two 40 µm wires. One of the jaws is connected to a micrometer

and the other is connected to an isometric force transducer. The tone of the arteries

was monitored using LabChart 7 software (AD Instruments, Pty. Ltd, Bella Vista,

NSW, Australia).

Page 77: Cerebrovascular effects of vasoactive drugs

Methods

53

Physiological salt solutions

Several physiological salt solutions (PSS) of slightly variable electrolyte

composition have been introduced by different researchers (

Table 2-1). Despite variation in composition between different PSS types, or

sometimes between the same PSS in different labs, they are all isotonic with

the blood. The solution is usually made by dissolving analytical grade

ingredients in purified water. The critical ions in most PSS are sodium,

potassium, bicarbonate and calcium. Most of the osmotic pressure is caused by

sodium and chloride ions. Bicarbonate is the principal component of the buffer

system controlling pH of the solution. In some cases, synthetic buffers such as

HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acidy be used instead of

bicarbonate. Calcium is needed for smooth muscle contraction. The

concentration of potassium can affect the tone of smooth muscle cells - low

potassium concentration causes membrane hyperpolarisation while high

concentration induces contraction of blood vessels through depolarization.

Phosphate and/or sulphate salts are added to increase the buffer capacity and

maintain normal anion balance. Some PSS contain EDTA

(ethylenediaminetetraamino acetic acid) for chelation of heavy metals that

may be present from salt impurities or leaching of metal surfaces. Glucose is

commonly included as a metabolic substrate. PSS containing bicarbonate are

often aerated continuously with a mixture of 95% O2 and 5% CO2 (carbogen).

Bicarbonate buffer is most effective in the presence of a high concentration of

CO2. High concentration of O2 (90-100%) is needed to compensate for the lack

of oxygen carriers such as haemoglobin (Moulds, 1983; Radnoti Glass

Technology, 2010)

Page 78: Cerebrovascular effects of vasoactive drugs

54

Table 2-1: The composition and application of common physiological salt solutions used in isolated tissue

experiments.

Frog Ringers Ringer Locke Tyrode Krebs-Henseleit McEwan

g/ml mM g/ml mM g/ml mM g/ml mM g/ml mM

NaCl 6.0-6.5 103-111 9.0 14 83.0 13.7 6.9 118 7.6 130

KCl 0.075-0.14 1-1.9 0.4 5.4 0.2 2.7 0.35 4.7 0.42 5.6

CaCl2 0.1-0.12 0.9-1.1 0.12-0.24 1.1-2.2 0.24 2.2 0.28 2.5 0.24 2.2

MgCl2 0.01-0.05 0.1-0.5

MgSO4.7H2O 0.29 1.2

NaH2PO4 0.01 0.1 0.05 0.04 0.16 1.2 0.14 1.0

NaHCO3 0.1-0.2 1.2-2.4 0.15-0.5 1.8-6.0 1.0 11.9 2.1 25 2.1 25

Glucose 0-2.0 0-11.1 1.0-2.0 5.5-11.5 1.0 5.5 2.0 11.1 2.0 11.1

Aeration Air Air or O2 Air or O2 or 5% CO2 + 95% O2

5% CO2 + 95% O2 5% CO2 + 95% O2

Uses Frog and other amphibian tissue

Mammalian perfused heart

Mammalian isolated smooth muscle

Tissue requiring good oxygenation (e.g., nerve, heart)

Adapted from Salmon (2014).

Page 79: Cerebrovascular effects of vasoactive drugs

Methods

55

Protocols for in vitro vascular studies

2.3.1. Preparation of Krebs’ PSS and dissolution of drugs

The Krebs’ solution was made by dissolving the following ingredients in a water

purified by double distillation and ion exchange technique (Milli-Q water)

(Angus and Wright, 2000) (Table 2-2). The concentration of Ca2+ was decreased

for cerebral artery experiments to minimize the occurrence of spontaneous

contractions. A 25x concentrated stock solution (Stock A) containing all

ingredients except sodium bicarbonate, glucose, CaCl2 and EDTA was initially

made and kept in cold storage (4oC) for up to one week at a time. The Stock A

was diluted on the day of each experiment; glucose and sodium bicarbonate

were added and after vigorously bubbling the solution with a mixture of 5% CO2

and O2 for 5 min, CaCl2 and EDTA were added. The solution was continuously

aerated with 95% O2 and 5% CO2 thereafter. In addition, an isotonic potassium

salt solution (KPSS) containing 124 mM K+ obtained by equimolar substitution of

NaCl with KCl in the Krebs’ PSS was similarly prepared. The KPSS was used to

fully depolarise the smooth muscle and contract the arteries. An isotonic

solution containing 40 mM K+, used for pre-contraction of the arteries in some

protocols, was made by replacing the corresponding amount of Na+ with K+ in

Krebs’ PSS. In experiments involving calcium-free PSS, the solution was made

without CaCl2 and EDTA was replaced by EGTA (ethylene glycol tetraacetic acid)

(1 mM).

All drugs were dissolved in water or other solvents according to the

manufacturers’ or suppliers’ instructions and diluted aliquots were made in

Milli-Q water just before use. At times, concentrated aliquots were stored in a

-20oC freezer and thawed before single use.

2.3.2. Animals

Experiments were approved by The University of Melbourne Animal Ethics

Committee in accordance with the Australian code for the care and use of

Page 80: Cerebrovascular effects of vasoactive drugs

Methods

56

animals for scientific purposes (8th edition, 2013, National Health and Medical

Research Council, Canberra). Rats were obtained from the Biomedical Animal

Facility (University of Melbourne) where they were housed in climate-controlled

conditions with a 12 h light/dark cycle and free access to normal pellet diet

and drinking water.

Table 2-2: The composition of Krebs’ physiological salt solution used in

this study

Ingredient Composition (mM)

Na+ 144

K+ 5.9

Ca2+ 1.5 (cerebral arteries)

2.5 (other arteries)

Mg2+ 1.2

Cl- 128.7

HCO3- 25

PO42- 1.18

SO42- 1.17

EDTA 0.026

Glucose 5.5

2.3.3. Extraction of the rat brain

Male or female Sprague-Dawley rats (300-350 g) were deeply anaesthetised by

spontaneous inhalation of 5% isoflurane (Baxter Healthcare Pty. Ltd.;

Toongabbie, NSW, Australia) in 95% O2 and killed by decapitation; the vertebral

column was cut close to the cranial bone to ease opening of the skull. The skin

and muscle on the dorsal part of the skull were removed with small scissors.

The skull bone was cut along the suture line with a special pair of angled iris

surgical scissors inserted from the back and the flap of bone on each side was

Page 81: Cerebrovascular effects of vasoactive drugs

Methods

57

removed with rongeurs until the whole brain was clearly visible. The dura

overlying the brain was then slightly lifted with fine pair of rat-tooth forceps

and cut with small sharp scissors. The olfactory bulb was disconnected by

passing a small spatula blade under the brain and gently pushing the bulbs to

the side. The brain was then lifted from the floor and the trigeminal nerve was

detached near the brain stem using the spatula. The brain was then removed

and placed in ice-cold Krebs’ physiological salt solution.

2.3.4. Dissection and setup of cerebral arteries

The brain was placed ventral side up in a petri dish containing ice-cold PSS and

continuously bubbled with a mixture of 5% O2 and 95% CO2. Fine dissecting

forceps and scissors were used to isolate the cerebral arteries. The surrounding

meningeal and brain tissues were cleared from the arteries under a dissection

microscope. The anterior cerebral artery, middle cerebral artery, posterior

communicating artery and basilar artery were carefully dissected and

immediately set up in the myograph (Figure 2-4). In some protocols mesenteric

arteries were dissected and set up to compare effects in the peripheral

circulation; the PSS used for mesenteric artery experiments contained 2.5 mM

CaCl2.

The vessels were cut into 2 mm long segments and mounted on two 40 µm

stainless steel wires in a myograph chamber (620M; Danish Myo Technology;

Aarhus, Denmark). One of the wires was connected to an isometric force

transducer while the other was connected to a screw micrometer used for

adjustment of passive force by varying the distance between the wires.

Measurement of vascular contractile force was recorded on a computer using

LabChart 7 software (AD Instruments, Pty. Ltd, Bella Vista, NSW, Australia).

The arteries were allowed to equilibrate for about 30 min in Krebs’ PSS at 37oC

(pH 7.4) with continuous aeration by a mixture of 95% O2 and 5% CO2.

Page 82: Cerebrovascular effects of vasoactive drugs

Methods

58

Figure 2-4: A photographic image of freshly isolated rat brain (ventral

surface).

The major cerebral arteries branching out from the Circle of Willis (anterior, middle

cerebral and posterior communicating arteries) and the basilar artery are shown.

2.3.5. Normalization

The aim of normalization is to set the initial passive conditions (resting

condition) under which the arteries give optimum responses upon

pharmacological stimulation or depolarization and to standardise the

experimental conditions. After resting for about 30 min, the arteries were

normalized while the contractile force was continuous measured. The

normalization was carried out by a stepwise distention of the tissue until the

tension equivalent to a transmural pressure of 100 mmHg was achieved using

the screw micrometer connected to one jaw of the myograph. The

normalization window (DMT module plugin) was opened by selecting the DMT

menu of the LabChart (Figure 2-5 and Figure 2-6). The vessel length, diameter

Page 83: Cerebrovascular effects of vasoactive drugs

Methods

59

of mounting wire, as well as the micrometer reading before (baseline) and after

every stretch were entered in the normalization window. The passive tension-

internal circumference curve was automatically generated and the internal

circumference of the vessel was estimated by the software. The internal

diameter of the artery was calculated from the corresponding internal

circumference at 100 mmHg.

𝐷𝟏𝟎𝟎 =𝑳𝟏𝟎𝟎

Where, D100 is an estimated lumen diameter and L100 represents the internal

circumference of the vessel where the point on the exponential curve of

effective transmural pressure was 100 mmHg.

The vessels were relaxed by adjusting the micrometer to an estimated reading

that corresponds to 90% of the internal diameter that was determined from the

curve. All experiments were conducted under the same passive pressure

(Mulvany and Halpern, 1977; Angus and Wright, 2000).

Page 84: Cerebrovascular effects of vasoactive drugs

Methods

60

Figure 2-5: A representative trace demonstrating normalization of the

basilar artery.

The vessel was stretched in a step-by-step fashion by rotating the micrometer

connected to one jaw of the myograph, each step lasting for about 1 min, in order

to achieve a target pressure of 13.3 KPa (100 mmHg). The artery was then relaxed

to a resting tone corresponding to 90% of the internal circumference (IC90).

Page 85: Cerebrovascular effects of vasoactive drugs

Methods

61

Figure 2-6: A screenshot display of a typical normalization window (DMT

module plugin) in LabChart7 (AD Instruments).

After the wires were brought together to touch each other the force reading was set

to zero on the myograph; the length of the vessel, diameter of the wire and the

baseline micrometer reading were entered in the space provided. The artery was

then stretched step-by-step and the micrometer readings were entered before each

next stretch, until the target pressure of 13.3 KPa (100 mmHg) was obtained. The

passive wall tension-internal circumference was automatically generated; the

internal circumference (IC100) and the 90% IC (IC90) (micrometer X1) were estimated

by the software.

2.3.6. Testing the effects of drugs

The viability of the arteries was initially tested by exposure to an isotonic

potassium salt solution (KPSS) containing 124 mM K+ obtained by equimolar

substitution of NaCl with KCl in the PSS. The KPSS-induced contraction was

Page 86: Cerebrovascular effects of vasoactive drugs

Methods

62

repeated after 5 min and the maximum response of the second contraction was

used as a reference for quantification of all vascular responses. Only vessels

with a KPSS-induced contraction of 2.5 mN or more were used for experimental

protocols. The presence of a functional endothelium was tested by the relaxant

response to acetylcholine (1-10 µM) in arteries pre-contracted with vasopressin

(0.1–1 nM) to 80-90% of the maximum KPSS-induced contraction. The

endothelium was considered intact when acetylcholine caused more than 50%

relaxation.

The effects of agonists were examined by addition of low-high concentration of

the agents with cumulative increments in half-log10 units until no further

response was elicited (maximum response). Responses were allowed to plateau

before addition of the next higher concentration of the drugs. The relaxation

responses were investigated in arteries pre-contracted to 80-100% of the KPSS-

induced maximum contraction using vasopressin or with K+ 40 mM isotonic

solution as required. In experiments involving target antagonism or enzyme

inhibition, the agents were added to the baths 20 min prior to the agonist unless

stated otherwise. The arteries were rinsed three times after every experiment

to wash out the drugs completely or once every 15 min between protocols.

2.3.7. Removal of endothelium

After normalization, the passive tone of the artery was relaxed (to about 0.5

mN). The bath chamber was temporarily removed from the myograph and a

straight and round section of a clean human hair was passed through the lumen

of the artery under a light microscope with high magnification. From the inner

surface of the vessel, the endothelium was rubbed off by slow back-and-forth

movement of the hair, without stretching the artery longitudinally, until all the

surface was covered. The chamber was mounted back on the myograph, the

PSS solution changed and the tissue was allowed to equilibrate. After 5-10 min,

the artery was stretched to its normalization pressure by adjusting the

micrometer and allowed to stabilize for 5-10 min. The vessel was then

contracted with KPSS to check its viability – tissues contracting less than 50% of

Page 87: Cerebrovascular effects of vasoactive drugs

Methods

63

the previous KPSS-induced contraction were not used for experiments.

Complete removal of the endothelium was confirmed by absence of relaxation

to acetylcholine 1-10 µM (Mulvany, 2004).

Collection and analysis of data

The absolute contraction force of an individual artery was initially collected on

a data pad provided by the LabChart software and expressed as percent of the

KPSS-induced contraction. All data were expressed as mean ± SEM. Data analysis

and sigmoid curve fitting (non-linear regression) were performed using

GraphPad Prism 5.0 (GraphPad software, San Diego, CA, USA). The pEC50 (-log10

EC50) value and the maximum response (Emax) elicited by the drugs were

derived from the sigmoid concentration-response curve. EC50 is defined as the

concentration of agonist that evokes a half-maximal response and pEC50 is the

negative logarithm of the EC50. Statistical comparison between groups of

arteries was performed using unpaired t test or one-way ANOVA followed by

Bonferroni’s, Tukey’s or Dunnett’s post-test, as appropriate. P<0.05 was

considered significant in all cases.

Page 88: Cerebrovascular effects of vasoactive drugs
Page 89: Cerebrovascular effects of vasoactive drugs

65

Effects of common vasoactive

agents in rat isolated cerebral

arteries

Page 90: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

66

Introduction

Cerebral arteries, like peripheral arteries, are reactive to pharmacological

stimulation owing to the presence of various receptors that mediate

constriction or dilatation. The sensitivity of cerebral arteries to endogenous

vasoconstrictor and vasodilator mediators is critical for maintenance of stable

cerebral blood flow in spite of changing peripheral arterial pressure (cerebral

pressure autoregulation). It is also important for regulating blood flow

according to the metabolic needs of the brain (flow-metabolism coupling)

(Peterson et al., 2011). An increased need or decreased supply of oxygen and

nutrients to the brain promotes cerebral artery dilatation in order to increase

the blood flow and supply more oxygen and nutrients to the brain. Conversely,

when there is less need or increased supply, often due to elevated peripheral

blood pressure, constriction of cerebral arteries limit the blood flow and thus

prevent brain damage (Paulson et al., 1990).

Endogenous mediators may be involved in the pathogenesis of different

cerebral blood flow disorders such as migraine or cerebral vasospasm (Crowley

et al., 2008). Most of the drugs employed in the treatment of these disorders

or new therapeutic agents under investigation target the tone of cerebral

arteries (Castanares-Zapatero and Hantson, 2011). In the same way, exogenous

vasoactive substances used for other conditions may affect blood flow to the

brain. Nevertheless, presence of functional blood-brain barrier is thought to

limit the direct effect of most systemically administered pharmacological

agents on the cerebrovascular smooth muscle (Ballabh et al., 2004). Vasoactive

agents may also indirectly affect the tone of cerebral arteries by inducing

systemic hypertension or hypotension, consequently activating the

autoregulatory vasoconstrictor or vasodilator response.

Several endogenously released factors affect the tone of vascular smooth

muscle cells (Table 3-1). The response of cerebral arteries to common

vasoactive drugs is generally similar to peripheral arteries, but some qualitative

and quantitative differences exist. Further, arteries located in different regions

Page 91: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

67

of the cerebral circulation may exhibit differential reactivity to the vasoactive

mediators. The aim of this chapter is to investigate the responses of arteries

isolated from different regions of the rat cerebral circulation to common

vasoactive mediators. In addition, previous studies on the effects of these

agents on cerebral arteries have been reviewed and compared with the results

of this study.

3.1.1. Effects of endothelial factors on cerebrovascular tone

The role of the endothelium in the regulation of cerebral circulation is discussed

in section 1.5.4. Briefly, vascular endothelium releases both constrictor and

dilator mediators. The most widely characterised constrictor agents produced

by the endothelium are endothelin (further discussion on endothelin is

presented in Chapter 5) and arachidonic acid metabolites such as thromboxane

A2, which is also produced by platelets and brain tissues. Thromboxane A2 is a

potent vasoconstrictor in cerebral as well as peripheral arteries (Ellis et al.,

1976; Ellis et al., 1977). Enhanced expression of thromboxane A2 receptors (TP)

is associated with a decrease in cerebral blood flow following subarachnoid

haemorrhage (Saema et al., 2010).

Nitric oxide, prostacyclin (PGI2) and endothelium-derived hyperpolarisation

(EDH) are the major dilators produced and released by the vascular endothelial

cells. Peptide dilators (e.g., bradykinin, substance P, calcitonin gene-related

peptide (CGRP) and arginine vasopressin) and certain neurotransmitters (e.g.,

acetylcholine and noradrenaline) induce vasodilatation by stimulating release

of endothelium-derived relaxing factors (Toda and Okamura, 1998). Deficiency

of endothelial vasodilator agents and/or increased production of

vasoconstrictor mediators have been implicated in the pathogenesis of cerebral

vasospasm and ischaemia (Suhardja, 2004).

3.1.2. The sympathetic nervous system

Cerebral arteries receive abundant noradrenaline-containing neurons from the

peripheral sympathetic nervous system. The majority of these neurons

Page 92: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

68

innervating the cerebral circulation originate from the superior cervical

ganglion. Some adrenergic projections originating from subcortical nuclei such

as the locus coeruleus also synapse on cerebral arteries (Raichle et al., 1975;

Micieli et al., 1994; Hamel, 2006). In terms of number of nerve axons per mass

of smooth muscle, cerebral arterial beds are probably more densely innervated

than other vascular beds. The nerve terminals contain similar amounts of

noradrenaline as those of well-innervated peripheral arteries and the release

and reuptake processes are not different (Edvinsson and Owman, 1974;

Edvinsson et al., 1993). Noradrenaline, a potent vasoconstrictor in peripheral

arteries, induces a weak constrictor effect in arteries isolated from certain

regions of the cerebral circulation. On the other hand, noradrenaline induces a

variable degree of relaxation in different cerebral arteries actively contracted

with an agonist. Consistent with these findings, in vivo sympathetic stimulation

barely affected cerebral blood flow under normal conditions. Heterogeneous

distribution of the constrictor and dilator adrenoceptors in smooth muscle and

endothelium of cerebral arteries may be responsible for the diversity of

responses to pharmacological stimulation. A further discussion of

cerebrovascular adrenoceptor stimulation is presented in Chapter 4.

3.1.3. The parasympathetic nervous system

It is well established that cerebral arteries are innervated by the extracerebral

parasympathetic nervous system originating from sphenopalatine, otic and

internal carotid ganglia. Biochemical investigations have shown the presence of

nerves containing acetylcholine in cerebral arteries. The distribution of

cholinergic nerves is similar to adrenergic nerves in cerebral arteries of most

species (Edvinsson and MacKenzie, 1976). In vitro or in situ application of

acetylcholine or cholinomimetic drugs can elicit a potent relaxation in cerebral

arteries (Edvinsson et al., 1977). However, the sensitivity of cerebral arteries

to exogenous acetylcholine varies among different species (Tsukahara et al.,

1986b). Acetylcholine causes relaxation of vascular smooth muscle cells by

releasing nitric oxide via activation of endothelial muscarinic (M3) receptors

(Dauphin and Hamel, 1990; Toda and Okamura, 1990; Parsons et al., 1991).

Page 93: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

69

Acetylcholine may also cause vasoconstriction of cerebral arteries by

stimulating M1 receptors located on smooth muscle cells (Dauphin et al., 1991).

The peripheral parasympathetic innervation is limited to the pial arteries, but

cholinergic neurons originating from the basal forebrain affect the cortical

microcirculation (Hamel, 2004).

3.1.4. 5-Hydroxytryptamine (5-HT)

Earlier histochemical and autoradiographic studies provided evidence that

nerves containing 5-HT (serotonin) are found in cerebral arteries. It was

subsequently demonstrated that major cerebral arteries of various species are

innervated by serotonergic nerves originating from the superior cervical ganglia

along with adrenergic neurons (Cowen et al., 1986; Lincoln, 1995). Intrinsic

serotonergic neurons projecting from raphe nucleus also innervate the cerebral

microcirculation (Cohen et al., 1996) indicating that 5-HT may be involved in

coupling cerebral blood flow to the metabolic needs of the brain (Nobler et al.,

1999). Although in vitro application of 5-HT induces a potent vasoconstrictor

effect mediated by 5-HT2A receptors in peripheral arteries, it has weak effects

in cerebral arteries. The 5-HT-induced constriction of cerebral arteries involves

activation of both 5-HT1B and 5-HT2A receptors (Kovacs et al., 2012) but 5-HT1B

receptors appear to be more important in cerebral arteries and are found to be

upregulated in subarachnoid haemorrhage (Ansar et al., 2007).

3.1.5. Neuropeptides

The cerebral arteries are also innervated by sensory nerve fibres, originating

largely from the trigeminal and pterygopalatine ganglion and containing

calcitonin gene-related peptide (CGRP), substance P, neurokinin A, pituitary

adenylate-cyclase activating peptide (PACAP) and vasoactive intestinal peptide

(VIP) (Ruskell and Simons, 1987; Tsai et al., 1988; Ando et al., 1990; Ando et

al., 1994; Baeres and Moller, 2004). The neuropeptides mostly induce in vitro

relaxation responses in cerebral arteries obtained from different species

(Edvinsson et al., 1985; Toda and Okamura, 1998). CGRP may be involved in the

Page 94: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

70

pathogenesis of migraine headache by inducing dilatation of cerebral and dural

blood vessels. Further, the antimigraine effects of triptans are related to

inhibition of CGRP release through activation of 5-HT1B/1D receptors in

trigeminovascular neurons (Tepper et al., 2002).

3.1.6. Bradykinin

Bradykinin belongs to a family of endogenous peptides called kinins which are

produced by the enzymatic action of kallikreins on kininogen precursors (Regoli

and Barabe, 1980). It has two types of G-protein coupled receptors - B1 and B2

receptors. Bradykinin relaxes blood vessels by activating B2 receptors

predominantly located on endothelial cells and inducing the release of nitric

oxide, cyclooxygenase products and EDH (Mombouli and Vanhoutte, 1995;

Golias et al., 2007; Rahman et al., 2014). It is a potent vasodilator peptide in

peripheral as well as cerebral arteries (Whalley and Wahl, 1983). Bradykinin

may also contract blood vessels by inducing release of endothelium-derived

contracting factors such as thromboxane A2 and prostaglandin H2 (Miyamoto et

al., 1999; Ihara et al., 2000). Bradykinin causes a variable degree of dilatation

in cerebral arteries isolated from different species (Görlach and Wahl, 1996).

Additionally, bradykinin increases the permeability of the blood-brain barrier

by opening the endothelial tight junctions (Wahl et al., 1999).

3.1.7. Vasopressin

Vasopressin is known to induce both constrictor and dilator effects in vascular

smooth muscle cells. The vasoconstrictor activity is caused by stimulation of

the vasopressin V1 receptors located on smooth muscle cells (Fernandez et al.,

2001). The dilator effect involves release of the endothelial nitric oxide via

activation of endothelial V1 receptors (Suzuki et al., 1992; Suzuki et al., 1994).

Vasopressin exerts a variable degree of constriction in cerebral arteries isolated

from different regions. This may be related to heterogeneous distribution of

the constrictor and dilator receptors in different arterial beds (Garcia-Villalon

et al., 1996). For instance, removal of endothelium enhanced the

Page 95: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

71

vasoconstrictor effect of vasopressin in rabbit basilar artery (Garcia-Villalon et

al., 1996), but not in human cerebral arteries (Martin de Aguilera et al., 1990).

Vasopressin may be involved in the development of cerebral vasospasm

following subarachnoid haemorrhage (Trandafir et al., 2004).

3.1.8. Angiotensin

It is widely accepted that angiotensin II causes constriction of peripheral as well

as cerebral arteries either by directly stimulating AT1 receptors located on

smooth muscle cells or eliciting release of endothelial prostaglandins in

different species (Wei et al., 1978; Edvinsson et al., 1979; Kanaide et al.,

2003). It can also induce endothelium-dependent relaxation by activating AT1

receptors located on vascular endothelium (Toda, 1984). Recent studies have

shown that AT2 receptors may also play role in the vasodilator effect of

angiotensin II (Carey et al., 2000). The vasoconstrictor effect of exogenous

angiotensin II in cerebral arteries can be attenuated due to simultaneous

activation of dilator receptors (Näveri et al., 1994; Vincent et al., 2005). The

AT1-mediated contraction of cerebral arteries has been found to be upregulated

in focal cerebral ischaemia (Stenman and Edvinsson, 2004; Ansar et al., 2007).

3.1.9. Soluble guanyly cyclase (sGC) stimulation/activation

Soluble guanylyl cyclase (sGC) is the primary target of the gaseous ligands NO

and CO (Evgenov et al., 2006; Bryan et al., 2009). Binding of NO to the haem

moiety activates sGC which then catalyses the conversion of GTP to cyclic GMP

(cGMP). cGMP induces vascular smooth muscle relaxation by a number of

mechanisms. These include inhibition of inositol-1,4,5-triphosphate generation,

enhancing efflux of cytosolic calcium, dephosphorylation of myosin light chain

kinase (MLCK), inhibition of calcium influx and potassium channel opening

(Priviero and Webb, 2010). Increased cGMP levels may also promote other

cellular functions such as smooth muscle proliferation, platelet aggregation,

and leukocyte recruitment (Zhong et al., 2011). Impaired NO/cGMP signalling

is involved in the aetiology of several cardiovascular disorders such as systemic

Page 96: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

72

arterial hypertension, pulmonary hypertension, coronary artery diseases and

peripheral vascular diseases including erectile dysfunction and atherosclerosis

(Stasch et al., 2011). A novel class of drugs directly acting on sGC has been

emerging during the last two decades. They can be subclassified into two

categories depending on the mode of action. The first category, called haem-

dependent sGC stimulators, constitutes drugs that activate sGC with intact

haem (e.g., YC-1, BAY 41-2272, BAY 41-8543, BAY 63-2521 and A-350619). They

sensitize sGC to physiological concentrations of NO depending on the presence

of reduced (ferrous) iron in the haem bound to sGC. The second group, known

as sGC activators, bind to and activate the enzyme without the need for haem

and NO binding (NO- and haem-independent) (Evgenov et al., 2006; Stasch et

al., 2011). The currently available sGC activators include cinaciguat (BAY 58-

2667), ataciguat (HMR 1766) and BAY 60-2770. Some of these drugs have

demonstrated promising potential for the treatment of cardiovascular diseases

(Schäfer et al., 2006; Weissmann et al., 2009; Andreas et al., 2010; Tamargo

et al., 2010).

Page 97: Cerebrovascular effects of vasoactive drugs

73

Table 3-1: Summary of the common mediators released by extrinsic and intrinsic perivascular nerves, their

respective receptors and the responses elicited in cerebral arteries.

Mediator Origin/source Receptor - location Vascular response

References

Peripheral (extrinsic)

NA Superior cervical ganglion α1 - SMC α2 - Endothelium β1, β2 - Endothelium

Constriction Dilatation Dilatation

(Edvinsson, 1975; Hamel, 2006)

ACH Pterygopalatine ganglion Otic ganglion

M5 - Endothelium Dilatation (Edvinsson, 1975); Hamner et al. (2012)

NO Pterygopalatine ganglion - Dilatation (Nozaki et al., 1993; Westcott and Segal, 2013)

CGRP Trigeminal ganglion Dilatation (Tsai et al., 1988; Edvinsson et al., 2001)

SP Multiple NK1R - Endothelium Dilatation (Shimizu et al., 1999) VIP Pterygopalatine ganglia

Otic ganglion VAPC1 - Endothelium Dilatation (Edvinsson et al., 2001)

NPY Superior cervical ganglion NPY1 - Endothelium Constriction (Nilsson et al., 1996; Edvinsson et al., 2001)

Central (intrinsic)

NA Locus coeruleus α1 - SMC α2 - Endothelium β1, β2 - Endothelium/SMC

Dilatation Dilatation Constriction

Cohen et al. (1997)

ACH Nucleus basalis M5 - Endothelium Dilatation Sato et al. (2001) 5-HT Raphe nucleus 5-HT1B - SMC Constriction (Lincoln, 1995) GABA Unidentified GABAA - SMC Dilatation Imai et al. (1991)

ACH, acetylcholine; CGRP, calcitonin gene-related peptide; GABA, gamma aminobutyric acid; 5-HT, 5-hydroxytryptamine; NA, noradrenaline; NO, nitric oxide; NPY, neuropeptide Y; SP, substance P; VIP, vasoactive intestinal peptide; and VSM, vascular smooth muscle.

Page 98: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

74

Materials and methods

3.2.1. Isolation of arteries

The major arteries were isolated from anterior (anterior and middle cerebral

arteries) and posterior (posterior communicating and basilar arteries) regions

of the rat cerebral circulation and the vasomotor responses to common

vasoactive agents were characterised using the wire myography technique as

previously described in Chapter 2. The concentration-contraction or

concentration-relaxation curves were constructed for each drug and the pEC50

values were derived whenever possible.

3.2.2. Experimental protocols

The viability of the arteries was initially assessed by contraction with KPSS, the

maximum value of which has been used as a reference to compare the vascular

responses induced by vasoactive drugs. Drugs were added to the baths in

cumulative concentrations where sufficient time was allowed for the responses

to plateau before adding the next higher concentration of the drug. Relaxant

effects of the drugs were similarly assessed in arteries pre-contracted with

vasopressin. All responses were expressed as the % of the KPSS-induced

maximum contraction.

3.2.3. Drugs and chemicals

The drugs used in this study were obtained from the following suppliers:

Acetylcholine, noradrenaline and 5-hydroxytryptamine (Sigma Aldrich, St Louis,

Missouri, USA); endothelin-1 (GenScript, Piscataway, NJ, USA); bradykinin and

vasopressin (AusPep, Parkville, Victoria, Australia); sodium nitroprusside

(Hospira Inc., Lake Forest, USA) and BAY 41-8543 (gift from Dr. Johannes-Peter

Stasch, Bayer Pharma AG, Berlin, Germany). All drugs were dissolved and/or

diluted in water except endothelin-1 and BAY 41-8543 which were initially

dissolved in 10% dimethylformamide and then diluted in water.

Page 99: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

75

Results

3.3.1. Effects of noradrenaline in cerebral arteries

Noradrenaline induced weak concentration-dependent contractile effects in rat

isolated anterior and middle cerebral arteries. The maximum contraction

(Emax) induced by noradrenaline was less than 20% of the KPSS-induced

maximum contraction in both arteries. The contraction pEC50 values for

noradrenaline were 5.7±0.1 (n=8) in the anterior cerebral artery and 5.6±0.1

(n=7) in the middle cerebral artery (Figure 3-1). The basilar and posterior

communicating arteries were not contracted by noradrenaline. In contrast,

noradrenaline induced relaxation responses in all four arteries when pre-

contracted with vasopressin (Figure 3-2). The relaxation pEC50 values of

noradrenaline were 6.2±0.1 (n=7), 6.5±0.2 (n=8), 6.8±0.3 (n=7) and 6.2±0.2

(n=6) in anterior cerebral, middle cerebral, posterior communicating and

basilar arteries, respectively.

Page 100: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

76

Figure 3-1: Contractile effects of noradrenaline in rat cerebral arteries.

Contractile effects of noradrenaline in rat isolated anterior cerebral, middle

cerebral, posterior communicating and basilar arteries. Data are shown as percent

of KPSS-induced maximum contraction. Vertical bars represent ±SEM (those not

shown are contained within the symbol); horizontal bars indicate EC50±SEM. n,

number of arteries from separate rats.

Page 101: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

77

Figure 3-2: Relaxant effects of noradrenaline in rat cerebral arteries.

Rat anterior cerebral, middle cerebral, posterior communicating and basilar arteries

were pre-contracted with vasopressin (AVP) and when the plateau was obtained,

cumulative concentrations of noradrenaline were added. Data are shown as percent

of KPSS-induced maximum contraction. Vertical bars represent ±SEM (those not

shown are contained within the symbol); horizontal bars indicate EC50±SEM. n,

number of arteries from separate rats.

Page 102: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

78

3.3.2. Contractile effects of endothelin-1 in rat cerebral

arteries

Endothelin-1 induced potent constrictor effects in rat anterior cerebral

cerebral, middle cerebral, posterior communicating and basilar arteries. The

middle cerebral and posterior communicating arteries tended to be more

sensitive to endothelin-1 than the anterior cerebral artery or basilar artery. The

pEC50 values of endothelin-1 were 8.4±0.06, 8.7±0.11, 8.8±0.14 and 8.3±0.05 in

anterior cerebral, middle cerebral, posterior communicating and basilar

arteries, respectively. The maximum effects caused by endothelin-1 in the

cerebral arteries ranged from 108±2% (n=11) (in basilar artery) to 122±2% (n=10)

(in anterior cerebral artery) of the KPSS-induced maximum contraction.

Page 103: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

79

Figure 3-3: Contractile effects of endothelin-1 in rat cerebral arteries.

Contractile effects of endothelin-1 in rat anterior cerebral, middle cerebral,

posterior communicating and basilar arteries. Data are shown as percent of KPSS-

induced maximum contraction. Vertical bars represent ± SEM (those not shown are

contained within the symbol); horizontal bars indicate EC50±SEM. n; number of

arteries from separate rats.

3.3.3. Contractile effects of 5-hydroxytryptamine (5-HT) in

rat cerebral arteries

The contractile effects of 5-HT were characterised in the rat middle cerebral

and basilar arteries. The contractile potency of 5-HT appeared to be similar in

the two arteries - the pEC50 values of 5-HT were 6.6±0.4 (n=4) in the middle

Page 104: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

80

cerebral and 6.8±0.03 in the basilar artery. However, the maximum contraction

induced by 5-HT was much greater in the rat middle cerebral artery (Emax

90±6%) than in the rat basilar artery which responded poorly (Emax 22±5% of

the KPSS-induced maximum contraction; p<0.05, unpaired t test) (Figure 3-4).

Figure 3-4: Contractile effects of 5-hydroxytryptamine (5-HT) in rat

middle cerebral and basilar arteries.

Data are shown as percent of KPSS-induced maximum contraction. Vertical bars

represent ±SEM (those not shown are contained within the symbol); horizontal bars

indicate EC50±SEM. n, number of arteries from separate rats.

3.3.4. Contractile effects of vasopressin in rat cerebral

arteries

Vasopressin induced a strong contraction in both rat middle cerebral and basilar

arteries (Figure 3-5). The maximum response induced by vasopressin was similar

in the two arteries - 110±8% in the middle cerebral and 100±2% of the KPSS-

induced maximum contraction in the basilar artery (p>0.05, unpaired t test).

However, vasopressin was more potent in the middle cerebral artery, pEC50

10.0±0.2 (n=4), than in the basilar artery, pEC50 9.1±0.1 (p<0.05, unpaired t

test; n=6). It was observed that some basilar arteries were resistant to

Page 105: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

81

vasopressin contraction or rapidly desensitized upon exposure to vasopressin

and their data were excluded from the analysis.

Figure 3-5: Contractile effects of vasopressin in rat middle cerebral and

basilar arteries.

Data are shown as percent of KPSS-induced maximum contraction. Vertical bars

represent ±SEM (those not shown are contained within the symbol); horizontal bars

indicate EC50±SEM. n, number of arteries from separate rats.

3.3.5. Effects of acetylcholine in rat cerebral arteries

Acetylcholine caused a concentration-dependent relaxation of rat isolated

middle cerebral and basilar arteries pre-contracted with vasopressin (Figure

3-6). The relaxation was more pronounced in basilar artery than in the middle

cerebral artery which was only weakly relaxed (EC50 could not be determined).

Acetylcholine induced a maximum relaxation of 83±8% of the pre-contraction

tone in basilar arteries with a pEC50 value of 6.2±0.2 (n=7). High concentrations

of acetylcholine (≥30 µM) induced a weak contraction in some arteries.

Page 106: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

82

Figure 3-6: Relaxant effects of acetylcholine in rat middle cerebral and

basilar arteries.

The arteries were pre-contracted with vasopressin (AVP) and when the plateau was

obtained, cumulative concentrations of acetylcholine were added. Data are shown

as percent of KPSS-induced maximum contraction. Vertical bars represent ±SEM

(those not shown are contained within the symbol); horizontal bars indicate

EC50±SEM. n, number of arteries from separate rats.

3.3.6. Relaxant effects of bradykinin in rat cerebral arteries

Bradykinin caused a potent relaxation in rat middle cerebral and basilar arteries

pre-contracted with vasopressin (Figure 3-7). The relaxation pEC50 value of

bradykinin was greater in the middle cerebral artery (8.1±0.3, n=5) than in the

basilar artery (7.2±0.2, n=4; p<0.05, unpaired t test). However, the maximum

relaxation induced by bradykinin was similar in the two arteries: 65±13% in

middle cerebral artery and 82±12% of the pre-contraction tone in the basilar

artery (p>0.05, unpaired t test). It appeared that the tone of middle cerebral

arteries did not relax further, or in some cases, started to constrict slightly

after a high concentration (10 µM) of bradykinin was added.

Page 107: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

83

Figure 3-7: Relaxant effects of bradykinin in rat middle cerebral and

basilar arteries.

The arteries were pre-contracted with vasopressin (AVP) and when the plateau was

obtained, cumulative concentrations of bradykinin were added. Data are shown as

percent of KPSS-induced maximum contraction. Vertical bars represent ±SEM (those

not shown are contained within the symbol); horizontal bars indicate EC50±SEM. n,

number of arteries from separate rats.

3.3.7. Relaxant effect of sodium nitroprusside in rat cerebral

arteries

Sodium nitroprusside induced a marked concentration-dependent relaxation in

middle cerebral and basilar arteries pre-contracted with vasopressin (Figure

3-8). While the basilar artery was completely relaxed by cumulative additions

of sodium nitroprusside, some contractile tone was left in the middle cerebral

artery. The relaxation potency of sodium nitroprusside was similar in middle

cerebral and basilar arteries with pEC50 values of 5.6±0.3 (n=5) and 6.2±0.3

(n=3), respectively (p>0.05, unpaired t test).

Page 108: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

84

Figure 3-8: Relaxant effects of sodium nitroprusside in rat middle

cerebral and basilar arteries.

The arteries were pre-contracted with vasopressin (AVP) and when the plateau was

obtained, cumulative concentrations of sodium nitroprusside were added. Data are

shown as percent of KPSS-induced maximum contraction. Vertical bars represent

±SEM (those not shown are contained within the symbol); horizontal bars indicate

EC50±SEM. n, number of arteries from separate rats.

3.3.8. The effects of BAY 41-8543 in rat cerebral arteries

The relaxant effects of BAY 41-8543, a soluble guanylyl cyclase stimulator, were

characterised in rat cerebral arteries. BAY 41-8543 caused concentration-

dependent relaxation of rat cerebral arteries isolated from different regions

(Figure 3-9). The pEC50 values were 8.3±0.1 (n=4), 7.6±0.2 (n=4), 7.7±0.2 (n=4)

and 8.2±0.2 (n=4) in anterior cerebral, middle cerebral, posterior cerebral and

basilar arteries, respectively. The potency of BAY 41-8543 was not significantly

different in the four arteries (p>0.05, one way ANOVA).

Page 109: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

85

Figure 3-9: Relaxant effects of BAY 41-8543 in rat cerebral arteries.

The anterior cerebral, middle cerebral, posterior communicating and basilar arteries

were pre-contracted with vasopressin (AVP) and when the plateau was obtained,

cumulative concentrations of BAY 41-8543 were added. Data are shown as percent

of KPSS-induced maximum contraction. Vertical bars represent ±SEM (those not

shown are contained within the symbol); horizontal bars indicate EC50±SEM. n,

number of arteries from separate rats.

Page 110: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

86

Discussion

The rat cerebral arteries were sensitive to most vasoactive drugs tested, similar

to other resistance arteries in the peripheral circulation. However, some

qualitative and quantitative differences in the response to vasoactive agents

existed between the cerebral and other arteries, as well as within the cerebral

arteries owing to the heterogeneous distribution of targets or because of

morphological differences.

Noradrenaline induced both constriction and relaxation responses that varied

according to the location of the artery. The constriction was induced only in

the anterior and middle cerebral arteries. This suggests that the distribution of

α1-adrenoceptors that mediate the constrictor response is limited to the

anterior or rostral region of the rat cerebral circulation. The magnitude of the

constrictor response, which was weak compared to peripheral vessels, may

reveal that the sympathetic nervous system has limited effects on cerebral

arteries. This is consistent with previous reports that the contraction induced

by noradrenaline was significantly less in cerebral arteries than peripheral

arteries (Hogestatt and Andersson, 1984). Vasodilator responses were induced

by noradrenaline in all arteries taken from different regions of the rat cerebral

circulation, demonstrating that the receptors mediating vasodilatation are

distributed throughout the major cerebral arteries. The mechanisms involved

in noradrenaline-induced responses in rat isolated cerebral arteries are

analysed in Chapter 4.

Endothelin-1 was shown to induce a potent vasoconstrictor response with no

significant regional variation in sensitivity between the four arteries. This

indicates that ETA receptors mediating contraction are present throughout the

rat cerebral cerebral circulation and suggests that the peptide may be involved

in pathogenesis of cerebral vasospasm. Further analysis of the mechanism

involved in endothelin-1-induced contraction of rat cerebral arteries is

presented in Chapter 5.

Page 111: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

87

5-Hydroxytryptamine induced potent constriction in rat cerebral arteries. The

response obtained in the middle cerebral artery (90%) was remarkably greater

than in the basilar artery (22%). The variation in response between arteries of

different regions of the cerebral circulation demonstrates heterogeneous

distribution of constrictor 5-HT receptors. Previous studies have shown disparity

in the magnitude of 5-HT-induced constriction of cerebral arteries obtained

from different species (Toda and Okamura, 1993). The constriction induced by

5-HT in cerebral arteries is mediated by 5-HT2B receptors (Deckert and Angus,

1992; Kaumann and Levy, 2006). Thus rat basilar arteries may have a sparse

population of the constrictor 5-HT2B receptors unlike the middle cerebral

arteries.

Vasopressin induced a strong constrictor response (100-110%) in rat cerebral

arteries, but with greater potency in the middle cerebral than in basilar

arteries. This reveals that the rat anterior cerebral circulation may have greater

sensitivity to vasopressin compared to the posterior circulation, as represented

by the basilar artery. Previous studies found that vasopressin caused potent

constriction in human and goat cerebral arteries (Lluch et al., 1984; Martin de

Aguilera et al., 1990), but endothelium-dependent relaxation was also reported

in canine basilar arteries which was converted to a weak constriction when the

endothelium was removed (Katusic et al., 1984; Suzuki et al., 1993). There may

be a similar dilator effect that attenuates the constrictor effect of vasopressin

in the rat basilar artery. However, this requires further investigation.

Acetylcholine relaxed cerebral arteries - greater relaxation was caused in the

basilar artery than in middle cerebral artery. This is an indication that

stimulation of the parasympathetic nervous system may increase cerebral blood

flow by dilating arteries in the rat. Acetylcholine has been reported to dilate

rabbit (Parsons et al., 1991), human (Tsukahara et al., 1986b) and dog basilar

arteries (Toda, 1979). However, it induced both dilator and constrictor

responses in dog (Tsukahara et al., 1986b) and pig cerebral arteries (Lee et al.,

1982). The variation in the acetylcholine-induced response between cerebral

arteries of different species, or sometimes between different vascular beds of

Page 112: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

88

the same species, may be due to heterogeneous distribution of muscarinic

receptors in endothelium mediating dilator responses and constrictor receptors

on the smooth muscle cells (Lee, 1982). Both M1 and M2 receptors may be

involved in mediating the constrictor and dilator responses depending on their

location on smooth muscle or endothelium (Garcia-Villalon et al., 1991;

Dauphin et al., 1994).

Bradykinin induced a dilator effect in the rat cerebral circulation with

significantly greater potency in the middle cerebral artery than in the basilar

artery. The current result is consistent with previously reported in vitro dilator

effects of bradykinin in cerebral arteries of different species (Wahl et al., 1983;

Whalley et al., 1983; Kamitani et al., 1985). It can be concluded that bradykinin

may be involved in the regulation of the basal tone of cerebral arteries by

promoting release of endothelial vasodilator mediators such as NO and

production of EDH.

The sensitivity of cerebral arteries to exogenous nitric oxide was also assessed.

Sodium nitroprusside, an inorganic nitric oxide-releasing drug, induced a

marked and sustained relaxation with similar potency in rat middle cerebral

and basilar arteries. This demonstrates the sensitivity of the healthy cerebral

arterial tissue to the relaxation effects of nitric oxide, a major regulator of

vascular tone and cerebral blood flow. Similar results have been previously

reported in cerebral arteries of various species (Kimura et al., 1998; Salom et

al., 1998; Salom et al., 1999; Schubert et al., 2004).

Similarly, stimulation of the sGC enzyme with BAY 41-8543 induced a potent

dilatation of the cerebral arteries. It can be inferred that basally-released nitric

oxide can relax contracted arteries if sGC is stimulated. This highlights the

critical role played by the NO/cGMP pathway in the regulation of basal tone of

cerebral arteries, and therefore, in the maintenance of cerebral blood flow.

Direct stimulation of sGC may be a new therapeutic option for the management

of cerebral ischaemia or any condition associated with compromised cerebral

Page 113: Cerebrovascular effects of vasoactive drugs

Common vasoactive agents

89

blood flow. However, sGC stimulators need to be further investigated in

cerebral arteries since only limited studies are available in this regard.

Overall, this study showed that cerebral arteries exhibit remarkable regional

variation in their responses to common vasoactive substances. Often, the

arteries were very sensitive to these agents, but in some cases they responded

weakly or not at all. Further, some mediators such as noradrenaline can induce

opposing effects depending on the type or location of the receptors stimulated.

Drugs which decrease blood flow to some regions such as the frontal cortex may

increase the flow or have no effect in other regions such as the brain stem.

Thus, results obtained from studies limited to only some parts of the cerebral

vascular bed should be examined carefully before conclusions are drawn for

generalised effects of drugs on cerebral blood flow.

Page 114: Cerebrovascular effects of vasoactive drugs
Page 115: Cerebrovascular effects of vasoactive drugs

91

Mechanisms involved in

adrenoceptor-mediated

responses in rat cerebral

arteries

Page 116: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

92

Introduction

The cerebral circulation receives abundant sympathetic innervation, mainly

originating from the superior cervical ganglion (Lee et al., 1976; Duckles, 1980;

Paulson et al., 1990; Lincoln, 1995; Mitsis et al., 2009; ter Laan et al., 2013).

Cerebral arteries also receive adrenergic projections from certain nuclei of the

central nervous system such as the locus coeruleus (Raichle et al., 1975; Micieli

et al., 1994; Hamel, 2006).

Electrical stimulation experiments and studies involving systemic

administration of sympathomimetic agents indicated that the sympathetic

nervous system may be involved in regulation of cerebral blood flow (Hamner

et al., 2010; ter Laan et al., 2013). However, the precise role of the

sympathetic nervous system in cerebral autoregulation is not clearly known.

Activation of the sympathetic nervous system with a cold pressor test did not

alter cerebral blood flow despite increasing arterial blood pressure. This finding

then led to an assertion that sympathetic activation may induce constriction of

cerebral arteries and thus prevent increases in cerebral blood flow (Roatta et

al., 1998). Other studies have shown that the sympathetic nervous system plays

a protective role by preventing increases in cerebral blood flow during acute

elevation of blood pressure (Heistad and Marcus, 1978; Tuor, 1992; Dunatov et

al., 2011) or maintaining it during acute hypotension (Ogoh et al., 2008). These

observations indicate that the sympathetic nervous system plays a dual role

inducing vasoconstriction under conditions of elevated blood pressure and

vasodilatation of cerebral arteries under hypotensive states. The constrictor

and dilator responses induced by sympathetic nerve activation may be

mediated by different adrenoceptor subtypes located in cerebral arteries.

Therefore, the location of specific adrenoceptors and their particular roles in

mediating these effects should be rigorously investigated in cerebral arteries.

Page 117: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

93

Previous studies have reported variable effects of noradrenaline in cerebral

arteries obtained from different species, along with limited mechanistic

studies. Noradrenaline generally induced variable degrees of contraction in

human, monkey, sheep, dog, cat, rabbit and rat middle cerebral arteries

(Edvinsson and Owman, 1974; Edvinsson et al., 1993). Relaxant effects of

noradrenaline have been observed in basilar arteries obtained from various

species including the rat (Winquist and Bohr, 1982; Edvinsson et al., 1993).

However, Tanishima (1983) reported that noradrenaline contracted the human

basilar artery. Both contractile and relaxant effects were induced by

catecholamines in the feline middle cerebral artery (Edvinsson and Owman,

1974). Variation in responses induced by noradrenaline in arteries obtained

from different regions of the cerebral circulation is probably related to the

distribution of adrenoceptors mediating vasoconstriction or vasodilatation. This

was demonstrated by differences in regional effects of noradrenaline in bovine

cerebral arteries where β-adrenoceptors mediated relaxation of the caudal

arteries and α-adrenoceptors mediated contraction of the rostral arteries.

Blockade of α-adrenoceptors converted the contraction to relaxation, while

inhibition of β-adrenoceptors attenuated the relaxant effect of noradrenaline

(Ayajiki and Toda, 1992).

While β-adrenoceptors are generally accepted to mediate relaxation of actively

contracted cerebral arteries (Edvinsson et al., 1993), the precise mechanisms

leading to vasodilatation are not clearly known. A study in rat isolated cerebral

arteries found that the relaxation induced by noradrenaline involves release of

nitric oxide through activation of the β-adrenoceptors located on the

endothelial cells (Hempelmann and Ziegler, 1993). Other studies, however,

indicated that activation of presynaptic β2-adrenoceptors located on nitrergic

neurons (previously known as non-adrenergic non-cholinergic neurons) releases

nitric oxide (Zhang et al., 1998; Lee et al., 2000; Chang et al., 2012). The

involvement of other factors such as prostacyclin or endothelium-derived

hyperpolarisation in mediating relaxation of cerebral arteries induced by β-

adrenoceptor activation and their possible connection with specific β-

adrenoceptor subtype is unknown. The relationship between changes in

Page 118: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

94

sympathetic activity and aetiology of cerebrovascular diseases such as the

vasospasm that often occurs following subarachnoid haemorrhage is poorly

understood. Sympathetic activity has reportedly been elevated in patients with

subarachnoid haemorrhage (Naredi et al., 2000; Lambert et al., 2002), but the

link between sympathetic activation and development of cerebral vasospasm

has not been established (Moussouttas et al., 2012). Damage to the vascular

endothelium or uncoupling of nitric oxide synthase during subarachnoid

haemorrhage (Sabri et al., 2011) may increase sensitivity of cerebral arteries

to vasoconstrictor agents including noradrenaline. Thus, a thorough

investigation of the mechanisms involved in adrenoceptor-mediated responses

in cerebral arteries is important to understand the process leading to the

development of cerebrovascular disorders.

The aim of this study was to analyse the regional variation in responses induced

by adrenoceptor activation, the adrenoceptor subtypes involved, as well as the

role of endothelium-derived relaxing factors in mediating the relaxant

responses in arterial segments isolated from different regions of the rat

cerebral circulation.

Page 119: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

95

Materials and methods

4.2.1. Methods

The responses of cerebral arteries obtained from different regions of the rat

cerebral circulation were analysed using wire myography as described in

Chapter 2.

4.2.2. Experimental protocols

Experiments were conducted as described in Chapter 2, unless specified

otherwise. Briefly, the viability of the arteries was initially assessed by

contraction with KPSS, the maximum value of which has been used as a

reference to compare the vascular responses induced by vasoactive drugs. Drugs

were added to the baths in cumulative concentrations where sufficient time

was allowed for the responses to plateau before adding the next higher

concentration of the drug, except where single concentrations of the drugs

were tested. In protocols involving assessment of relaxant effects, arteries

were pre-contracted to about 80-100% of the KPSS-induced maximum

contraction using vasopressin followed by addition of the test drugs. The

inhibitors or antagonists were added 20-25 min before testing an agonist. All

responses were expressed as the % of the KPSS-induced maximum contraction.

4.2.3. Drugs and chemicals

The drugs used in this study were obtained from the following suppliers: (-)-

Adrenaline, atenolol, formoterol, (-)-isoprenaline, (-)-noradrenaline,

methoxamine, N-nitro-L-arginine methyl ester (L-NAME) and prazosin (Sigma

Aldrich, St Louis, Missouri, USA); ICI 118,551 (Tocris Bioscience, Bristol, UK);

propranolol (Imperial Chemical Industries Ltd, Macclesfield, UK); and apamin,

charybdotoxin and vasopressin (AusPep, Parkville, Victoria, Australia).

Page 120: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

96

Results

4.3.1. Contractile effects of α1-adrenoceptor stimulation in

cerebral arteries

The effects of α1-adrenoceptor stimulation in arteries taken from different

region of the rat cerebral circulation were investigated using the α1-

adrenoceptor selective agonist methoxamine. Methoxamine caused

concentration-dependent contraction of the rat anterior (Figure 4-1A) and

middle cerebral (Figure 4-1B) arteries. The potency of methoxamine was similar

in the anterior cerebral (pEC50 5.2±0.08, n=4) and middle cerebral (pEC50

5.3±0.1, n=6) arteries. Similarly, the maximum contractile effects elicited by

methoxamine were not significantly different in the anterior (79±5%) and

middle cerebral (58±8% of the KPSS-induced maximum contraction, P>0.05)

arteries. However, methoxamine failed to contract either the posterior

communicating (Figure 4-1C) or the basilar (Figure 4-1D) artery isolated from

the rat.

4.3.2. Relaxant effects of β-adrenoceptor stimulation in

cerebral arteries

Stimulation of β-adrenoceptors using isoprenaline (nonselective β1- and β2-

adrenoceptor agonist) caused concentration-dependent relaxation of rat

cerebral arteries actively constricted with vasopressin (1-3 nM for basilar, or

0.1-1 nM for other arteries). Isoprenaline (1 nM-1 M) induced similar degrees

of relaxation in the rat anterior cerebral (Figure 4-2A), middle cerebral (Figure

4-2B), posterior communicating (Figure 4-2C) and basilar arteries (Figure 4-2D)

pre-contracted with vasopressin. There was no significant difference between

the relaxation potencies of isoprenaline in the four arteries (P>0.05) – the pEC50

values were 7.4±0.3 (n=5), 7.4±0.1 (n=7), 7.1±0.4 (n=5) and 6.8±0.2 (n=7) in

anterior cerebral, middle cerebral, posterior communicating and basilar

arteries, respectively.

Page 121: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

97

Figure 4-1: Effects of α1-adrenoceptor stimulation in rat cerebral

arteries.

Effects of α1-adrenoceptor activation with methoxamine in A) anterior cerebral; B)

middle cerebral; C) posterior communicating; and D) basilar arteries isolated from

the rat. Data are shown as percent of KPSS-induced maximum contraction. Vertical

bars represent ±SEM (those not shown are contained within the symbol); horizontal

bars indicate EC50±SEM. n, number of arteries from separate rats.

Page 122: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

98

Figure 4-2: Relaxant effects of β-adrenoceptor stimulation in cerebral

arteries

Relaxant effect of isoprenaline in rat A) anterior cerebral; B) middle cerebral; C)

posterior communicating; and D) basilar arteries pre-contracted with vasopressin

(AVP) 0.1-3 nM. Data are shown as percent of KPSS-induced maximum contraction.

Vertical bars represent ±SEM (those not shown are contained within the symbol);

horizontal bars indicate EC50±SEM. n, number of arteries from separate rats.

Page 123: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

99

4.3.3. Effects of endothelium removal and inhibition of nitric

oxide production on isoprenaline-induced relaxation of

cerebral arteries

The roles of endothelium and nitric oxide in mediating the relaxation induced

by β-adrenoceptor stimulation with isoprenaline were investigated in arteries

isolated from anterior (middle cerebral artery) and posterior regions (basilar

artery) of the rat cerebral circulation. Physical removal of the endothelium did

not affect the relaxant effect of isoprenaline (1 nM-1 M) in the middle cerebral

artery (Figure 4-3A). However, removal of endothelium inhibited the

isoprenaline-induced relaxation in rat basilar artery (Figure 4-3B). The

relaxation pEC50 value of isoprenaline in the endothelium-denuded middle

cerebral artery was not significantly different from corresponding arteries with

intact endothelium (P>0.05). Inhibition of nitric oxide production with L-NAME

100 µM did not significantly affect the isoprenaline-induced relaxation either in

middle cerebral (Figure 4-4A) or basilar arteries (Figure 4-4B) isolated from the

rat. The relaxation potency of isoprenaline in the presence of L-NAME was

similar in the middle cerebral (pEC50 6.9±0.3, n=7) and basilar arteries (pEC50

6.8±0.2, n=7; P>0.05).

Page 124: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

100

Figure 4-3: Effects of endothelium removal on isoprenaline-induced

relaxation of cerebral arteries.

Relaxant effect of isoprenaline in rat A) middle cerebral; and B) basilar arteries with

or without endothelium. The arteries were pre-contracted with vasopressin (AVP)

0.1-3 nM and subsequently relaxed with cumulative addition of isoprenaline. The

endothelium was removed from the arteries by rubbing the lumen with a human hair.

Data are shown as percent of KPSS-induced maximum contraction. Vertical bars

represent ±SEM (those not shown are contained within the symbol); horizontal bars

indicate EC50±SEM. n, number of arteries from separate rats.

Page 125: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

101

Figure 4-4: Effects of inhibition of nitric oxide production on

isoprenaline-induced relaxation of cerebral arteries.

The effects of nitric oxide production inhibition with N-nitro-L-arginine methyl ester

(L-NAME) on isoprenaline-induced relaxation of rat A) middle cerebral; and B) basilar

arteries. The arteries were pre-contracted with vasopressin (AVP) 0.1-3 nM and

subsequently relaxed with cumulative addition of isoprenaline. L-NAME was added

20 min prior to AVP contraction. Data are shown as percent of KPSS-induced

maximum contraction. Vertical bars represent ±SEM (those not shown are contained

within the symbol); horizontal bars indicate EC50±SEM. n, number of arteries from

separate rats.

4.3.4. Contractile effects of noradrenaline and the role of

endothelium and nitric oxide

The contractile effects of noradrenaline, an endogenous neurotransmitter

released by sympathetic nerves, were characterised in rat cerebral arteries. As

with methoxamine, noradrenaline induced contraction only in anterior and

middle cerebral arteries. However, the contractile responses induced by

noradrenaline were relatively weak Emax ~17% of the KPSS-induced maximum

contraction in anterior (Figure 4-5A) and middle cerebral arteries (Figure 4-5B).

There was no significant difference in pEC50 values of noradrenaline in anterior

Page 126: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

102

cerebral (5.6±0.06, n=7) and middle cerebral arteries (5.7±0.12, n=7; P>0.05).

However, noradrenaline did not contract rat isolated posterior communicating

(Figure 4-5C) and basilar arteries (Figure 4-5D). Removal of endothelium from

the middle cerebral artery did not affect the contraction induced by

noradrenaline (Figure 4-6). However, nonselective inhibition of the nitric oxide

synthase enzyme with L-NAME 100 µM significantly increased the contraction

induced by noradrenaline in both anterior (Emax 69±10%) (Figure 4-7A) and

middle cerebral arteries (Emax 69±5%) (Figure 4-7B) compared to the weak

response obtained in the absence of L-NAME (P>0.05). No contractile effects of

noradrenaline were observed in posterior communicating or basilar arteries

pre-treated with L-NAME.

Page 127: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

103

Figure 4-5: Contractile effects of noradrenaline in rat cerebral arteries

The effects of noradrenaline in rat isolated A) anterior cerebral; B) middle cerebral;

C) posterior communicating; and D) basilar arteries. Data are shown as percent of

KPSS-induced maximum contraction. Vertical bars represent ±SEM (those not shown

are contained within the symbol); horizontal bars indicate EC50±SEM. n, number of

arteries from separate rats.

Page 128: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

104

Figure 4-6: Effect of endothelium removal on the noradrenaline-induced

contraction of middle cerebral artery.

Contractile effects of noradrenaline in the rat middle cerebral artery with or without

endothelium. The endothelium was removed from the arteries by rubbing the lumen

with a human hair. Data are shown as percent KPSS-induced maximum contraction.

Vertical bars represent ±SEM (those not shown are contained within the symbol). n,

number of arteries from separate rats.

Page 129: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

105

Figure 4-7: Effects of inhibition of nitric oxide production on

noradrenaline-induced contraction of cerebral arteries.

The effects of nitric oxide production inhibition with N-nitro-L-arginine methyl ester

(L-NAME 100 M) on noradrenaline-induced contraction of rat A) anterior; and B)

middle cerebral arteries. L-NAME was added 20 min prior to the noradrenaline

contraction. Data are shown as percent of KPSS-induced maximum contraction.

Vertical bars represent ±SEM (those not shown are contained within the symbol);

horizontal bars indicate EC50±SEM. n, number of arteries from separate rats.

4.3.5. Relaxant effects of noradrenaline in rat cerebral

arteries and the role of endothelium and nitric oxide

Noradrenaline elicited a concentration-dependent relaxation of rat cerebral

arteries actively contracted with vasopressin. The maximum relaxation

response to noradrenaline appeared to be lesser in anterior (~60%) (Figure 4-8A)

and middle cerebral arteries (~52%) (Figure 4-8B) than in posterior

communicating arteries (~69%) (Figure 4-8C) and basilar arteries (~76% of the

pre-contraction tone) (Figure 4-7D). The relaxation pEC50 values of

noradrenaline were 6.5±0.2 (n=7), 6.2±0.1 (n=7), 6.8±0.3 (n=8) and 6.2±0.2

(n=6) in anterior cerebral, middle cerebral, posterior communicating and

Page 130: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

106

basilar arteries, respectively. No significant difference was found in relaxant

potency of noradrenaline between the four arteries (P>0.05). Removal of the

endothelium inhibited the relaxant effects of noradrenaline in the basilar artery

as shown in Figure 4-9. The absence of spontaneous relaxation in the control

arteries left contracted during experimental period further confirmed that the

relaxation responses were induced by noradrenaline. Pre-treating the arteries

with L-NAME 100 µM inhibited the relaxant effects of noradrenaline in rat

cerebral arteries (Figure 4-10A-D). Noradrenaline slightly increased the pre-

contraction tone in anterior and middle cerebral arteries pre-treated with L-

NAME as shown in Figure 4-10A and B.

Page 131: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

107

Figure 4-8: Relaxant effects of noradrenaline in rat cerebral arteries.

Relaxant effects of noradrenaline in rat isolated A) anterior cerebral; B) middle

cerebral; C) posterior communicating; and D) basilar arteries pre-contracted with

vasopressin (AVP) 0.1-3 nM. Arteries in the treatment group were relaxed by

cumulative addition of noradrenaline, while those in the time control group were

left contracted during this time. Data are shown as percent of KPSS-induced

maximum contraction. Vertical bars represent ±SEM (those not shown are contained

within the symbol); horizontal bars indicate EC50±SEM. n, number of arteries from

separate rats.

Page 132: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

108

Figure 4-9: Effects of endothelium removal on noradrenaline-induced

relaxation of rat basilar artery.

Relaxant effect of noradrenaline in rat basilar artery pre-contracted with

vasopressin (AVP) 0.1-3 nM. The endothelium was removed from the arteries by

rubbing the lumen with a human hair. Data are shown as percent of KPSS-induced

maximum contraction. Vertical bars represent ±SEM (those not shown are

contained within the symbol); horizontal bars indicate EC50±SEM. n, number of

arteries from separate rats.

Page 133: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

109

Figure 4-10: Effects of inhibition of nitric oxide production on the

noradrenaline-induced contraction of cerebral arteries.

The effect of inhibition of nitric oxide production with N-nitro-L-arginine methyl

ester (L-NAME) on noradrenaline-induced relaxation of rat A) anterior cerebral; B)

middle cerebral; C) posterior communicating; and D) basilar arteries pre-contracted

with vasopressin (AVP) 0.1-3 nM. L-NAME was added 20 min prior to the

noradrenaline contraction. Data are shown as percent of KPSS-induced maximum

contraction. Vertical bars represent ±SEM (those not shown are contained within the

symbol); horizontal bars indicate EC50±SEM. n, number of arteries from separate rats.

Page 134: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

110

4.3.6. Roles of α- and β-adrenoceptors in the responses

induced by noradrenaline in rat cerebral arteries

The roles of α- and β-adrenoceptors in the responses induced by noradrenaline

were further analysed in rat isolated cerebral arteries. The contractile

responses were analysed in middle cerebral artery, while the relaxant responses

were analysed in all four arteries in the presence of the adrenoceptor

antagonists prazosin, propranolol, atenolol or ICI 118551. Inhibition of α1-

adrenoceptors with prazosin 0.1 µM completely inhibited the contraction

induced by noradrenaline (10 nM-10 µM) in the rat middle cerebral artery

(Figure 4-11A). Pre-treatment of the arteries with propranolol 0.1 µM, a

nonselective β-adrenoceptor antagonist, increased the contractile response to

noradrenaline of the middle cerebral artery to about 52% of the KPSS-induced

maximum contraction (Figure 4-11B). Inhibition of β1-adrenoceptors with

atenolol 1 µM increased the contractile effect of noradrenaline in the middle

cerebral artery (Figure 4-11C). Similarly, pre-treating the arteries with the β2-

adrenoceptor antagonist ICI 118551 (1 µM) increased the contractile response

to noradrenaline (Figure 4-11D). Both atenolol and ICI 118551 caused similar

increases in the maximum contractile effects of noradrenaline (Emax 41-43% of

KPSS-induced contraction) in the rat middle cerebral artery. The pEC50 of

noradrenaline was not significantly changed by the antagonists (vehicle,

5.7±0.12; propranolol, 6.2±0.09; atenolol, 6.2±0.13; and ICI 118551, 6.0±0.13;

P>0.05). Stimulation of α2-adrenoceptors with clonidine (10 nM-1 µM) did not

induce any contraction in the rat middle cerebral artery (n=2; data not shown).

Non-selective inhibition of the β-adrenoceptors with propranolol 1 µM caused a

rightward shift of the noradrenaline concentration-relaxation curve in rat

cerebral arteries (Figure 4-12). Noradrenaline initially caused a small increase

of the pre-contraction tone in some anterior and middle cerebral arteries pre-

treated with propranolol (Figure 4-12A and B). However, propranolol diminished

the maximum relaxant effect of noradrenaline in these arteries. Specific

inhibition of β1-adrenoceptors using atenolol 1 µM also caused a rightward shift

of the concentration-relaxation curve without affecting the maximum response

Page 135: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

111

in the basilar artery (Figure 4-13A). Similarly, ICI 118551 0.1 µM, a β2-

adrenoceptor specific antagonist, shifted the noradrenaline concentration-

relaxation curve to the right as shown in Figure 4-13B. In another set of

experiments, the basilar artery was relaxed with selective agonists to

characterise the presence of β1- and β2-adrenoceptors. Adrenaline, a selective

β2-adrenoceptor agonist, caused a concentration-dependent relaxation of the

basilar artery with a pEC50 of 6.4±0.08 (n=4) as shown in Figure 4-14A. However,

another β2-selective agonist formoterol (0.1 nM-1 M) caused poor relaxant

effects in the basilar artery (Figure 4-14B). Likewise, prenalterol (10 nM-10 µM),

a β1-adrenoceptor partial agonist, did not relax basilar arteries pre-contracted

with vasopressin (n=2; data not shown). Presence of α2-adrenoceptors was

tested using a selective agonist clonidine (10 nM-1 µM). Clonidine did not cause

any relaxant effect in rat middle cerebral or basilar arteries pre-contracted

with vasopressin (Figure 4-15).

Page 136: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

112

Figure 4-11: Effects of adrenoceptor antagonism on the noradrenaline-

induced contraction in middle cerebral artery.

The effects of A) prazosin 0.1 µM; B) propranolol 0.1 µM; C) atenolol 1 µM; and D)

ICI 118551 1 µM pre-treatment on the noradrenaline-induced contraction of rat

middle cerebral arteries. The antagonists were added 20 min prior to noradrenaline

contraction. Data are shown as percent of KPSS-induced maximum contraction.

Vertical bars represent ±SEM (those not shown are contained within the symbol);

horizontal bars indicate EC50±SEM. n, number of arteries from separate rats. *P<0.05

compared to the vehicle group.

Page 137: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

113

Figure 4-12: Effects of β-adrenoceptor antagonism on the noradrenaline-

induced relaxation of cerebral arteries.

The effects of nonselective inhibition of β-adrenoceptors with propranolol 1 µM on

the noradrenaline-induced relaxation of rat A) anterior cerebral; B) middle cerebral;

C) posterior communicating; and D) basilar arteries pre-contracted with vasopressin

(AVP) 0.1-3 nM. Propranolol was added 20 min prior to AVP contraction. Data are

shown as percent of KPSS-induced maximum contraction. Vertical bars represent

±SEM (those not shown are contained within the symbol); horizontal bars indicate

EC50±SEM. n, number of arteries from separate rats.

Page 138: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

114

Figure 4-13: The roles of β1- and β2-adrenoceptors in noradrenaline-

induced relaxation of basilar arteries.

The effects of A) atenolol 1 µM; and B) ICI 118551 0.1 µM pre-treatment on the

noradrenaline-induced relaxation of the rat basilar artery pre-contracted with

vasopressin (AVP). The antagonists were added 20 min prior to AVP contraction. Data

are shown as percent of KPSS-induced maximum contraction. Vertical bars represent

±SEM (those not shown are contained within the symbol); horizontal bars indicate

EC50±SEM. n, number of arteries from separate rats.

Page 139: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

115

Figure 4-14: Relaxant effects of adrenaline and formoterol in rat

basilar arteries.

Relaxant effects of A) adrenaline; and B) formoterol in the rat basilar artery pre-

contracted with vasopressin (AVP). Arteries were relaxed by cumulative

concentrations of adrenaline or formoterol. Data are shown as percent of KPSS-

induced maximum contraction. Vertical bars represent ±SEM (those not shown are

contained within the symbol); horizontal bars indicate EC50±SEM. n, number of

arteries from separate rats.

Page 140: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

116

Figure 4-15: Relaxant effects of clonidine in rat cerebral arteries.

The effects of clonidine in rat A) middle cerebral; and B) basilar arteries pre-

contracted with vasopressin (AVP). There arteries were pre-contracted with

vasopressin (0.1-3 nM) and when the plateau was attained, clonidine was added in

cumulative concentrations. Data are shown as percent of KPSS-induced maximum

contraction. Vertical bars represent ± SEM (those not shown are contained within

the symbol). n, number of arteries from separate rats.

4.3.7. Involvement of the endothelium-derived

hyperpolarisation (EDH) in β-adrenoceptor-mediated

relaxation of cerebral arteries

Cerebral arteries were pre-contracted with a depolarizing K+ solution

(equimolar Krebs’ solution containing 40 mM K+) and subsequently relaxed by

cumulative addition of the nonselective β-adrenoceptor agonist isoprenaline

(Figure 4-16). Isoprenaline induced a slight relaxation in both middle cerebral

and basilar arteries. Addition of sodium nitroprusside 1-10 µM (an NO donor

drug) after isoprenaline caused a complete relaxation of the arteries. Inhibition

of EDH production in basilar arteries with a combination of apamin 50 nM and

charybdotoxin 50 nM slightly shifted the concentration-relaxation curve of

Page 141: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

117

noradrenaline, but the pEC50 was not significantly changed (Figure 4-17A). The

isoprenaline-induced relaxation of the basilar artery was, however, markedly

inhibited by the combination (the EC50 value could not be derived after the

inhibition) (Figure 4-17B). Combination of apamin 50 nM and charybdotoxin 50

nM in the presence of L-NAME 100 µM and indomethacin 3 µM only slightly

inhibited the isoprenaline-induced relaxation in the middle cerebral artery

(Figure 4-18A), but completely inhibited the relaxation in the rat basilar artery

(Figure 4-18B). A moderate to strong constriction was initially observed in some

middle cerebral arteries after addition of apamin and charybdotoxin, but

subsided in most arteries within 10 min.

Page 142: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

118

Figure 4-16: Relaxant effects of isoprenaline in cerebral arteries pre-

contracted with K+

Relaxant effects of isoprenaline in rat A) middle cerebral; and B) basilar arteries

pre-contracted with Krebs’ buffer solution containing K+ 40 mM (K40). Data are

shown as percent of KPSS-induced maximum contraction. Vertical bars represent ±

SEM (those not shown are contained within the symbol). n, number of arteries from

separate rats.

Page 143: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

119

Figure 4-17: Effects of apamin and charybdotoxin combination on the

relaxation induced by noradrenaline and isoprenaline in rat

basilar arteries.

Effects of apamin 50 nM and charybdotoxin (ChTx) 50 nM pre-treatment on A)

noradrenaline-induced; and B) isoprenaline-induced relaxation of rat basilar artery

pre-contracted with vasopressin (AVP). The antagonists (inhibitors) were added 20

min prior to AVP contraction. Data are shown as percent of KPSS-induced maximum

contraction. Vertical bars represent ± SEM (those not shown are contained within

the symbol); horizontal bars indicate EC50±SEM. n, number of arteries from separate

rats.

Page 144: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

120

Figure 4-18: Effects of endothelial-derived EDH, NO and PGI2 inhibition

on the isoprenaline-induced relaxation of cerebral arteries.

Effects of endothelium-derived hyperpolarisation (EDH), nitric oxide (NO) and

prostacyclin (PGI2) inhibition on isoprenaline-induced relaxation in the rat A) middle

cerebral; and B) basilar arteries pre-contracted with vasopressin (AVP). EDH

production was inhibited by using a combination of apamin (Apam) 50 nM and

charybdotoxin (ChTx) 50 nM. L-NAME 100 µM and indomethacin (Indo) 3 µM were

added to inhibit NO and PGI2 production. The antagonists (inhibitors) were added 20

min prior to AVP contraction. Data are shown as percent of KPSS-induced maximum

contraction. Vertical bars represent ± SEM (those not shown are contained within

the symbol); horizontal bars indicate EC50±SEM. n, number of arteries from separate

rats.

Page 145: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

121

Discussion

This chapter analysed the effects of adrenoceptor activation, the location and

subtypes of receptors involved and the role of endothelial factors in different

arteries of the rat cerebral circulation. Activation of adrenoceptors in rat

cerebral arteries caused heterogeneous responses depending on the regional

location of the arteries. Both contractile and relaxant effects were induced in

anterior and middle cerebral arteries, while only relaxant effects were induced

in posterior communicating and basilar arteries. The relaxation response in the

middle cerebral artery is endothelium-independent, but involved release of

endothelial NO and EDH in the basilar artery. α1-Adrenoceptors mediate the

contractile responses in rat anterior and middle cerebral arteries, while β1- and

β2-adrenoceptors mediate the relaxant effects in rat cerebral arteries.

Selective stimulation of α1-adrenoceptors with methoxamine contracted

arteries isolated from the anterior region of the rat cerebral circulation

anterior and middle cerebral arteries. However, no contractile response was

induced by methoxamine in arteries obtained from the caudal region of the rat

cerebral circulation, namely the posterior communicating and basilar arteries.

Noradrenaline elicited similar effects as methoxamine in cerebral arteries, but

the constrictor responses were relatively weak. The contractile responses were

inhibited by prazosin, a selective α1-adrenoceptor antagonist. These findings

indicate that the distribution of α1-adrenoceptors may be limited to arteries in

the anterior parts of the rat cerebral circulation. Stimulation of α2-

adrenoceptors with clonidine did not cause contraction in rat cerebral arteries.

Similar findings were previously reported by Hogestatt and Andersson (1984)

that noradrenaline induced an α1-adrenoceptor-mediated contraction in rat

middle cerebral artery, but not in the basilar artery. Similarly, noradrenaline

contracted bovine artery strips taken from anterior cerebral, middle cerebral

and internal carotid arteries through α1-adrenoceptor stimulation (Ayajiki and

Toda, 1990).

Page 146: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

122

The relaxant effects of adrenoceptor stimulation were also investigated in rat

cerebral arteries pre-contracted with vasopressin. Nonspecific activation of the

β-adrenoceptors with isoprenaline caused strong relaxation in cerebral arteries

pre-contracted with vasopressin. Similarly, noradrenaline induced relaxant

effects that varied between arteries taken from different regions greater

relaxation was obtained in caudal (posterior communicating and basilar) than

rostral (anterior and middle cerebral) arteries. No relaxation was induced by α-

adrenoceptor stimulation in cerebral arteries. This indicates that β-

adrenoceptors mediate the relaxation responses in rat cerebral arteries. The

relaxation effects in anterior and middle cerebral arteries counteract the

contraction induced by noradrenaline. As a result, noradrenaline induced weak

contractile responses that were attenuated by simultaneous stimulation of β-

adrenoceptors, unlike methoxamine which caused a relatively strong

contraction of anterior and middle cerebral arteries. The assertion was

supported by increases in the contractile responses to noradrenaline when β-

adrenoceptors were inhibited with propranolol; or selective β1-(atenolol) and

β2-adrenoceptor (ICI 118551) antagonists. These findings indicate that both β1-

and β2-adrenoceptors are involved in the relaxation induced by noradrenaline.

Similar findings were previously reported in rat middle cerebral arteries where

pre-treatment with propranolol enhanced the maximum contraction induced by

noradrenaline (Hogestatt and Andersson, 1984). Additionally, the presence of

β-adrenoceptors was confirmed by inhibition of the noradrenaline-induced

relaxation caused by adrenoceptor antagonists in rat cerebral arteries pre-

contracted with vasopressin. Propranolol 1 µM significantly attenuated the

relaxation, while atenolol 1 µM and ICI 118551 1 µM shifted the concentration-

relaxation curve of noradrenaline to the right. This shows involvement of both

β1- and β2-adrenoceptors in the relaxation induced by noradrenaline.

Relaxation induced by adrenaline, a drug with preferential affinity for β2-

adrenoceptors (pKi 6.2), reveals the presence of these receptors in the rat

basilar artery. However, the lack of response to formoterol, a selective β2-

adrenoceptor agonist (Baker, 2010), is intriguing and these observations need

further investigation.

Page 147: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

123

In the middle cerebral artery, removal of the endothelium neither inhibited the

relaxation induced by isoprenaline nor enhanced the noradrenaline-induced

contraction. The findings suggest an absence of relaxant β-adrenoceptors on

endothelial cells, but these may be located on the smooth muscle cells of the

rat middle cerebral artery. In contrast, removal of the endothelium inhibited

the relaxation induced by noradrenaline or isoprenaline in the basilar artery.

This indicates that β-adrenoceptors are located on the endothelium of the rat

basilar artery. Previous studies have shown the presence of both endothelial

and smooth muscle β-adrenoceptors in various peripheral arteries isolated from

different species (see Guimarães and Moura (2001) for a detailed review). It

appears that the presence of endothelial adrenoceptors in cerebral arteries is

now widely accepted (Winquist and Bohr, 1982; Hempelmann and Ziegler,

1993). However, most studies were conducted in basilar artery preparations

and did not compare the endothelial/smooth muscle distribution of β-

adrenoceptors in other cerebral arteries. Thus, this study has shown

heterogeneous regional distribution of endothelial and smooth muscle β-

adrenoceptors in rat cerebral arteries.

Inhibition of NO production with L-NAME inhibited the relaxation responses to

noradrenaline (up to a maximum concentration of 30 µM) in cerebral arteries.

Furthermore, L-NAME increased the contraction induced by noradrenaline in

anterior and middle cerebral arteries in contrast to the finding that the

contractile effect of noradrenaline was not enhanced by removing the

endothelium in the middle cerebral artery. However, L-NAME alone or in

combination with indomethacin did not inhibit the relaxation induced by

isoprenaline in rat middle cerebral and basilar arteries. This disparity observed

in the inhibitory effects of L-NAME on the responses induced by noradrenaline

and isoprenaline may possibly be related to a potentiation of vasopressin-

induced pre-contractile tone that has resisted the relaxant effects of

noradrenaline, a relatively weaker relaxant than isoprenaline. The increases in

the contractile responses to noradrenaline in anterior and middle cerebral

arteries pre-treated with L-NAME also illustrates a potentiation effect. L-NAME

can potentiate the effects of vasoconstrictor agonist by inhibiting the basal

Page 148: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

124

production of nitric oxide which is responsible for regulating tone of cerebral

arteries (Faraci, 1994).

The difference in efficacy between noradrenaline and isoprenaline with regard

to the quantity of cAMP generated has been previously noted. For example,

isoprenaline produced three times more cAMP than noradrenaline at 10-fold

less concentration in human lymphocytes (MacGregor et al., 1996). Whether

these variations are related to the differences in relative affinities for the β-

adrenoceptor subtypes or efficacy of the individual agonist is unknown. The

variations are unlikely to be due to differences in selectivity of the agonists for

β-adrenoceptor subtypes since both drugs have slightly greater affinities for β1

(pKi: noradrenaline 6.0 and isoprenaline 6.6-7.0) than β2-adrenoceptors (pKi:

noradrenaline 5.4 and isoprenaline 6.4) (IUPHAR/BPS

http://www.guidetopharmacology.org/). The difference in efficacies of

noradrenaline and isoprenaline in inducing relaxation of cerebral arteries

following activation of either β1 or β2-adrenoceptor subtypes is unknown and

needs to be investigated.

The possible role of EDH in adrenoceptor-mediated relaxation of rat cerebral

arteries was also analysed using a combination of apamin 50 nM and

charybdotoxin 50 nM, a combination that has been shown to block EDH by

selectively inhibiting potassium channels in the endothelium without affecting

the channels in smooth muscle cells (Doughty et al., 1999). The chemical

identity of EDH is not clearly identified, but it is generally accepted as an

agonist-induced endothelium-dependent relaxation of smooth muscle cells due

to an increased K+ conductance. It may involve efflux of ions or secondary

mediators from the endothelium into the myoendothelial space and/or transfer

into smooth muscle cells via the gap junctions (Ozkor and Quyyumi, 2011). This

study found that apamin 50 nM and charybdotoxin 50 nM only slightly inhibited

the relaxation induced by noradrenaline, but significantly inhibited the

isoprenaline-induced relaxation in the basilar artery. This indicates the

presence of EDH in the rat basilar artery that is favourably released by

isoprenaline. Further, a combination of apamin 50 nM and charybdotoxin 50 nM,

Page 149: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

125

L-NAME 100 µM and indomethacin 3 µM, inhibited the relaxation induced by

isoprenaline in the basilar artery, but not in the middle cerebral artery. This

shows an involvement of EDH and NO in the relaxation of the basilar artery

induced by β-adrenoceptor stimulation. In addition, these results reiterate the

fact that relaxation of the rat middle cerebral artery induced by β-

adrenoceptor activation is primarily an endothelium-independent phenomenon.

In another attempt to attenuate the production of EDH, arteries were pre-

contracted by using a depolarizing concentration of K+ (40 mM), as previously

indicated (Ozkor and Quyyumi, 2011). However, only weak relaxations were

induced by isoprenaline in both middle cerebral and basilar arteries pre-

contracted with 40 mM K+ demonstrating that high K+ may simultaneously inhibit

both NO and EDH-mediated relaxations.

Stimulation of vascular β-adrenoceptors has been known to induce

hyperpolarisation associated with smooth muscle relaxation (Goto et al., 2001).

Stimulation of β-adrenoceptors on endothelial cells causes increases in

intracellular calcium ([Ca2+]i ) that subsequently lead to synthesis of NO or

generation of EDH. It appears that NO-mediated relaxations require lower

concentrations of endothelial [Ca2+]i than EDH-mediated relaxation. This was

demonstrated in uridine triphosphate-induced relaxation of rat cerebral

arteries where a lower threshold concentration of endothelial [Ca2+]i (~220 nM)

was required for production of NO than for generation of EDH (~340 nM)

(Marrelli, 2001). Thus, stimulation of β-adrenoceptors with isoprenaline may

cause high [Ca2+]i that induces generation of EDH in the rat basilar artery.

Taken together, this study found that adrenoceptor activation induced variable

responses in the rat cerebral circulation due to heterogeneous distribution of

the constrictor and dilator adrenoceptors as summarised in Figure 4-19.

Constrictor α1-adrenoceptors are found in arteries taken from medial and

anterior locations, but β-adrenoceptors mediating relaxation are distributed in

both anterior and posterior regions of rat cerebral arterial circulation. The β-

adrenoceptors are located on smooth muscle in middle cerebral arteries, unlike

in the basilar artery where they are located only on the endothelium. Activation

Page 150: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

126

of endothelial β-adrenoceptors in the basilar artery induces release of variable

proportions of EDH and NO depending on the agonist used. Isoprenaline favours

release of EDH, while noradrenaline induces preferential release of NO in the

rat basilar artery.

Figure 4-19: Summary of the location and effects mediated by β-

adrenoceptors and effects of noradrenaline and

isoprenaline in rat cerebral arteries.

A) β-adrenoceptors mediating relaxation are located on smooth muscle and

endothelium of the rat middle cerebral and basilar artery, respectively, while α1-

adrenoceptors are located only on the smooth muscle of the middle cerebral

artery and mediate a relaxation response. B) Noradrenaline activated β1/β2-

adrenoceptors predominantly inducing release of nitric oxide (NO), but

isoprenaline favours the release of endothelium-derived hyperpolarisation (EDH)

in the rat basilar artery.

Page 151: Cerebrovascular effects of vasoactive drugs

Adrenoceptors

127

Limitations and future directions

Due to time limitations on the part of the candidate to submit this thesis,

experiments pertaining to further characterisation of β-adrenoceptors in rat

cerebral arteries have not been completed. Thus, the following points have

been identified for future investigation based on the findings of this study:

To examine the relationship between specific activation of β1/β2-

adrenoceptor subtypes and the release of NO and EDH and the

relevance of relaxations induced by noradrenaline and isoprenaline in

the rat basilar artery.

To assess the link between the efficacy of β-adrenoceptor agonists

(ligands) and the release of NO and EDH in the basilar artery and

beyond.

Page 152: Cerebrovascular effects of vasoactive drugs
Page 153: Cerebrovascular effects of vasoactive drugs

129

Effects of endothelin in the rat

cerebral circulation and the

role of voltage-operated and

non-voltage-operated calcium

channels

Page 154: Cerebrovascular effects of vasoactive drugs

Endothelin

130

Introduction

Endothelins are vasoactive peptides mainly produced and released by

endothelial cells. They were initially isolated from endothelial cells and

subsequently characterised by researchers in Japan (Yanagisawa et al., 1988;

Barton and Yanagisawa, 2008). The family includes endothelin-1, endothelin-2

and endothelin-3; endothelin-1 (a 21-amino acid peptide) being the most

powerful vasoconstrictor agent. Endothelins are synthesised as 212-amino acid

precursor peptides, called preproendothelins that are subsequently converted

to the respective big-endothelin intermediates (or proendothelins) by furin-like

endopeptidases. The big-endothelins are then converted to endothelins

primarily by the membrane-bound endothelin converting enzymes as shown in

Figure 5-1 (Barton and Yanagisawa, 2008). Apart from their vascular effects,

endothelins also have potent effects in the heart and kidney, as well as in the

central nervous, respiratory, endocrine, urogenital, and gastrointestinal

systems (Neylon, 1999).

Endothelins have two distinct G-protein-coupled receptors - ETA and ETB

(Davenport, 2002). ETA receptors have high affinities for endothelin-1 and

endothelin-2 but about 100-fold lower affinity for endothelin-3. The affinities

of ETB receptors are similar for all three endothelins (Kedzierski and

Yanagisawa, 2001). ETA receptors located on vascular smooth muscle cells

mediate the vasoconstrictor and growth promoting effects of endothelins, while

endothelial ETB receptors mediate vasodilatation, growth inhibition and

clearance of ETA receptors (Kawanabe and Nauli, 2005). However, endothelin

ETB receptors may also be located on vascular smooth muscle cells and mediate

vasoconstriction (Haynes et al., 1995).

The production of endothelin is mostly regulated at the level of mRNA

transcription since most cells producing endothelins lack storage vesicles.

Endothelin-1 mRNA transcription is upregulated by tumour necrosis-alpha,

growth factor-beta interleukins, noradrenaline, angiotensin II, thrombin,

insulin and hypocapnia. The expression of endothelin-1 is downregulated by

Page 155: Cerebrovascular effects of vasoactive drugs

Endothelin

131

nitric oxide, prostacyclin and atrial natriuretic peptide and hypoxia, while

initially increased then decreased by shear stress or stretch (Kedzierski and

Yanagisawa, 2001).

Preclinical and clinical studies have shown endothelin-1 is involved in the

pathogenesis of various diseases including pulmonary and arterial hypertension,

heart failure, renal diseases, atherosclerosis, diabetes and cancer (Kedzierski

and Yanagisawa, 2001). Endothelin-1 has also been identified as a potential

cause of ischaemic stroke and cerebral vasospasm that often occurs after

subarachnoid haemorrhage. Studies have reported increased concentrations of

endothelin-1 in cerebrospinal fluid of subarachnoid haemorrhage patients

during cerebral vasospasm (Kobayashi et al., 1995; Kästner et al., 2005). In

addition, increased sensitivity of cerebral vessels to endogenous

vasoconstrictor agents may also contribute to the pathogenesis of the

vasospasm due to upregulation of receptors (Edvinsson and Povlsen, 2011). This

indicates that even relatively low concentrations of vasoconstrictor mediators

may cause significant vasospasm.

Page 156: Cerebrovascular effects of vasoactive drugs

Endothelin

132

Figure 5-1: The endothelin pathway.

Endothelins are initially expressed as preproendothelin precursor peptides

(preproET-1, preproET-2 and preproET-3) that are subsequently cleaved by furin-

like proteases to form big-endothelins (proendothelins). Big-endothelins are then

converted to the respective endothelins by endothelin converting enzymes (ECE).

The endothelin peptides activate the G-protein-coupled endothelin ETA and ETB

receptors eliciting different physiological responses (From Kedzierski and

Yanagisawa (2001)).

The vasoconstrictor action of endothelin-1 depends predominantly on influx of

calcium from the extracellular space. It can also cause release of calcium from

the sarcoplasmic/endoplasmic reticulum that normally constitutes a small

proportion of the calcium source (QingHua et al., 2009). Despite initial reports

that endothelin-1 activates voltage-operated calcium channels (VOCC) in

porcine coronary artery smooth muscle cells (Goto et al., 1989), it appears that

these channels constitute only a small part in the vasoconstrictor response since

nifedipine, a specific inhibitor of L-type VOCC, did not inhibit calcium influx in

this tissue (Kawanabe et al., 2002; Kawanabe and Nauli, 2005). Endothelin-1

Page 157: Cerebrovascular effects of vasoactive drugs

Endothelin

133

has also been reported to enhance calcium entry into cardiac smooth muscle

cells via T-type VOCC in rat ventricular myocytes (Furukawa et al., 1992). Non-

voltage-operated calcium channels, namely receptor-operated calcium

channels and store-operated calcium channels, are known to play a principal

role in the sustained increase of intracellular calcium that maintains the

contractile effect of endothelin-1 (Enoki et al., 1995; Iwamuro et al., 1998;

Iwamuro et al., 1999; Zhang et al., 1999; Zhang et al., 2000; Kawanabe et al.,

2002). On the other hand, the dynamic range of global cytosolic calcium

mediating contraction in cerebral arteries is quite narrow (Wellman, 2006) and,

as a result, cerebral arteries are typically sensitive to constriction induced by

an agonist or increased intraluminal pressure. Understanding the functional

relationship between endothelin-1 and calcium channels in the cerebral

circulation may help to unravel the mechanism of endothelin-1-induced

vasospasm. The aim of this study was to characterise vasoconstrictor effects of

endothelin-1 and the roles of L- and T-type VOCC and non-VOCC in arterial

segments isolated from different regions of the rat cerebral circulation using

wire myography.

Page 158: Cerebrovascular effects of vasoactive drugs

Endothelin

134

Materials and methods

5.2.1. Methods

Arteries isolated from different regions of the rat cerebral circulation (anterior

cerebral, middle cerebral, posterior communicating and basilar arteries) were

characterised in vitro using wire myography as described in section 2.3. Rat

small mesenteric arteries were also isolated and characterised in a similar

manner.

5.2.2. Experimental protocols

The effects of endothelin-1 (0.01–100 nM) were examined by addition of

cumulative concentrations in half-log10 increments. Responses were allowed to

plateau before addition of the next higher concentration. The functional

presence of endothelin ETB receptors in cerebral arteries was assessed using

sarafotoxin S6c, a selective endothelin ETB receptor agonist. The vessels were

pre-contracted to 80–90% of the maximum KPSS-induced contraction using

vasopressin (0.1–1 nM), and relaxed with cumulative concentrations of

sarafotoxin S6c (0.01–3 nM). Nifedipine (1 M), NNC 55-0396 (1 M) or SK&F

96365 (10 or 30 M) were used to assess the roles of L-type VOCC, T-type VOCC

or SOCC and ROCC, respectively. Combinations of 1 M nifedipine with 10 or 30

M SK&F 96365 were also used to investigate effects of combined blockade of

L-type VOCC and non-voltage-operated calcium channels. All calcium channel

inhibitors were added 20–25 min before addition of endothelin-1.

In a second series, the ability of these calcium channel antagonists to relax

endothelin-1-induced contraction was investigated. The vessels were

contracted with endothelin-1 (10 nM) followed by addition of cumulative

concentrations of nifedipine, NNC 55-0396 or SK&F 96365. In a third series, the

middle cerebral artery was pre-contracted with endothelin-1 (10 nM) before

adding a single concentration of nifedipine (0.01 or 1 M), SK&F 96365 (1 or 10

M) or their combinations. Some vessels acted as time controls after the

endothelin-1 pre-contraction.

Page 159: Cerebrovascular effects of vasoactive drugs

Endothelin

135

5.2.3. Drugs and chemicals

The drugs used in this study were obtained from the following suppliers:

Endothelin-1 (GenScript, Piscataway, NJ, USA), vasopressin and sarafotoxin S6c

(AusPep, Parkville, Victoria, Australia), bosentan (gift from Actelion

Pharmaceutical Ltd., Allschwil, Switzerland), nifedipine (Sigma Aldrich, St

Louis, Missouri, USA), NNC 55-0396 and SK&F 96365 (Tocris Bioscience, Bristol,

UK). Endothelin-1 was dissolved in 10% dimethylformamide (DMF). Nifedipine

was dissolved in dimethylsulfoxide (DMSO) and diluted in water. Vasopressin,

bosentan, sarafotoxin S6c, NNC 55-0396 and SK&F 96365 were dissolved in

water.

Page 160: Cerebrovascular effects of vasoactive drugs

Endothelin

136

Results

5.3.1. Effects of endothelin receptor stimulation in rat

cerebral arteries

The effects of endothelin receptor activation were investigated in rat anterior

cerebral, middle cerebral, posterior communicating and basilar arteries.

Endothelin-1 induced potent constrictor effects in cerebral and mesenteric

arteries. The contraction induced by endothelin-1 was characterised by

vasomotion (a wave-like oscillation of tone), typically in the middle cerebral

artery, which was abolished by the calcium channel inhibitors (Figure 5-2). The

middle cerebral and posterior communicating arteries tended to be more

sensitive to endothelin-1 than anterior cerebral artery or basilar artery (Figure

5-3 and Table 5-1). The maximum effects caused by endothelin-1 in the cerebral

arteries ranged from 108±2% (n=11) (in basilar artery) to 122±2% (n=10) (in

anterior cerebral artery) of the KPSS-induced maximum contraction; similar

effects were obtained in mesenteric arteries as shown in Table 5-1. Sarafotoxin

S6c (0.01-1 nM) caused significant relaxation of vasopressin pre-contracted

arteries with similar potency in each vessel (pEC50 10.2±0.2-10.4±0.2). The

maximum relaxation was, however, variable between arteries taken from

different regions of the rat cerebral circulation - anterior cerebral artery and

middle cerebral artery (77%), posterior communicating artery (88%) and basilar

artery (100%) of vasopressin pre-contraction (Figure 5-3). Sarafotoxin S6c did

not induce any contraction in cerebral arteries, with or without endothelium.

Page 161: Cerebrovascular effects of vasoactive drugs

Endothelin

137

Figure 5-2: Representative trace of the endothelin-induced contraction

of cerebral arteries.

Individual experimental traces of endothelin-1-induced vascular contraction in

middle cerebral arteries showing (left panel) the oscillations in tone and (right

panel) the effect of pre-treatment with the L-type calcium channel inhibitor

nifedipine 1 M.

Page 162: Cerebrovascular effects of vasoactive drugs

Endothelin

138

Figure 5-3: Contractile effects of endothelin-1 in rat cerebral arteries.

Contractile effects of endothelin-1 in rat anterior cerebral, middle cerebral,

posterior communicating and basilar arteries. Data are shown as percent of KPSS-

induced maximum contraction. Vertical bars represent ± SEM (those not shown are

contained within the symbol); horizontal bars indicate EC50±SEM. n, number of

arteries from separate rats.

Page 163: Cerebrovascular effects of vasoactive drugs

Endothelin

139

Figure 5-4: Relaxant effects of sarafotoxin S6C in rat cerebral arteries.

Relaxant effects of sarafotoxin S6C in rat anterior cerebral, middle cerebral,

posterior communicating and basilar arteries pre-contracted with vasopressin (AVP)

0.1-1 nM. Data are shown as percent of KPSS-induced maximum contraction. Vertical

bars represent ± SEM (those not shown are contained within the symbol); horizontal

bars indicate EC50±SEM. n, number of arteries from separate rats.

5.3.2. Antagonism of endothelin receptors in rat cerebral

arteries

Specific inhibition of the ETA receptors with bosentan caused a rightward shift

of the endothelin-1 concentration-response curve in rat cerebral and

mesenteric arteries (Figure 5-5 and Table 5-1). At 0.1 µM, bosentan caused a

significant inhibition of endothelin-1-induced contraction only in the anterior

Page 164: Cerebrovascular effects of vasoactive drugs

Endothelin

140

cerebral artery (1.5 fold shift) and basilar artery (2.5 fold shift). However

bosentan 1 µM, significantly antagonised endothelin-1 in all arteries with fold

rightward shifts in EC50 of 4, 5, 8, 4 and 5 in the anterior cerebral, middle

cerebral, posterior communicating, basilar and mesenteric arteries,

respectively (P<0.05, compared with respective vehicle group) (Table 5-1).

Page 165: Cerebrovascular effects of vasoactive drugs

Endothelin

141

Figure 5-5: Effects of bosentan on the endothelin-induced contraction of

cerebral arteries.

Effects of bosentan (Bos) 0.1 or 1 µM on the contraction induced by endothelin-1 in

rat anterior cerebral, middle cerebral, posterior communicating and basilar arteries.

Bosentan was added 20 min before endothelin contraction. Data are shown as

percent of KPSS-induced maximum contraction. Vertical bars represent ± SEM (those

not shown are contained within the symbol); horizontal bars indicate EC50±SEM. n,

number of arteries from separate rats.

Page 166: Cerebrovascular effects of vasoactive drugs

142

Table 5-1: Comparison of the pEC50 and Emax values of ET-1 and the fold shift caused by bosentan pre-treatment in

rat cerebral and mesenteric arteries

Pre-treatment ACA MCA Pcom BA Mes

Vehicle

n 10 10 9 11 5

Emax 122±2 119±2 117±4 108±2 109±3

pEC50 8.4±0.06 8.7±0.11 8.8±0.14 8.3±0.05 8.4±0.12

Bosentan 0.1 µM

n 7 6 7 7 6

Emax 122±3 112±4 123±2 109±3 116±2

pEC50 8.2±0.04* 8.3±0.14 8.5±0.16 7.9±0.08* 8.2±0.08

EC50 Fold shifta 1.5 - - 2.5 -

Bosentan 1 µM

n 6 6 5 6 5

Emax 117±4 113±5 123±4 110±4 117±1

pEC50 7.8±0.07* 8.0±0.07* 7.9±0.11* 7.7±0.04* 7.7±0.10*

EC50 fold shifta 4 5 8 4 5

ET-1, endothelin-1; ACA, anterior cerebral artery; MCA, middle cerebral artery; Pcom, posterior communicating artery; BA, basilar

artery; Mes, mesenteric artery. Emax, the maximum contractile response produced by ET-1 expressed as % of KPSS-induced maximum

contraction; EC50, the concentration of ET-1 that caused 50% of the maximum response; pEC50, the negative logarithm of EC50; n,

the number of arteries (each from a separate rat). *P<0.05, compared to respective vehicle, one-way ANOVA with Dunnett’s post-

test for multiple comparisons. aRightward shift caused by bosentan.

Page 167: Cerebrovascular effects of vasoactive drugs

Endothelin

143

5.3.3. Effects of inhibiting nitric oxide production and

endothelium removal on the endothelin-1-induced

contraction of cerebral arteries

Inhibition of endothelial nitric oxide production with L-NAME 100 µM caused a

significant leftward shift of the endothelin-1 concentration-contraction curve

in rat anterior cerebral artery (p<0.05 compared to vehicle, unpaired t test).

L-NAME did not, however, affect the endothelin-1 response in middle cerebral,

posterior communicating and basilar arteries (Figure 5-6). Physical removal of

the endothelium increased the potency of endothelin-1 in both anterior

cerebral and basilar arteries, but it did not significantly change the pEC50 of

endothelin-1 in the rat middle cerebral and posterior communicating arteries

(Figure 5-7). The maximum contraction elicited by endothelin-1 in the rat

cerebral arteries was not affected by L-NAME treatment or removal of the

endothelium.

Page 168: Cerebrovascular effects of vasoactive drugs

Endothelin

144

Figure 5-6: Effects of inhibiting nitric oxide production on the

endothelin-1-induced contraction of cerebral arteries.

Contractile effects of endothelin-1 in rat anterior cerebral, middle cerebral,

posterior communicating and basilar arteries pre-treated with N-nitro-L-arginine

methyl ester (L-NAME). L-NAME was added 20 min before endothelin-1 contraction.

Data are shown as percent of KPSS-induced maximum contraction. Vertical bars

represent ±SEM (those not shown are contained within the symbol); horizontal bars

indicate EC50±SEM. n, number of arteries from separate rats. *P<0.05 compared to

vehicle, unpaired t test.

Page 169: Cerebrovascular effects of vasoactive drugs

Endothelin

145

Figure 5-7: Effects of endothelium removal on endothelin-1-induced

contraction of cerebral arteries.

Contractile effects of endothelin-1 in endothelium-denuded rat anterior cerebral,

middle cerebral, posterior communicating and basilar arteries. The endothelium was

removed from the arteries by rubbing the lumen with a human hair. Data are shown

as percent of KPSS-induced maximum contraction. Vertical bars represent ±SEM

(those not shown are contained within the symbol); horizontal bars indicate

EC50±SEM. n, number of arteries from separate rats. *P<0.05 compared to vehicle,

unpaired t test.

5.3.4. Contractile effects of endothelin in calcium-free

physiological salt solution

Page 170: Cerebrovascular effects of vasoactive drugs

Endothelin

146

Calcium-free physiological salt solution was used to analyse the endothelin-1-

induced release of calcium from intracellular stores. Only a small contraction

was obtained with addition of endothelin-1 in the absence of extracellular

calcium (Figure 5-8). The estimated pEC50 value of endothelin-1 in calcium-free

solution, however, was not significantly different from the respective vehicle

(normal PSS) in all arteries (P>0.05) (Table 5-2). The maximum contraction

induced by endothelin-1 ranged from about 13% (anterior cerebral artery) to

24% (basilar artery) of the maximum KPSS-induced contraction (Table 5-3).

Page 171: Cerebrovascular effects of vasoactive drugs

Endothelin

147

Figure 5-8: The contractile effects of endothelin-1 in calcium-free

medium.

The contractile effects of endothelin-1 in rat cerebral arteries: A. in physiological

salt solution (PSS) containing 1.5 mM calcium; and B. in PSS without calcium. Data

are shown as percentage of KPSS-induced maximum contraction. Vertical bars

represent ± SEM (those not shown are contained within the symbol); horizontal bars

indicate EC50±SEM. n, number of arteries from separate rats. ET-1, endothelin-1;

ACA, anterior cerebral artery; MCA, middle cerebral artery; Pcom, posterior

communicating artery and BA, basilar artery.

5.3.5. Effects of nifedipine pre-treatment on endothelin-1-

induced contraction

Inhibition of the L-type VOCC with 1 µM nifedipine did not affect the pEC50

values of endothelin-1 in the cerebral arteries (P>0.05) (Table 5-2). Similarly,

the maximum contraction induced by endothelin-1 was not significantly

decreased except in the middle cerebral artery (P<0.05) where a maximum

effect was decreased by about 19% compared with the (Table 5-3 and Figure

5-9A). Nifedipine did not affect the baseline tone of the arteries during the pre-

treatment period.

Page 172: Cerebrovascular effects of vasoactive drugs

Endothelin

148

5.3.6. Effects of NNC 55-0396 pre-treatment on endothelin-1-

induced contraction

Pre-treatment of the different cerebral arteries with NNC 55-0396 1 µM, a

putative selective T-type voltage-operated calcium channel blocker did not

significantly change the pEC50 of endothelin-1 (Table 5-2). The maximum

contractile effect of endothelin-1 was, however, significantly decreased in the

cerebral arteries (Table 5-3; shown for middle cerebral artery in Figure 5-9B).

NNC 55-0396 1 µM did not affect the baseline arterial tone, but moderate to

strong contractions were observed in most vessels after addition of 3 or 10 µM.

Thus, a higher concentration of NNC 55-0396 was not considered in this study.

5.3.7. Effects of SK&F 96365 pre-treatment on endothelin-1-

induced contraction

Pre-treatment of the cerebral arteries with SK&F 96365 10 µM was without

effect, while 30 µM caused a concentration-dependent decrease in the

maximum endothelin-1 contractile response without significantly altering the

pEC50 values (Table 5-2 and Table 5-3). As shown in Figure 5-9C for the middle

cerebral artery, the maximum contractile effect of endothelin-1 was not

significantly affected by 10 µM of SK&F 96365. At 30 µM, however, SK&F 96365

significantly decreased the maximum contractile effects of endothelin-1

(P<0.05) in the cerebral arteries, with changes in Emax of 23% in basilar arteries

to 41% in middle cerebral arteries compared with respective vehicle pre-

treatment values (Table 5-3 and Figure 5-9C). SK&F 96365 100 µM completely

abolished the contraction induced by endothelin-1 in cerebral arteries. SK&F

96365 10 or 30 µM did not affect the baseline arterial tone, while a slight

decrease was observed in some arteries after addition of 100 µM.

Page 173: Cerebrovascular effects of vasoactive drugs

Endothelin

149

5.3.8. Combined effects of nifedipine and SK&F 96365 on

endothelin-1-induced contraction

Rat cerebral arteries were pre-treated with a combination of nifedipine 1 µM

with SK&F 96365 10 µM or 30 µM to assess the effects of simultaneous blockade

of L-type VOCC and non-VOCC on the contractile effects of endothelin-1.

Combinations of nifedipine and SK&F 96365 did not significantly change the

pEC50 of endothelin-1 (Table 5-2); however, maximum contractile effects of

endothelin-1 were significantly attenuated (P<0.05; Table 5-3). Thus, the

response to endothelin-1 10 nM (alone) was 110±4% of the KPSS-induced

contraction and the combination of nifedipine 1 µM with SK&F 96365 10 µM

decreased the response to 44±7% of KPSS-induced contraction (Figure 5-9D).

Similarly, combination of nifedipine 1 µM with SK&F 96365 30 µM caused a

further decrease in the maximum effect of endothelin-1 (36% in middle

cerebral artery, Figure 5-9D; and 52% in basilar artery, Table 5-3; P<0.05). In

all arteries, the resting baseline tone was not affected by either combination

of antagonists.

Further examination of the responses to endothelin-1 at 3 nM (taken from the

concentration–response curves when neither nifedipine 1 µM nor SK&F 96365

10 µM affected the endothelin-1 contraction), showed that the combined

treatment attenuated the response to just 17±5% of KPSS-induced contraction

in the middle cerebral artery (Figure 5-10). The combination of nifedipine 1

µM and NNC 55-0396 1 µM did not show this enhanced effect.

Page 174: Cerebrovascular effects of vasoactive drugs

Endothelin

150

Figure 5-9: Effect of calcium channel inhibition on endothelin-induced

contraction of middle cerebral arteries.

Effects of endothelin-1 in rat isolated middle cerebral arteries pre-treated with:

A. nifedipine; B. NNC 55-0396; C. SK&F 96365; or D. Combination of nifedipine and

SK&F 96365. ET-1, endothelin-1; SK&F, SK&F 96365; NNC, NNC 55-0396; Nifed,

nifedipine. Data are shown as percentage of KPSS-induced maximum contraction.

Vertical bars represent ± SEM (those not shown are contained within the symbol);

horizontal bars indicate EC50±SEM. n, number of arteries from separate rats. The

calcium channel antagonists were added 20 min prior to endothelin-1 contraction.

Page 175: Cerebrovascular effects of vasoactive drugs

Endothelin

151

Figure 5-10: Comparison of the effects of calcium channel antagonists on

endothelin-1-induced contraction of cerebral arteries.

The effects of pre-incubation with calcium channel antagonists, alone or in

combination, on the contraction induced by endothelin-1 at 3 nM in rat middle

cerebral arteries (data are derived from the ET-1 concentration-response curves

depicted in Figure 5-9). Data are shown as percentage of KPSS-induced maximum

contraction. Vertical bars represent ± SEM. The calcium channel antagonists were

added 20 min prior to endothelin-1 contraction. Presence or absence of a drug is

indicated by (+) or (-), respectively. ET-1, endothelin-1; SK&F, SK&F 96365; NNC,

NNC 55-0396; Nifed, nifedipine. *P<0.05 compared to vehicle (endothelin-1 alone),

one way ANOVA followed by Dunnett’s post-test.

5.3.9. Relaxation of endothelin-1-induced contraction with

calcium channel blockers

In a separate series of experiments, arteries were pre-contracted with

endothelin-1 to assess the relaxant effects of various calcium channel

Page 176: Cerebrovascular effects of vasoactive drugs

Endothelin

152

antagonists following pre-contraction induced by endothelin-1 (Figure 5-11).

Over the same time course that the channel antagonists were tested, the

plateau of endothelin-1 (10 nM)-induced contraction did not change in the time

control group. SK&F 96365 caused concentration-dependent relaxation of rat

cerebral arteries pre-contracted with endothelin-1 (10 nM). The relaxation

potency of SK&F 96365 was similar in all cerebral arteries with no significant

difference in the pEC50 values observed (e.g., 4.9±0.11, n=3 in anterior cerebral

artery). The endothelin-1mediated contraction was almost completely relaxed

in all arteries by 100 µM SK&F 96365. Nifedipine relaxed the endothelin-1-

induced contraction with similar potency in all vessels (pEC50 in anterior

cerebral artery 7.7±0.14, n=4). Anterior cerebral artery, middle cerebral artery

and posterior communicating artery were relaxed by about 65%, 75% and 77%,

respectively, with nifedipine 1 µM while in basilar artery the contraction was

relaxed only by 52%. In contrast, endothelin-1 pre-contraction was not

effectively reversed by NNC 55-0396 (Figure 5-11). The arteries started to re-

contract when the concentration of NNC 55-0396 was further increased.

Page 177: Cerebrovascular effects of vasoactive drugs

153

Table 5-2: Endothelin-1 pEC50 values in rat isolated cerebral arteries in the presence of various calcium channel

antagonists

Treatment

ACA MCA Pcom BA

n pEC50 n pEC50 n pEC50 n pEC50

Vehicle 10 8.4±0.06 10 8.7±0.11 9 8.8±0.14 11 8.3±0.05

Nifedipine 1 µM 4 8.7±0.22 5 8.8±0.22 4 8.7±0.19 4 8.7±0.19

NNC 55-0396 1 µM 5 8.6±0.08 5 8.4±0.06 5 8.6±0.15 5 8.4±0.12

SK&F 96365 10 µM 5 8.7±0.13 4 8.6±0.06 5 8.8±0.09 4 8.6±0.16

SK&F 96365 30 µM 4 8.5±0.08 4 8.3±0.09 4 8.6±0.07 4 8.5±0.07

SK&F 96365 10 µM + Nifedipine 1 µM 4 8.3±0.09 5 8.2±0.05 3 8.4±0.06 4 8.3±0.04

SK&F 96365 30 µM + Nifedipine 1 µM 4 8.3±0.06 4 8.3±0.11 4 8.5±0.10 4 8.3±0.15

Calcium-free PSS 3 8.6±0.04a 3 8.6±0.04a 3 8.7±0.09a 3 8.5±0.14a

ACA, anterior cerebral artery; MCA, middle cerebral artery; Pcom, posterior communicating artery; BA, basilar artery, pEC50, the negative

logarithm of EC50 (concentration of endothelin-1 that caused 50% maximum contraction); PSS, physiological salt solution; n, number of

arteries (each from separate animals). The calcium channel antagonists were added 20 min before endothelin-1. apEC50 values are only

estimates.

Page 178: Cerebrovascular effects of vasoactive drugs

154

Table 5-3: The effects of various calcium channel antagonist pre-treatments on the Emax of endothelin-1 in rat

isolated cerebral arteries

ACA MCA Pcom BA

Treatment Emax %Emax Emax %Emax Emax %Emax Emax %Emax

Vehicle 122±2 100 119±2 100 117±4 100 108±2 100

Nifedipine 1 µM 101±4 83 96±7* 81 102±10 87 90±6 83

NNC 55-0396 1 µM 81±4* 66 87±4* 73 96±3* 82 89±9* 82

SK&F 96365 10 µM 127±9 104 120±9 100 111±5 95 113±7 105

SK&F 96365 30 µM 84±9* 69 70±3* 59 74±11* 63 83±2* 77

SK&F 96365 10 µM + Nifedipine 1 µM 66±12*† 54 62±3*† 52 85±6* 73 85±9* 79

SK&F 96365 30 µM + Nifedipine 1 µM 39±9*† 32 36±6*† 30 45±4*† 38 52±5*† 48

Calcium-free PSS 16±2* 13 13±3* 11 14±2* 12 24±1* 22

ACA, anterior cerebral artery; MCA, middle cerebral artery; Pcom, posterior communicating artery; BA, basilar artery; Emax, the

maximum contractile effect of endothelin-1 expressed as % KPSS-induced maximum contraction; % Emax, expressed as % respective

vehicle Emax. The calcium channel antagonists were added 20 min before endothelin-1 contraction. *P<0.05 compared to respective

vehicle, one way ANOVA followed by Dunnett’s post hoc test. †P<0.05 compared to nifedipine 1 µM treatment, one way ANOVA

followed by Bonferroni’s post hoc test between selected pairs.

Page 179: Cerebrovascular effects of vasoactive drugs

155

Figure 5-11: Relaxant effects of calcium channel antagonists in cerebral arteries pre-contracted with endothelin-

1.

Relaxation of the endothelin-1 induced contraction in rat isolated cerebral arteries using A. SK&F 96365; B. nifedipine; and C. NNC

55-0396. ET 1, endothelin-1; ACA, anterior cerebral artery; MCA, middle cerebral artery; Pcom, posterior communicating artery;

and BA, basilar artery. Data are shown as percentage of KPSS-induced contraction. Vertical bars represent ±SEM; horizontal bars

indicate EC50±SEM. The arteries were pre-contracted with 10 nM endothelin-1 and cumulative concentrations of each of the

respective antagonists were added after the endothelin response had plateaued. n, number of arteries from different rats.

Page 180: Cerebrovascular effects of vasoactive drugs

Endothelin

156

Figure 5-12: Relaxation of endothelin-1 pre-contracted cerebral arteries

with combinations of calcium channel antagonists

Relaxation of endothelin-1 (10 nM)-pre-contracted middle cerebral arteries with

calcium channel antagonists alone or in combination at low-high concentrations. A.

Low concentration of SK&F 96365 (1 µM) and nifedipine (0.01 µM) and their

combination; B. Low concentration of SK&F 96365 (1 µM) and high concentration of

nifedipine (1 µM) and their combination; C. High concentration of SK&F 96365 (10

µM) and low concentration of nifedipine (0.01 µM) and their combination; and D.

High concentration of SK&F 96365 (10 µM) and nifedipine (1 µM) and their

combination. Data are shown as percentage of the contraction induced by 10 nM

endothelin-1. Error bars are ±SEM. *P<0.05; one-way ANOVA followed by Dunnett’s

post-test. n≥5 for each treatment group.

Page 181: Cerebrovascular effects of vasoactive drugs

Endothelin

157

Discussion

In this in vitro study the effects of endothelin receptor activation and role of

calcium channels in arterial segments isolated from different regions of the rat

cerebral circulation were analysed. Endothelin-1 was shown to induce a potent

vasoconstrictor response with no significant regional variation in sensitivity

within the cerebral circulation. Endothelin-1 caused similar effects in

mesenteric arteries. Bosentan, a dual endothelin ETA and ETB antagonist,

shifted the contraction-response curve to the right. On the other hand,

activation of endothelin ETB receptors using sarafotoxin S6c did not induce

vasoconstriction in the cerebral arteries. The findings show that

vasoconstriction of cerebral arteries induced by endothelin-1 may be

dependent on activation of ETA receptors only. However, sarafotoxin S6c caused

relaxation of pre-contracted cerebral arteries supporting the presence of

endothelial ETB receptors (Nilsson et al., 2008). Inhibition of endothelial nitric

oxide production and removal of endothelium caused a variable increment in

sensitivity to endothelin-1. This indicates a heterogeneous distribution of ETA

and ETB receptors in cerebral arteries.

The interdependence between endothelin receptor activation and calcium

mobilization involves complex mechanisms and appears to vary in different cells

and tissues (Tykocki and Watts, 2010). The diversity of signals induced by

endothelin-1 can be attributed to disparity in the level of expression of various

channels involved in calcium mobilization (Neylon, 1999). The current study

demonstrates that endothelin-1 induces contraction of cerebral arteries both

by releasing calcium from intracellular stores and influx of extracellular

calcium. However, the vasoconstrictor actions of endothelin-1 largely depend

on an influx of calcium from extracellular sources as evidenced by the weak

response (<22%) elicited by endothelin-1 in cerebral arteries in calcium-free

solution. This shows that the calcium released from internal stores accounts for

a small proportion of the calcium needed for endothelin-1-induced contraction

of these arteries. On the other hand, the threshold concentration of endothelin-

1 that caused vascular contraction was higher in calcium-free solution than in

Page 182: Cerebrovascular effects of vasoactive drugs

Endothelin

158

calcium-containing buffer. This may indicate that low concentrations of

endothelin-1 activate influx of extracellular calcium, while higher

concentrations are needed to release calcium from internal stores. The finding

is consistent with an earlier report by Komuro et al. (1997) that removal of

extracellular calcium abolished effects of low concentrations of endothelin-1

in rabbit isolated aortic rings. However, it differs from a previous report that

endothelin-1-induced contraction of rabbit basilar artery was exclusively

dependent on influx of extracellular calcium (Kawanabe et al., 2002). The

variation between this latter finding and our results may be due to species

difference. In addition, endothelin may enhance the sensitivity of the vascular

smooth muscle cell contractile machinery to calcium via the protein kinase C

pathway, which might have contributed to the long-lasting contraction induced

by endothelin (Labadia et al., 1997; Cain et al., 2002). In line with this,

endothelin may also activate RhoA/Rho kinase and thus inactivate myosin light

chain phosphatase leading to the maintenance of contraction for long periods

(Scherer et al., 2002; Wynne et al., 2009).

Pre-treatment with nifedipine 1 µM, a concentration that is reported to

maximally inhibit the L-type VOCC (Iwamuro et al., 1998; Navarro-Gonzalez et

al., 2009), caused suppression of the contraction induced by higher

concentrations of endothelin-1 in cerebral arteries but did not affect the low

concentration responses. This suggests that L-type VOCC may not play a

significant role in the influx of extracellular calcium that is needed for

contraction induced by lower concentrations (<1 nM) of endothelin-1.

Interestingly, inhibition of the T-type VOCC with NNC 55-0396 1 µM decreased

the contraction response induced by low as well as high concentrations of

endothelin-1. The observation that the responses to low concentrations of

endothelin-1 (unaffected by nifedipine) were inhibited by NNC 55-0396

demonstrates that T-type VOCC may be activated by low concentrations of

endothelin-1 in the rat cerebral arteries. Kuo et al. (2010) also demonstrated a

mibefradil- and NNC 55-0396-sensitive response in smaller branches of rat

basilar artery and there is an increasing body of evidence that T-type VOCC are

expressed in cerebral vasculature (Abd El-Rahman et al., 2013; Harraz and

Page 183: Cerebrovascular effects of vasoactive drugs

Endothelin

159

Welsh, 2013). While the results from this study revealed a potential role of T-

type VOCC in endothelin-1-induced vasoconstriction of cerebral arteries,

rigorous experimental investigation is needed for further understanding of the

actual role of T channels in cerebral arteries.

The nifedipine (L) or NNC 55-0396 (T)-insensitive component of the contraction

induced by endothelin-1 was blocked by SK&F 96365, a putative non-VOCC

inhibitor, suggesting that the residual response may be mediated by non-VOCC

activation. It is worth noting that SK&F 96365 (1-10 µM) reportedly blocked

multiple VOCC including T-type channels in HEK 293 cells (Singh et al., 2010).

But there are substantial reservations about the findings of VOCC blockade in

HEK 293 cells compared with native tissue. Therefore, this drug was not used

in this study for the purpose of blocking a specific calcium channel – rather it

was used to block the voltage-insensitive component of calcium influx or the

non-VOCC-mediated calcium entry. SK&F 96365 caused concentration-

dependent inhibition of the endothelin-1-induced contraction of the cerebral

arteries demonstrating roles of the non-VOCC in addition to the L- and T-type

VOCC. A previous study by Saleh et al. (2009) reported that endothelin-1 causes

activation of canonical transient receptor potential type-1 (TRPC1) channels in

rabbit coronary artery myocytes. TRPC1 channels are often loosely referred to

as ROCC or SOCC in order to highlight that they may be gated due to receptor

activation or secondary to depletion of intracellular calcium storage. Complete

inhibition of endothelin-1-induced contraction, however, shows that high

concentrations of SK&F 96365 (100 µM) non-specifically block other calcium

channels including those located on intracellular stores.

Addition of nifedipine (0.01-1 µM) to cerebral vessels pre-contracted with a high

concentration (10 nM) of endothelin-1 resulted in marked relaxation. The

maximum relaxation caused by nifedipine 1 µM was about 74% in the middle

cerebral artery. In contrast, NNC 55-0396 (0.1-1 µM) did not cause significant

relaxation of the arteries pre-contracted with endothelin-1. This implies that

L-type VOCC, which can be activated secondary to depolarization caused by

opening of other calcium channels, play greater role in the maintenance phase

Page 184: Cerebrovascular effects of vasoactive drugs

Endothelin

160

of contraction caused by higher concentrations of endothelin-1 in rat cerebral

arteries, as previously suggested (Neylon, 1999; Navarro-Gonzalez et al., 2009).

SK&F 96365 (1-100 µM) also caused a concentration-dependant relaxation of

arteries pre-contracted with endothelin-1 with a complete reversal observed at

100 µM.

Activation of G-protein-coupled endothelin ETA receptors stimulates

phospholipase C which hydrolyses the membrane bound phosphatidyl 4, 5

bisphosphate (PIP2) into inositol triphosphate (IP3) and diacylglycerol (DAG). IP3

and DAG then cause release of calcium from intracellular stores

(sarcoplasmic/endoplasmic reticulum) and opening of non-VOCC directly

and/or indirectly through depletion of intracellular calcium stores (Motte et

al., 2006). The findings of the current study indicate that the initiation and

maintenance phases of endothelin-1-induced contraction may involve a

different profile of calcium channel activation. The contraction induced by low

concentrations of endothelin-1 (<1 nM) may be initiated by release of calcium

from intracellular stores as well as activation of non-VOCC, which in turn cause

membrane depolarization that gradually leads to opening of the low voltage-

activated calcium channels (T-type VOCC) which normally open at membrane

potential of -65 to -45 mV (Kuo et al., 2011). Higher concentrations of

endothelin-1 caused opening of L-type VOCC and the maintenance phase of

contraction caused by high concentrations of endothelin-1 largely depends on

influx of calcium through L-type VOCC and the non-VOCC play a lesser role

(Figure 5-13).

Combination treatment with nifedipine and SK&F 96365 revealed an interaction

between VOCC and non-VOCC in mediating the contraction of cerebral arteries

induced by endothelin-1. Pre-treatment with either nifedipine (1 µM) or SK&F

96365 (10 µM) alone did not significantly inhibit responses to low concentrations

of endothelin-1, but their combination significantly decreased the overall

responses, including those induced by low endothelin-1 concentrations, as

illustrated in Figure 5-10. The synergy observed between the antagonists in

Page 185: Cerebrovascular effects of vasoactive drugs

Endothelin

161

inhibiting the endothelin-1-induced contraction may illustrate interdependence

of VOCC and non-VOCC in mobilization of the extracellular calcium.

The relative contribution of non-VOCC and VOCC is altered during the

contraction phase. Thus, NNC 55-0396 sensitivity was decreased in pre-

contracted arteries. In contrast, arteries pre-contracted with endothelin-1

were more sensitive to nifedipine and SKF 96365. Thus, nifedipine, caused

about 74% relaxation of endothelin-1 pre-contraction, in contrast to the weak

inhibitory effect (<25%) on endothelin-1 contraction when used as pre-

treatment, and SK&F 96365 caused complete concentration-dependent

relaxation of the pre-contracted arteries. In addition, in endothelin-1 pre-

contracted arteries additive responses were obtained when the arteries were

relaxed using combinations of nifedipine and SK&F 96365. This is in contrast to

the pre-treatment findings and suggests that nifedipine and SK&F 96365 relax

the endothelin-1-pre-contracted arteries through independent inhibition of the

VOCC and non-VOCC. It can be implied that relatively low concentrations of

these calcium channel antagonists may be more effective in reversing rather

than preventing contraction, a finding that may point to the therapeutic

management of cerebral vasospasm.

In conclusion, the potent ETA receptor-mediated vasoconstrictor effect of

endothelin-1 involves mobilization of calcium through activation of non-VOCC

as well as L-and T-type VOCC. Combination of nifedipine and SK&F 96365

demonstrated synergistic inhibition of endothelin-1-induced vasoconstriction

when added prior to endothelin. Interestingly, nifedipine and SK&F 96365 were

more effective in relaxing the contraction induced by endothelin-1 than

preventing it through pre-treatment. This work suggests that non-VOCC may

work in concert with VOCC in mediating induction of vasoconstrictor effects of

endothelin-1 in cerebral arteries and that the L-type VOCC antagonists are not

sufficient to fully relax endothelin-1 pre-contracted cerebral arteries.

Page 186: Cerebrovascular effects of vasoactive drugs

Endothelin

162

Figure 5-13: A schematic representation of the contribution of different

calcium channels in the contractile response to endothelin-

1.

The results calculated from the area under the concentration-response curve

(expressed as %), contrasting the effect of adding antagonists before or during

endothelin-1 contraction. The contributions were allocated according to i) the effect

of the L-type VOCC antagonist nifedipine (1 µM), the T-type VOCC antagonist NNC

55-0396 (1 µM) on the endothelin-1 concentration-response curve and the residual

response unaffected by either agent; and ii) the contribution from L-type VOCC and

non-VOCC antagonists to the relaxation of endothelin-1 (10 µM) pre-contracted

middle cerebral arteries. VOCC; voltage-operated calcium channels.

Page 187: Cerebrovascular effects of vasoactive drugs

163

Cerebrovascular effects of

vasoactive substances in vivo

Page 188: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

164

Introduction

Cerebral autoregulation (or pressure autoregulation) is defined as the ability of

the cerebral circulation to maintain adequate cerebral blood flow in spite of

changes in perfusion pressure due to variation in peripheral blood pressure

within the range of 60 to 150 mmHg (Paulson et al., 1990; van Beek et al.,

2008). The main goal of autoregulation is to maintain cerebral blood flow in

face of varying perfusion pressure by changing the cerebrovascular resistance.

The relationship between cerebral blood flow, perfusion pressure and vascular

resistance can be expressed using Ohm’s law which states that flow [current] is

directly proportional to pressure [voltage] but inversely proportional to the

vascular resistance (Cipolla, 2009; Partington and Farmery, 2014).

𝑪𝑩𝑭 =𝑪𝑷𝑷

𝑪𝑽𝑹

Where, CBF, cerebral blood flow; CPP, cerebral perfusion pressure; and CVR,

cerebral vascular resistance.

Cerebral perfusion pressure is the difference between mean arterial pressure

(MAP) and intracranial pressure (ICP) or central venous pressure, whichever is

highest.

𝑪𝑷𝑷 = 𝑴𝑨𝑷− 𝑰𝑪𝑷

The normal intracranial pressure is maintained within 5-13 mmHg in adults with

only minor variation associated with arterial pulse pressure or change in body

posture (Partington and Farmery, 2014).

Under high blood pressure conditions, destructive increases in cerebral blood

flow are prevented by vasoconstriction and when the blood pressure decreases,

cerebral blood flow is maintained through vasodilatation of the cerebral

arteries. In addition cerebral blood flow is also dependent on the metabolic

needs of the brain (flow-metabolism coupling). Brain regions with higher

metabolic activities require more blood flow than other areas. The process of

Page 189: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

165

autoregulation involves various mechanisms including vascular and neural

systems to provide a beat-to-beat control over the cerebral arterial diameter.

The interplay between various factors in the regulation of cerebral blood flow

is illustrated in Figure 6-1.

Figure 6-1: Major physiological factors affecting cerebral blood flow and

the effects of vasoactive drugs.

Cerebral blood flow (CBF) is principally dependent on mean arterial pressure (MAP),

intracranial pressure (ICP) and cerebral vascular resistance (CVP). Autoregulation

maintains CBF by varying the cerebrovascular resistance (CVR) in spite of changes in

MAP within the autoregulatory range. Innervation of the cerebral arteries control

the vascular tone, and thus the CVR. Vasoactive drugs circulating in blood may affect

CBF through direct effect on CVR or indirectly by altering MAP through their effects

on cardiac output (CO) and/or total peripheral resistance (TPR). Some drugs may

also augment the neural effects on CBF.

The large pial arteries may not play a significant role in maintaining cerebral

blood flow since their diameters do not change in response to changes in

peripheral blood pressure (Giller et al., 1993). This assumption has been

employed in transcranial Doppler sonography which measures the blood flow

velocity in major arteries such as the middle cerebral artery as a surrogate

Page 190: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

166

marker of cerebral blood flow. Therefore, small branches of the large cerebral

arteries, such as branches of the middle cerebral artery, and arterioles may be

directly involved in the regulation of cerebral blood flow through compensatory

vasoconstriction or vasodilatation in response to decreases or increases in blood

pressure, respectively. Previous studies suggested that changes in cerebral

vascular resistance caused by systemic administration of vasoactive substances

such as adrenaline and angiotensin II are induced by the changes in arterial

blood pressure, not direct effects of the agents on cerebral arteries (Olesen,

1972). Most vasoactive agents circulating in blood have limited access to

cerebral vascular smooth muscle cells due to the presence of the blood-brain

barrier in the cerebral circulation (Pardridge, 2012). However, it has not been

clear whether vasoactive substances are exclusively prevented by the blood-

brain barrier from affecting the cerebral circulation. Further, instantaneous

onset of autoregulation by a change in blood pressure may augment any vascular

effect of the drug in the cerebral circulation. It has been reported that

autoregulation, as determined by continuous oximetry analysis, returns the

oxygen saturation disturbed by changes in arterial blood pressure to its basal

level in about 40-45 seconds (Ekström-Jodal, 1970). This study attempted to

dissect the direct versus indirect effects of vasoactive drugs by administering

them into systemic circulation via femoral vein or directly into the cerebral

circulation through the internal carotid artery. Drug administration through the

internal carotid artery can initially allow the distribution of vasoactive drugs to

the cerebral circulation undiluted which then cause localized effects on

cerebral arteries before affecting the peripheral circulation. Thus, drugs

administered via carotid artery should change the diameter of cerebral arteries

before affecting blood pressure, unlike simultaneous responses in cerebral

artery and blood pressure induced by intravenous administration of the drugs.

The aim of this study was to analyse the changes in diameter of rat small

cerebral arteries, as well as in mean arterial blood pressure, caused by the

infusion of vasoactive substances through the femoral vein or via direct delivery

into the cerebral circulation through the internal carotid artery.

Page 191: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

167

Materials and methods

6.2.1. Anaesthesia and pre-surgical preparation

The experiments were performed in accordance with the Australian code for

the care and use of animals for scientific purposes (8th edition, 2013, National

Health and Medical Research Council, Canberra) and were approved by The

University of Melbourne Animal Ethics Committee. Male or female Long Evans

rats (300-350 g) were deeply anaesthetized by intramuscular injection of a

mixture of ketamine (60 mg/kg) and xylazine (5 mg/kg). Throughout the

experiment, the body temperature was maintained at 37oC using a heat pad

regulated by feedback from a rectally inserted thermometer. The depth of

anaesthesia was regularly monitored and confirmed by lack of reaction to toe-

pinch tests, absence of blood pressure fluctuations in response to pinching and

regular observations of the animal for pupil dilatation. The anaesthesia was

administered every 30 min or as required.

6.2.2. Cannulation of arteries and veins

The right femoral artery was cannulated with a blunt-ended catheter

(inner/outer diameter: 0.28/0.61 mm, Microtube Extrusions, NSW, Australia)

and used for blood pressure monitoring. The right femoral vein was cannulated

and used for systemic administration of the drugs. The external carotid artery

was used for direct administration of drugs into the cerebral circulation. The

right common carotid artery, right external carotid artery and right internal

carotid artery were isolated and carefully separated from the adjacent vagus

nerves. The external carotid artery was clipped with a microvascular clip near

the bifurcation. A small puncture was made on the external carotid artery and

a PE-10 catheter was retrogradely inserted and advanced to the bifurcation

point of internal and external carotid arteries without blocking blood flow

through the internal carotid artery as shown in Figure 6-2. The catheter was

then secured in place by tying with a 6.0 silk suture and then applying a tissue

glue in order to avoid slipping back due to arterial pressure. The surgical wound

Page 192: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

168

area was sutured to avoid incidence of bleeding before the animal was turned

over into the prone position for cranial window preparation.

Figure 6-2: Carotid artery cannulation.

A catheter with a smaller tubing connected to the tip was inserted through the

external carotid artery and advanced to the point of the bifurcation of internal

and external carotid arteries without blocking the blood flow through the internal

carotid artery. Suture and tissue glue were used to secure the tubing in place and

prevent movements.

6.2.3. Cranial window preparation

After cannulation, the head of the animal was shaved and symmetrically fixed

by ear bars, front teeth and nosebar in a stereotactic frame (David Kopf® Small

animal stereotactic instrument, Model 900, Tujunga, USA). The skin overlying

the periosteum of the skull was removed with sharp scissors. The periosteum

was wiped away with gauze and the skull was covered with a squirt of hydrogen

peroxide in order to minimize bleeding. A small surgical cautery was used

whenever the bleeding was not stopped by the above means. A small area on

the left temporal bone (approximately 3 mm x 3 mm area: 0.80 mm posterior

Rostral

Page 193: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

169

and 1.5 mm lateral relative to bregma) was gradually thinned with a silicon

carbide grinding stone (Dremel 84922 silicon carbide grinding stone 4.8 mm,

Dremel, USA) attached to a drill (Dremel 300 Series variable speed rotary tool,

Dremel, USA) until the meningeal arteries could be observed through the skull

(Figure 6-3). A rubber polishing point (Dremel 462 Rubber Polishing Cone Point,

Dremel, USA) and/or a scalpel blade was then used to smooth the skull surface

in order to reduce reflection during the imaging process. The area was

continuously rinsed with sterile saline to avoid overheating of the skull and to

improve visualization of the field. Care was taken during the procedure to avoid

application of too much pressure that may crack the skull. The eyes of the

animal were covered with a transparent ophthalmic lubricant ointment to

prevent damage during the surgical procedure.

6.2.4. Imaging and image analysis

A standard Olympus stereo microscope fitted with a 10x magnification objective

was used to visualize the brain vessels through the skull with the help of a

Polychome V microscope illumination system (TILL Photonics LLC, Pleasanton,

CA, USA) as shown in Figure 6-3. A clear gel was applied to the skull as an

immersion medium into which the microscope objective was placed. The image

was captured with a Neo scientific CMOS camera (Andor Technology Ltd,

Belfast, UK) controlled by Metamorph® imaging software (Universal Imaging

Corporation, West Chester, PA). The arteries were differentiated from the veins

by the direction of their bifurcation (towards the midline for arteries) and by

the constriction induced when the animals inhaled a 100% oxygen for about 60

s. Images with 500 x 500 pixel frame were captured during the experiments and

stored for offline analysis. The images were analysed using ImageJ software

(U.S. National Institutes of Health, Bethesda, MA, USA). The image stacks were

initially averaged with the Grouped ZProjector plug-in and the diameters of the

arteries were obtained by selecting a line in the ‘analyse’ menu that

perpendicularly crossed through the vessel from edge to edge, followed by

selection of the ‘measure’ menu in the software. Data were then copied to a

spreadsheet for further statistical analyses.

Page 194: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

170

Figure 6-3: Cranial window preparation and imaging.

After the skin was removed from the skull, the cranial bone was gradually thinned

using a silicon carbide grinding stone attached to a driller and the area was

continuously flushed with saline to prevent heating of the skull (left panel). The

arteries were visualized with a stereo microscope fitted with a 10x magnification

objective lens (right panel). The image was captured with a Neo scientific CMOS

camera (Andor Technology Ltd, Belfast, UK) controlled by Metamorph® imaging

software (Universal Imaging Corporation, West Chester, PA).

6.2.5. Drug administration and haemodynamic monitoring

The animal was allowed to stabilize for at least 30 min post-surgery before

drugs were infused via the femoral vein or carotid artery using a syringe infusion

pump (Harvard Apparatus Inc., Holliston, MA). The blood pressure and heart

rate of the animal were continuously monitored during the experiment using a

pressure transducer (Transpact, Abbott Critical Care Systems, Sligo, IRE)

connected to femoral artery catheter and the data were recorded using

Page 195: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

171

PowerLab data acquisition system and Chart software (AD Instruments, Pty. Ltd,

Bella Vista, NSW, Australia). A stock concentration of each drug was made in

normal saline and was administered by continuous infusion via the femoral vein

or carotid artery. The doses were progressively increased by changing the

infusion rate after the blood pressure response had plateaued following each

previous dose rate. The rates of infusion used in the protocols were initially

determined by running pilot experiments wherein the drugs were administered

by step-by-step increment of the infusion rate until the blood pressure had been

changed by 15-20 mmHg. Drug administration was stopped when the mean

arterial pressure was increased to a maximum of 150-160 mmHg or decreased

to a minimum of 50 mmHg. Sufficient time was allowed for the effects of every

drug to wear off before another drug was tested. At the end of the experiments,

the rats were euthanized by an overdose of intravenous injection of

pentobarbitone (>100 mg/kg).

6.2.6. Experimental protocols

Rats were randomly assigned into groups of 5-7 for administration of

phenylephrine (0.3-10 µg/kg/min), prenalterol (0.03-10 g/kg/min),

formoterol (0.03-3 g/kg/min), vasopressin (0.1-10 ng/kg/min) or saline

(0.003-1 ml/kg/min) via the femoral vein or carotid artery. The animals

received the drugs in a randomized order allowing sufficient time between the

drugs for parameters to return to baseline. For femoral vein administration, the

drugs were infused at the same rate (0.003-1 ml/kg/min) with each rate lasting

for about 2 min before increasing to the next higher rate. However, the dose

rate varied for individual drugs. Only two doses of each drug were used for

intra-carotid administration. A separate group of animals received infusion of

the drugs at the same dose rates as intra-carotid infusion via the femoral vein

to compare the time-course of effects between peripheral (through femoral

vein) and local (through carotid artery) administration. The blood pressure and

heart rate were continuously monitored via a cannula inserted in the femoral

artery. Simultaneously, the images of the cerebral arteries corresponding to

the maximum effect of a specific doses of each drug were obtained through the

Page 196: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

172

cranial window before the infusion was started, before each increment of the

rate and before the infusion was stopped at the end of the experiment.

6.2.7. Statistical analysis

All data were expressed as mean ± SEM and the analysis was performed using

GraphPad Prism 5.0 (GraphPad software, San Diego, CA, USA). Values before

drug infusion were taken as baseline and the percentage changes of the mean

arterial blood pressure (MAP) and the cerebral artery diameter were calculated

for each animal. An unpaired t test was used to compare the intravenous and

intra-arterial effects of drugs between animals as required. P<0.05 was

considered significant in all cases.

Page 197: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

173

Results

6.3.1. Effects of intravenous vasoactive agents on blood

pressure and cerebral arterial diameter

The effects on blood pressure and cerebral artery diameter of phenylephrine (a

selective α1-adrenoceptor agonist drug), vasopressin (a nonspecific V1 and V2

receptor agonist peptide), prenalterol (a selective β1-adrenoceptor agonist

drug) and formoterol (a selective β2-adrenoceptor agonist drug) were

investigated in anaesthetized rats. The baseline mean arterial blood pressure

and heart rate diameters of cerebral arteries of the rats are shown in Table

6-1.

An equivalent rate of infusion of normal saline (0.003-1 ml/min; in half-log unit

increments) given through the femoral vein did not change the mean arterial

pressure and heart rate of the rats (Figure 6-5A). Phenylephrine (0.3-10

g/kg/min) induced a dose-dependent increase in mean arterial blood

pressure. The mean arterial blood pressure was increased by 81±7% after

intravenous infusion of phenylephrine 10 g/kg/min, but there was no change

in heart rate (Figure 6-6A). Similarly, intravenous administration of vasopressin

(0.1-10 ng/kg/min) increased the rat arterial blood pressure as shown in Figure

6-7A. Vasopressin 10 ng/kg/min increased the mean arterial pressure by 88±6%

before the infusion was stopped, but the heart rate was not significantly

changed. Infusion of prenalterol (0.03-0.3 µg/kg/min) increased the mean

arterial pressure by 21±5%. The heart rate was significantly increased by 29±6%

after infusion of prenalterol 10g/kg/min (Figure 6-8A). The blood pressure was

decreased by about 24±9% after intravenous administration of formoterol.

Formoterol 0.3 µg/kg/min increased the heart rate by 10±0.6% (Figure 6-9A).

The diameter of small distal branches of the left middle cerebral artery were

monitored through the cranial window as the drugs were infused. Images were

taken before the drug infusion was increased to the next higher rate or dose,

or before stopping the last dose (Figure 6-4). The average baseline outer

Page 198: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

174

diameter of the arteries ranged from 50-70 µm (Table 6-1). The diameter of rat

cerebral arteries was unaffected by intravenous infusion of normal saline (up

to a maximum rate of 1 ml/min; P>0.05, compared to baseline) (Figure 6-5B).

Phenylephrine, vasopressin and prenalterol decreased cerebral artery

diameter. The rat cerebral arteries were dilated by infusion of β2-adrenoceptor

agonist formoterol. Phenylephrine 10 µg/kg/min constricted the arteries by

22±3% of the baseline diameter (Figure 6-6B), while the maximum constriction

induced by vasopressin 10 ng/min was 20±5% (Figure 6-7B). It appeared that the

cerebral artery started to constrict before mean arterial pressure increased

upon infusion of vasopressin 1 ng/kg/min – increases in blood pressure were

observed only after the dose rate was increased to 3.2 ng/kg/min. Prenalterol

10 µg/kg/min decreased the cerebral artery diameter by 8±4% at the maximum

infusion rate (Figure 6-8B). The arteries were dilated by 13±5% after infusion of

formoterol (0.03-0.3µg/kg/min) (Figure 6-9B).

Table 6-1: Baseline values of rat blood pressure, heart rate and cerebral

artery diameter before intravenous administration of the

drugs.

S

(n=6)

PE

(n=7)

PR

(n=7)

FO

(n=6)

AVP

(n=5)

MAP (mmHg) 98±6 97±3 77±2 87±3 76±4

HR (bpm) 328±19 343±18 337±5 323±11 350±18

D (m) 50±4 70±15 67±14 63±13 68±14

S, saline; PE, phenylephrine; PR, prenalterol; FO, formoterol; AVP, vasopressin; MAP, mean arterial pressure; HR, heart rate; D, cerebral artery outer diameter: and n,

number of rats in each group. Data are shown and mean±SEM.

Page 199: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

175

Figure 6-4: Representative images of cerebral arteries illustrating the

effects of vasoactive drug infusion

The images show the diameter of cerebral arteries before and after intravenous

infusion of phenylephrine 10 g/kg/min (top) and formoterol 0.3 g/kg/min

(bottom) in different rats.

Page 200: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

176

Figure 6-5: Effects of saline on blood pressure, heart rate and cerebral

artery outer diameter.

Effects of equivalent rate of intravenous saline administration on A) mean arterial

pressure (MAP) and heart rate (HR); and B) cerebral artery outer diameter (D) in the

rat. Normal saline was infused through the femoral vein in anaesthetised rats in

increasing rate, each rate lasting for 2 min. Data are expressed as % change from

the baseline MAP, HR or D. Vertical bars are ±SEM. n, number of animals in the group.

Page 201: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

177

Figure 6-6: Effects of phenylephrine on blood pressure, heart rate and

cerebral artery outer diameter.

Effects of intravenous phenylephrine administration on A) mean arterial pressure

(MAP) and heart rate (HR); and B) cerebral artery outer diameter (D) in the rat.

Phenylephrine was infused through the femoral vein in anaesthetised rats in

increasing doses, each dose rate lasting for 2 min. Data are expressed as % change

from the baseline MAP, HR or D. Vertical bars are ±SEM. n, number of animals in the

group.

A

Page 202: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

178

Figure 6-7: Effects of vasopressin on blood pressure, heart rate and

cerebral artery outer diameter.

Effects of intravenous vasopressin administration on A) mean arterial pressure (MAP)

and heart rate (HR); and B) cerebral artery outer diameter (D) in the rat. Vasopressin

was infused through the femoral vein in anaesthetised rats in increasing dose, each

dose rate lasting for 2 min. Data are expressed as % change from the baseline MAP,

HR or D. Vertical bars are ±SEM. n, number of animals in the group.

Page 203: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

179

Figure 6-8: Effects of prenalterol on blood pressure, heart rate and

cerebral artery outer diameter.

Effects of intravenous prenalterol administration on A) mean arterial pressure (MAP)

and heart rate (HR); and B) cerebral artery outer diameter (D) in the rat. Prenalterol

was infused through the femoral vein in anaesthetised rats in increasing dose, each

dose rate lasting for 2 min. Data are expressed as % change from the baseline MAP,

HR or D. Vertical bars are ±SEM. n, number of animals in the group.

Page 204: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

180

Figure 6-9: Effects of formoterol on blood pressure, heart rate and

cerebral artery outer diameter.

Effects of intravenous formoterol administration on A) mean arterial pressure (MAP)

and heart rate (HR); and cerebral artery outer diameter (D) in the rat. Formoterol

was infused through the femoral vein in anaesthetised rats in increasing dose, each

dose rate lasting for 2 min. Data are expressed as % change from the baseline MAP,

HR and D. Vertical bars are ±SEM. n, number of animals in the group.

Page 205: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

181

6.3.2. Intravenous versus intra-arterial effects of drug

infusion on blood pressure and cerebral arterial

diameter

In an attempt to ascertain the direct effects on the cerebral arteries, drugs

were directly infused into the cerebral circulation through the carotid artery.

Only two doses of each drug were given in order to minimize the redistribution

the drugs to the peripheral circulation and thereby change the blood pressure

before affecting cerebral arteries. Separately, the drugs were infused at the

same dose rate via the femoral vein to compare with the intra-carotid artery

administration. The baseline mean arterial blood pressure and heart rate of

each group before infusion of the drugs are shown in Table 6-2. The drugs were

infused at the following dose rates, each lasting for 2 min, or until the maximum

limit of blood pressure was reached: phenylephrine (3 and 10 µg/kg/min),

vasopressin (3 and 10 ng/kg/min) and formoterol (0.3 and 1 µg/kg/min).

Intravenous and intra-arterial phenylephrine (3 µg/kg/min) increased the blood

pressure by 32±10% and 14±4%, respectively, but no statistically significant

difference was found between the two routes (P>0.05, unpaired t test).

Similarly, the increase in mean arterial pressure caused by phenylephrine (10

µg/kg/min) administration was comparable by intravenous (81±14%) and intra-

arterial (80±10%) administration (Figure 6-10A). The respective increase in blood

pressure induced by vasopressin (3 ng/kg/min) was 12±4% and 17±1% when

administered intravenously and intra-arterially. Similarly, at the 10 ng/kg/min

dose rate, vasopressin caused an increase in mean arterial pressure of 69±12%

by intravenous infusion and 84±9% after intra-arterial infusion (Figure 6-11A) –

no significant difference was found between the two routes (P>0.05).

Intravenous and intra-arterial infusion of formoterol (0.3 µg/kg/min) decreased

the blood pressure by 1±1% and 7±3%, respectively. The blood pressure was

decreased by similar extent when the dose of formoterol was increased to 1

µg/kg/min (intravenous, -37±7%; and intra-arterial, -43±13%) as shown in Figure

Page 206: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

182

6-12A. Rat blood pressure was unaffected by the same rate of saline infusion –

intravenous or intra-arterial.

Table 6-2: Baseline rat blood pressure and heart rate before intra-femoral

vein or intra-carotid artery administration of the drugs

S (n=5-7)

PE (n=5-7)

FO (n=5-7)

AVP (n=5-6)

MAPi.v.(mmHg) 85±2 92±7 98±6 86±2

MAPi.a.(mmHg) 74±6 83±5 89±7 77±3

HRi.v. (bpm) 348±22 346±12 322±37 335±19

HRi.a. (bpm) 337±6 352±23 334±15 354±17

S, saline; PE, phenylephrine; PR, prenalterol; FO, formoterol; AVP, vasopressin; MAP, mean arterial pressure; HR, heart rate; i.v., intravenous; i.a., intra-arterial; and n, number of rats in each group.

Both phenylephrine and vasopressin induced constriction of rat cerebral

arteries when administered through the femoral vein or carotid artery. The

changes in arterial diameter were similar for both intravenous and intra-arterial

phenylephrine (3 µg/kg/min) infusion (-8±3% for i.v.). At the higher dose of

phenylephrine (10 µg/kg/min), greater constriction of cerebral arteries was

obtained upon direct infusion into the cerebral circulation via the carotid artery

(-30±3%) than systemic administration through femoral vein (-16±2%, P<0.05,

unpaired t test) (Figure 6-10B).

Vasopressin (3 ng/kg/min) caused comparable decreases in cerebral arterial

diameter by intravenous (-3±2%) and intra-arterial routes of infusion (-13±5%,

P>0.05, unpaired t test). Likewise, vasopressin (10 ng/kg/min) induced a similar

degree of constriction by intravenous (-22±3%) and intra-arterial administration

(-36±6%) (Figure 6-11B).

Infusion of formoterol (0.3 µg/kg/min) increased the cerebral arterial diameter

by 8±9% and 6±4% when given by intravenous and intra-arterial routes,

Page 207: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

183

respectively. A further dilatation was induced by increasing the dose of

formoterol to 1 µg/kg/min, either intravenous (37±7%) or intra-arterial (41±3%)

(Figure 6-12B), but there was no statistically significant difference between the

two routes. No observable change in rat cerebral artery diameter was induced

by infusion of saline at the same volume rate as other drugs (P>0.05, compared

to baseline).

Page 208: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

184

Figure 6-10: Intravenous versus intra-arterial effects of phenylephrine

on blood pressure and cerebral artery outer diameter.

Comparison of the effects of intravenous (via femoral vein) and intra-arterial (via

carotid artery) phenylephrine administration on A) mean arterial pressure (MAP);

and B) cerebral artery outer diameter (D) in the rat. Phenylephrine was infused at

two dose rates (3 and 10 µg/Kg/min), each dose lasting for 2 min. Data are expressed

as % change from the baseline MAP or D values. Vertical bars are ±SEM. *P<0.05,

unpaired t test. n, number of animals in the group.

Page 209: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

185

Figure 6-11: Intravenous versus intra-arterial effects of vasopressin on

blood pressure and cerebral artery outer diameter.

Comparison of the effects of intravenous (via femoral vein) and intra-arterial (via

carotid artery) vasopressin administration on A) mean arterial pressure (MAP); and

B) cerebral artery outer diameter (D) in the rat. Vasopressin was infused at two dose

rates (3 and 10 ng/Kg/min), each dose lasting for 2 min. Data are expressed as %

change from the baseline MAP or D values. Vertical bars are ±SEM. n, number of

animals in the group.

Page 210: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

186

Figure 6-12: Intravenous versus intra-arterial effects of formoterol on

blood pressure and cerebral artery outer diameter.

Comparison of the effects of intravenous (via femoral vein) and intra-arterial (via

internal carotid artery) formoterol administration on A) mean arterial pressure

(MAP); and B) cerebral artery outer diameter (D) in the rat. Formoterol was infused

at two dose rates (0.3 and 1 µg/Kg/min), each dose lasting for 2 min. Data are

expressed as % change from the baseline MAP or D values. Vertical bars are ±SEM. n,

number of animals in the group.

Page 211: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

187

6.3.3. Time-course of effects of vasopressor agents on blood

pressure and cerebral artery diameter

The mean arterial pressure and cerebral artery diameter were measured every

30 seconds to analyse the time-course effect of the drugs at two rates (low-

and high-rate) of infusion each lasting for 2 min. The blood pressure and arterial

diameter were only slightly affected by the first phase of phenylephrine (3

µg/min) (Figure 6-13A), vasopressin (3 ng/min) (Figure 6-14A) and formoterol

(0.3 µg/min) (Figure 6-15A). Increasing the infusion rate by half an order of

magnitude (half-log10 unit increments) markedly affected both the blood

pressure and vessel diameter. The onset of action was similar for both

intravenous and intra-arterial administration of the drugs. Each drug appeared

to cause a similar pattern of change (slope of the curves) in mean arterial

pressure and cerebral artery diameter after intravenous or intra-arterial

administration. Phenylephrine caused a significantly greater maximum

constriction by intra-carotid than by intravenous administration (Figure 6-13B).

However, the maximum effects on cerebral artery diameter were similar by

either intravenous or intra-arterial administration for both vasopressin (Figure

6-14B) and formoterol (Figure 6-15B)

Page 212: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

188

Figure 6-13: The time-course of effects of intravenous and intra-arterial

phenylephrine on blood pressure and cerebral artery outer

diameter.

The time-course effect of phenylephrine infusion via femoral vein or carotid artery

on A) mean arterial pressure (MAP); and B) cerebral artery outer diameter (D) in the

rat. Phenylephrine was infused at 3 and 10 µg/kg/min rates and the measurements

taken every 30 s. Data are expressed as % change from the baseline MAP or D.

Vertical bars are ±SEM. *P<0.05, unpaired t test between intravenous and intra-

arterial routes for each time point. n, number of animals in each group.

Page 213: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

189

Figure 6-14: The time-course of effects of intravenous and intra-arterial

vasopressin on blood pressure and cerebral artery outer

diameter.

The time-course effect of vasopressin infusion via femoral vein or carotid artery on

the rat mean arterial pressure (MAP) and cerebral artery outer diameter (D) in the

rat. Vasopressin was infused at 3 and 10 µg/kg/min rates and the measurements

taken every 30 s. Data are expressed as % change from the baseline MAP or D.

Vertical bars are ±SEM. n, number of animals in each group.

Page 214: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

190

Figure 6-15: The time-course of effects of intravenous and intra-arterial

formoterol on blood pressure and cerebral artery outer

diameter.

The time-course effect of formoterol infusion via femoral vein or carotid artery on

A) mean arterial pressure; and B) cerebral artery outer diameter (D) in the rat.

Formoterol was infused at 0.3 and 1 µg/kg/min rates and the measurements taken

every 30 s. Data are expressed as % change from the baseline MAP or D. Vertical bars

are ±SEM. n, number of animals in each group.

Page 215: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

191

Discussion

In this study, the blood pressure and cerebrovascular responses to systemically

administered vasoactive agents in anaesthetised rats were investigated. The

cranial window was made to visualize the vessels through a ‘thinned’ but intact

skull without releasing the intracranial pressure. The current results

demonstrate that administration of vasoactive drugs in vivo affects both the

peripheral blood pressure and diameter of cerebral arteries. The changes in

calibre of cerebral arteries appear to be indirect autoregulatory responses to

changes in peripheral blood pressure induced by vasoactive agents.

Phenylephrine and vasopressin increase arterial blood pressure by inducing

vasoconstriction of peripheral arteries through activation of α1-adrenoceptors

and V1 receptors, respectively. Formoterol decreases the blood pressure by

activating β2-adrenoceptors that mediate dilatation of peripheral arteries.

Prenalterol (a β1-adrenoceptor agonist) does not cause significant effects in

peripheral arteries, but moderately increases blood pressure due to its positive

chronotropic and inotropic effects (Hedberg et al., 1982). In this study,

intravenous administration of phenylephrine and vasopressin induced

comparable increases in arterial blood pressure with simultaneous constriction

of the cerebral arteries, while formoterol induced hypotension along with

cerebral vasodilatation. Prenalterol had weak effects on blood pressure as well

as diameter of the rat cerebral arteries. Cerebral arteries can be constricted

or dilated either by direct actions of vasoactive agents or in response to changes

in peripheral blood pressure, or both. In vitro studies have shown the presence

in rat cerebral arteries of α1-adenoceptors mediating constriction and β1 and

β2-adrenoceptors mediating dilatation as discussed in Chapter 4. Vasopressin

also induces a potent constriction of cerebral arteries in vitro (see Chapter 3).

There were simultaneous and parallel changes in blood pressure and cerebral

arterial diameter induced by intravenous administration of the agents. As a

result, it is not possible to distinguish whether the cerebrovascular effects are

caused by direct actions of the vasoactive drugs on cerebral arteries or are

Page 216: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

192

autoregulatory responses induced by blood pressure changes. Further, the

direct vascular effects of the drugs may be concealed due to the autoregulatory

responses since the agents were administered over a wide range of doses for

long period of time. Thus, higher doses of the vasoactive drugs were directly

infused through the carotid artery so that any direct effects of the drugs on

cerebrovascular smooth muscle would be observed before affecting the blood

pressure due to redistribution to the peripheral circulation. Again, there were

no differences in the onset time of the effects of the drugs on blood pressure

and diameter changes between intravenous and intra-carotid administration.

There were no significant differences in the magnitude of responses between

the two routes except that intra-arterial phenylephrine (10 µg/kg/min) caused

a significantly greater constriction of cerebral arteries than intravenous

administration.

These findings show that the cerebrovascular reactions to systemic vasoactive

drug administration are predominantly indirect autoregulatory responses to the

changes in peripheral blood pressure or perfusion pressure rather than direct

effects on the arterial smooth muscle cells. The presence of a selective blood-

brain barrier in cerebral arteries prevents most substances circulating in the

blood from accessing the vascular smooth muscle cells (discussed in section

1.3). While phenylephrine, formoterol or prenalterol can exert their effect on

cerebral vasculature in vitro, their direct in vivo effect in cerebral arteries may

be prevented by the blood-brain barrier. Phenylephrine does not cross the

blood-brain barrier under normal conditions, but higher blood pressure may

disrupt the integrity of the blood-brain barrier such that circulating drugs can

access the cerebrovascular smooth muscle and induce vasoconstriction as

discussed below. Similarly, vasopressin causes a potent constriction of isolated

cerebral arteries but its direct effect on cerebral vasculature in vivo may

limited by the blood-brain barrier (Zaidi and Heller, 1974). Infusion of

vasopressin has been reported to cause endothelium-dependent dilatation of

large cerebral arteries but increase the resistance of small arteries due to

activation of cerebral autoregulation in response to increased arterial pressure

(Faraci and Heistad, 1990). However, the results from this study do not reflect

Page 217: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

193

direct effects of vasopressin on cerebral arteries since no difference was found

between intravenous and intra-carotid administration of vasopressin.

Nevertheless, the possibility of overlap between the autoregulatory response

and direct constriction induced by the drugs cannot be excluded. Differences

in the degree of vasoconstriction induced by phenylephrine and vasopressin in

spite of similar changes in arterial blood pressure may indicate additional

effects related to direct actions of the agents on the cerebral vasculature.

Elevated arterial blood pressure may disrupt the function of the blood-brain

barrier so that vasoactive agents may directly affect the tone of cerebral

arteries (Ito et al., 1980; Tsai et al., 1989; Cole et al., 1991). Previous studies

have shown that acute vasopressor-induced elevation of blood pressure or

chronic hypertension increased the permeability of the blood-brain barrier

(Domer et al., 1980; Johansson, 1980). For example, infusion of phenylephrine

(30 µg/kg/min) for 5 min induced a severe increase in rat blood pressure (from

122 to 187 mmHg) that was associated with disruption of the blood-brain barrier

that led to the leakage of fluorescent albumin (Mayhan, 1991). Similarly,

vasopressin and adrenaline caused disruption of the blood-brain barrier after

haemorrhagic circulatory arrest in pigs (Semenas et al., 2014). In this study, rat

blood pressure was increased to about 150-160 mmHg by phenylephrine and

vasopressin. This may be an additional reason why intra-carotid administration

of vasopressin tended to cause a slightly greater constriction of cerebral artery

than intravenous administration in spite of similar effects on blood pressure

induced by the two drugs. However, it has been shown that there were more

disruptions of the blood-brain barrier after a rapid increase than after stepwise

increase in blood pressure (Baumbach and Heistad, 1985). Thus, the

contribution of the direct effects of vasoactive drugs to overall cerebrovascular

responses as a consequence of blood-brain barrier disruption was minimized in

this study because the drugs were infused gradually in increasing doses. The

extent of leakage through a pressure-disrupted blood-brain barrier may be

variable for different vasoactive agents and requires further investigation.

Page 218: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

194

Additionally, this study emphasizes the greater role of small calibre cerebral

arteries rather than large cerebral arteries in the regulation of cerebral blood

flow in face of changing systemic blood pressure as previously indicated

(Olesen, 1972; Auer et al., 1987). Small branches of the rat middle cerebral

arteries responded to changes in arterial pressure by constriction or dilatation,

which are critical elements of cerebral autoregulation. The size dependence of

responses in cat pial arteries was previously reported by Kontos et al. (1978),

but only vessels larger than 200 µm in diameter responded to changes in arterial

blood pressure. Small arteries of less than 100 µm dilated when the pressure

was less than 90 or greater than 170 mmHg. The difference between the

previous study and our results may be related to species and/or methodological

differences. Kontos et al. used a cranial window in the cat where the

intracranial pressure was artificially controlled through catheter inserted

through the skull. However, the cranial window in this study was prepared in

rat without breaking the cranial bone, thus maintained a natural intracranial

pressure. This study also allowed the normal variation of intracranial pressure

due to changes in arterial blood pressure.

The current study has further implications to the clinical practice wherein

vasoactive drugs are sometimes administered through the carotid artery in

order to obtain localized effects in the cerebral circulation, or to minimize their

systemic adverse effects. For example, intra-arterial nimodipine or nicardipine

has been used to prevent or reverse the cerebral vasospasm following

subarachnoid haemorrhage (Biondi et al., 2004). It has been shown that

nicardipine induces angiographic cerebral vasodilatation as well as hypotension

which indicates that a decrease in arterial pressure may have induced the

cerebrovascular dilatation by activating autoregulation (Linfante et al., 2008).

Thus, intra-carotid administration of vasoactive drugs may not cause local

effects in the cerebral circulation unless the agents are highly lipophilic and

cross the blood-brain barrier (Pluta et al., 1997).

In summary, systemic administration of vasoactive drugs primarily affects the

systemic arterial pressure which then induces the autoregulatory response in

Page 219: Cerebrovascular effects of vasoactive drugs

Vasoactive agents in vivo

195

cerebral arteries. Small calibre cerebral arteries may play a significant role in

the regulation of cerebral blood flow.

Page 220: Cerebrovascular effects of vasoactive drugs
Page 221: Cerebrovascular effects of vasoactive drugs

197

Effects of vasopressor agents on

human cerebral oxygen

saturation during anaesthesia

Page 222: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

198

Introduction

7.1.1. Background to clinical problem

In healthy subjects, a stable cerebral blood flow of approximately 50 ml/100

g/min is maintained in the face of considerable fluctuations in peripheral

arterial blood pressure (Lassen, 1959). This autoregulation occurs within the

mean arterial blood pressure range of 60-150 mmHg, and adjusts perfusion for

normal physiological changes in brain function including intellectual work,

sensory stimulation, muscle movement and sleep (Lassen, 1974). However,

when mean arterial pressure is outside the autoregulatory range, changes in

cerebrovascular resistance are unable to compensate for extreme mean arterial

pressure fluctuations, and perfusion becomes pressure-passive. The lower limit

of the autoregulatory blood pressure threshold varies significantly in individuals

(Drummond, 1997), and is shifted to the right (higher blood pressure) in disease

states such hypertension and diabetes (Bentsen et al., 1975). In episodes of

acute hypotension due to haemorrhage or overuse of vasoactive drugs, cerebral

hypoperfusion may occur, potentially leading to cerebral ischaemia and

infarction (Agnoli et al., 1968; MacKenzie et al., 1979). Further, the

autoregulation can be disrupted by certain disease conditions such as head

injury, acute ischaemic stroke, brain tumour or haematoma (Paulson et al.,

1989; Paulson et al., 1990; Carnevale and Lembo, 2010).

The concept of cerebral perfusion pressure is central to cerebral protection,

and states that cerebral blood flow is determined primarily by the pressure

gradient across the cerebral vascular bed (Moraine et al., 2000). The driving

force for flow equates to the difference between mean arterial pressure and

intra-cranial pressure or central venous pressure. The normal value of

intracranial pressure in adults who are in the supine position ranges from 7-10

mmHg, but can be elevated in pathological conditions such as traumatic brain

injury. Maintaining cerebral blood flow through prevention of hypotension and

by lowering elevated intracranial pressure is a priority in the management of

acute brain injury, particularly in traumatic brain injury and subarachnoid

Page 223: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

199

haemorrhage. Current guidelines recommend mean arterial pressure to be

maintained above 90 mmHg (Bratton et al., 2007), but ensuring the intracranial

pressure should be less than 20 mmHg (Kirkman and Smith, 2014).

General anaesthesia may alter cerebral autoregulation with effects on cerebral

vascular reactivity and cerebral perfusion pressure. The lower limit of

autoregulation may be increased by both volatile anaesthetic agents (Dagal and

Lam, 2009) and by hypocapnia (Meng and Gelb, 2015). In some patients

excessive hypotension may occur during surgery placing them at risk of cerebral

ischaemia. This is of particular concern when anaesthetized patients are placed

into an upright “beach chair” or “barber’s chair” position, as commonly

employed in arthroscopic shoulder procedures. The “beach chair” position is

associated with significant hypotension, due to gravitational pooling of blood in

the lower parts of the body (Buhre et al., 2000). The risk of cerebral

hypoperfusion and ischaemic injury with the beach chair position was first

reported in a case series by Pohl and Cullen (2005). There has since been

significant discussion relating to the safe management of patients in the beach

chair position and importantly how to monitor for cerebral ischaemia (Cullen

and Kirby, 2007). The seriousness of this problem was illustrated by a recent

death of a 50 year old man after undergoing surgery in the beach chair position

(The Australian, 2013).

In recent years the monitoring of cerebral oxygen saturation using near-infrared

spectroscopy (NIRS) has emerged as a potential monitor of intraoperative

cerebral blood flow during anaesthesia in the chair position (the principle of

NIRS oximetry is discussed in the following section). It has been shown that

patients undergoing shoulder surgery in the beach chair position had lower

cerebral oxygen saturation compared to the lateral decubitus position (Laflam

et al., 2015). Unlike transcranial Doppler measurements that are based on

determination of the red blood cell velocity, cerebral oximetry measures

haemoglobin oxygen saturation within the frontal lobe microcirculation. Though

NIRS provides an indirect measurement of tissue perfusion, continuous

Page 224: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

200

monitoring of regional cerebral oxygen saturation however has the potential to

detect episodes of silent ischaemia during surgery.

A current approach to protect patients during beach chair surgery is to increase

cerebral perfusion pressure with vasopressor drug administration (Murphy and

Szokol, 2011). Clinically used drugs include the sympathomimetics

noradrenaline, phenylephrine and ephedrine, and the vasoactive peptide

vasopressin. Whilst noradrenaline and phenylephrine are effective in increasing

arterial blood pressure, recent studies indicate these drugs may also induce

cerebral oxygen desaturation (Brassard et al., 2009; Meng et al., 2011; Soeding

et al., 2013). Vasopressor agents have the potential to constrict both systemic

and cerebral arteries. In normal patients, however, the unique structural blood-

brain barrier prevents most blood-borne substances, including catecholamines,

from accessing the cerebral vascular smooth muscle and brain parenchymal

tissue. In pathological conditions such as traumatic brain injury or subarachnoid

haemorrhage the permeability of the blood-brain barrier to circulating drugs

may be altered (Hardebo and Owman, 1980; Myburgh, 2005). The question

arises whether all vasopressor drugs adversely alter regional cerebral perfusion,

either via direct constriction of cerebral arteries, or indirectly by changing

arterial blood pressure, and importantly in relation to differences in the doses

of each agent used. This study employed a NIRS oximetry technique to analyse

the effect of vasopressor agents on cerebral oxygen saturation.

7.1.2. Principle of cerebral oximetry

In cardiac surgery, NIRS monitoring is becoming routine management (Douds et

al., 2014) and it is increasingly being used during shoulder surgery. It is a

noninvasive technique based on the use of near infrared light (700-1000 nm)

reflectance spectroscopy. The NIRS optode, consisting of a light emitter and

detector(s) separated by 3-4 centimetres, is placed on the forehead overlying

the prefrontal cortex. Near-infrared light is transmitted through the scalp and

is finally reflected back to the detectors which measure the intensity (Figure

7-1). The reflected light is scattered and its intensity is attenuated as it passes

Page 225: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

201

through the scalp due to oxygen-dependent absorption by certain

chromophores, such as haemoglobin, myoglobin and cytochrome oxidase (Delpy

and Cope, 1997). Oxygenated and deoxygenated haemoglobin have different

absorption spectra, thus their relative concentration can be determined based

on the degree of differential near-infrared light absorption. This can be used as

a marker to monitor changes in cerebral blood flow (Ferrari et al., 2004;

Highton et al., 2010).

The NIRS equipment available to date varies in the analysis of the reflected NIR

light. Currently, there are four methods used in NIRS oximetry – continuous

wave, frequency domain, time resolution and spatially resolved spectroscopy.

In the continuous wave technique, the NIR light is emitted at a constant

frequency and amplitude and the changes in light intensity are related to

changes in haemoglobin concentration (Scholkmann et al., 2014). The

frequency domain technique employs an amplitude modulated light (50 to 500

MHz) and relates the changes in amplitude and phase of the back-scattered

signal to changes in haemoglobin concentration (Delpy and Cope, 1997). Time

resolution spectroscopy involves emission of a short pulse NIR (<100

picoseconds) and calculates the haemoglobin concentration based on temporal

delay of the light propagated through the tissue (Wabnitz et al., 2010). The

spatially-resolved spectroscopy (SRS) technique uses a complex algorithm that

determines the absolute concentration of haemoglobin and oxyhaemoglobin

based on localized attenuation of the NIR light. The light attenuation is

measured as a function of distance using multiple detectors; i.e., the slope of

NIR light attenuation vs distance from the light source is used to determine the

absolute ratio of oxyhaemoglobin to deoxyhaemoglobin concentration (Highton

et al., 2010). Thus, regional cerebral oxygen saturation (rScO2) can be

calculated using the following relationship:

Page 226: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

202

𝒓𝑺𝒄𝑶𝟐 =[𝑶𝟐𝑯𝒃]

[𝑶𝟐𝑯𝒃+𝑯𝑯𝒃]× 𝟏𝟎𝟎%,

where, O2Hb is oxyhaemoglobin and HHb is deoxyhaemoglobin.

Using NIRS for monitoring cerebral oxygen has several limitations. The most

common drawback of this technology is the possibility of interference from

extracranial tissues such as the scalp. To overcome this problem, multiple light

emitting and detector systems are used to identify extracranial reflectance. A

special algorithm that subtracts the light absorbed by the scalp from the entire

absorption is used to determine the oxygen-dependent absorption of NIR light

by the brain tissues (Figure 7-1) (Zheng et al., 2013). Nevertheless, NIRS

oximetry a non-invasive method that offers a direct and monitoring of cerebral

oxygen saturation. It can be used to continuously monitor the dynamic changes

in cerebral oxygen saturation induced by postural changes or vasoactive drug

administration in neurocritical care.

The aim of this study was to compare the effects of clinically-used vasopressor

drugs on cerebral oxygen saturation in anaesthetised subjects during both

supine and beach chair positioning. A primary question was to identify whether

vasopressor drug infusion could prevent postural hypotension associated with

head-up positioning, and secondly identify the effect of each drug on the

incidence of cerebral oxygen desaturation in the supine and beach chair

positions.

Page 227: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

203

Figure 7-1: Application of near infrared spectroscopy (NIRS) for

monitoring regional cerebral oxygen saturation.

The setup consists of a 2-channel Equinox (Model 7600) oximetry device set (top left)

with the optode placed on each side of the forehead in a “beach chair”-seated

subject (top right). The components of the optode - a NIR light source and dual

detectors - are shown (lower left). The first detector located closer to the source

detects light reflected from the superficial tissue (skin/skull/dura), whilst the

second sensor located 3-4 cm further from the emitter detects light reflected from

deep brain tissues (lower right).

Page 228: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

204

Materials and methods

7.2.1. Patient recruitment

After institutional approval was obtained from the Melbourne Health and

Clinical Trial Registration Ethics Committee (ANZCTR No 12610001075077), 24

patients undergoing elective shoulder arthroscopy in the upright position were

recruited for this study. A written informed consent was obtained from each

patient. Patients were excluded if there was a history of cerebrovascular event,

significant cardiac disease (New York Heart Association symptoms class 3, or

pacemaker), carotid endarterectomy, uncontrolled hypertension, uncontrolled

diabetes, contraindications to interscalene block or a body mass index (BMI)

≥35.

7.2.2. Anaesthesia and patient preparation for experiments

Patients were randomized to receive an infusion of either saline, noradrenaline

(Levophed®, Hospira Ptd Ltd, VIC, Australia) or vasopressin (Pitressin®, Link

Medical Products Pty. Ltd, NSW, Australia) following induction of general

anaesthesia. A standardized sevoflurane general anaesthetic technique,

combined with interscalene brachial plexus anaesthesia, was used in all

patients (Figure 7-2). On arrival to the operating theatre, patients received

warmed Hartmann’s solution 15 ml/kg intravenously (i.v.) and were sedated

with midazolam 0.05 mg/kg. Interscalene brachial plexus anaesthesia was

performed under ultrasound guidance, using 0.75% ropivacaine (30–35 ml). The

radial artery was directly cannulated for continuous arterial pressure

monitoring. The pressure transducer was positioned at the level of the tragus

throughout surgery to reflect cerebral perfusion pressure. Near-infrared

spectroscopy (NIRS) optode sensors (EQUINOX 7600, Nonin Medical Inc,

Plymouth, MA USA) were placed bilaterally on each forehead 1–2 cm above the

brow, and sensor edges taped to prevent interference from ambient light.

Cerebral oxygen saturation (ScO2) was measured from both left and right

sensors. Systemic pulse oximetry, ECG, and bispectral index monitoring for

Page 229: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

205

depth of anaesthesia (BIS, Aspect Medical Systems, Newton, MA, USA) were

applied.

The ScO2 and haemodynamic variables were measured with patients inspiring

room air, and after pre-oxygenation as shown in Figure 7-3. General anaesthesia

was induced intravenous administration of fentanyl (1 mg/kg ), propofol (1.5–

2.0 mg/kg) and atracurium (0.5 mg/kg), and the trachea intubated. Patients

were ventilated with oxygen and the end-tidal carbon dioxide maintained at

4.7–5.3 kPa. Anaesthesia was maintained with sevoflurane by targeting end

tidal concentration (ETSEVO) at 1.5 % and BIS value at 40-60%. ScO2, and

haemodynamic parameters were continuously monitored. Temperature was

monitored via a nasopharyngeal probe.

7.2.3. Experimental protocols

After induction of anaesthesia, the patient was observed for a period of 5-10

min during which time mean arterial pressure, heart rate and anaesthetic depth

stabilised, before the study drug was infused (Figure 7-2). The drugs were

diluted with sterile saline in a 60 ml syringe to make the following

concentrations: noradrenaline 20 µg/ml or vasopressin 0.3 units/ml (1 unit

vasopressin corresponds to 17 µg). The vasopressor drug, or control saline was

infused by increasing the rates in 5 steps to a maximum dose over 15 min. At

maximum infusion rate, noradrenaline infusion was 0.3 µg/kg/min and

vasopressin 4 milliunits/kg/min. Measurements were taken before the infusion

was started and just before the infusion rate was increased to the next higher

level. The patient was then quickly placed in the beach chair position while the

last infusion was continued (Figure 7-2).

Page 230: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

206

Figure 7-2: Study protocol for patients randomized to receive either

saline, noradrenaline or vasopressin infusion.

Interscalene brachial plexus anaesthesia was initially performed under ultrasound

guidance while the patient was spontaneously breathing. General anaesthesia was

induced after the patient was pre-oxygenated. About 5-10 min was allowed for

haemodynamic stabilization before the infusion was started. A vasopressor drug or

saline was continuously administered with gradual increment in rate of infusion,

each rate lasting for 2.5 min. The patient was then placed into the beach chair

position (BCP) while the drug or saline was being infused at the maximum rate. The

MAP, HR, ScO2, BIS, ETCO2, ETSEVO, Paw and temperature were continuously monitored

while the patient was lying supine as well as in the beach chair position. The diagram

is only informative about the sequence of activities and not drawn to scale. MAP,

mean arterial pressure; HR, heart rate; ETCO2, end tidal carbon dioxide

concentration; ETSEVO, end tidal sevoflurane concentration; BIS, bispectral index

value; and Paw, airway pressure.

Page 231: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

207

7.2.4. Intervention

A prolonged decrease in mean arterial pressure below 60 mmHg was treated

with metaraminol (sympathomimetic amine) 500 µg intravenous bolus, and/or

atropine (muscarinic antagonist) 300 µg intravenous bolus injection if

bradycardia (heart rate below 45 beats/min) was present. A sustained mean

blood pressure greater than 110 mmHg was treated by initially decreasing the

infusion and if necessary, treating with an intravenous glyceryl trinitrate (25 µg

bolus). A further criterion for intervention was a significant decrease in ScO2

(more than 25% decrease below the baseline, or absolute value below 50%).

7.2.5. Statistical analysis

Data are presented as numbers or mean±SEM. Mean arterial blood pressure and

heart rate measurements were taken after the induction of anaesthesia were

used as baseline recordings. The effects of treatments on mean arterial

pressure and heart rate were expressed as the difference between measured

values and the post-induction baseline ( baseline). For cerebral oxygen

saturation (ScO2) data are expressed as percentage change (ScO2%) relative to

the values before induction of anaesthesia (basal) in order to demonstrate the

effects of anaesthesia on cerebral oxygen saturation compared to spontaneous

breathing before anaesthesia. Data analysis was performed using GraphPad

Prism 5.0 (GraphPad software, San Diego, CA, USA). Statistical comparison

between groups was performed using unpaired t test or one-way ANOVA

followed by Tukey’s post-test for intergroup comparison. Dunnett’s post-test

was used whenever treatment groups were compared to the saline group.

P<0.05 was considered significant in all cases.

Page 232: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

208

Results

7.3.1. Patient characteristics and intra-operative parameters

The patients involved in this study were admitted for a right or left shoulder

arthroscopic repair surgery. The patient characteristics are shown in Table 7-1.

There was no significant difference in the age, body weight, body mass index

(BMI) between the groups taking saline, noradrenaline or vasopressin (P>0.05).

Similar doses of propofol were used to induce general anaesthesia in all groups.

The end tidal carbon dioxide concentration (ETCO2), end tidal sevoflurane

concentration (ETSEVO), bispectral index value (BIS), airway pressure (Paw) and

body temperature were maintained constant throughout the experiment and no

significant difference was found between the control (saline) and treatment

groups for each of these variables (P>0.05) (Table 7-2).

Page 233: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

209

Figure 7-3: A representative measurement of haemodynamic and

anaesthesia parameters recorded in a study patient.

A representative monitor showing haemodynamic and anaesthesia parameters

including ECG trace, heart rate, blood pressure, % oxygen administered and end tidal

sevoflurane and carbon dioxide concentrations.

Table 7-1: Characteristics of the patients randomly assigned to saline,

noradrenaline or vasopressin infusion groups

Parameter Saline Noradrenaline Vasopressin

Male/female (n) 6/3 5/2 6/2

Age (years) 45 (19-69) 39 (22-62) 34 (20-47)

Weight (kg) 78.0±3.7 80.1±4.6 77.1±2.2

BMI (kg/m2) 25.0±0.8 25.5±0.9 24.6±0.6

Values are mean±SEM. Age mean (range). BMI, body mass index (body

weight/height2; n, number of patients in each group.

Page 234: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

210

Table 7-2: Intraoperative details of the patients assigned to saline,

noradrenaline and vasopressin groups

Parameter Saline (n=9)

Noradrenaline (n=7)

Vasopressin (n=8)

P values

Propofol (mg) 173±12 171±16 181±7.1 0.82

ETSEVO (%) 1.6±0.03 1.6±0.04 1.6±0.04 0.51

ETCO2 (mmHg) 35.8±0.2 36.0±0.4 36.0±0.3 0.38

BIS 36.7±1.5 39.9±2.9 37.2±1.8 0.37

T (oC) 35.6±0.1 36.0±0.1 35.8±0.1 0.07

Paw (cmH2O) 16.8±0.8 16.5±0.6 15.0±0.4 0.14

Values are mean±SEM. ETCO2, end tidal carbon dioxide concentration; ETSEVO, end

tidal sevoflurane concentration; BIS, bispectral index value; T, body temperature;

and Paw, airway pressure. P, comparison between all groups, one-way ANOVA.

7.3.2. Effects of vasopressor agents on mean arterial blood

pressure and heart rate

There was no significant difference in the mean arterial pressure before

induction of general anaesthesia between the groups receiving saline (104±9),

noradrenaline (99±5) or vasopressin infusion (96±4 mmHg) (P>0.05). Induction

of general anaesthesia considerably decreased the blood pressure (by 40 mmHg

in saline group) without significant effect on heart rate. Following induction of

anaesthesia, mean arterial pressure and heart rate stabilised with similar values

in each group (Table 7-3). After stabilization, infusion of saline caused no

change in heart rate and mean arterial pressure in the supine position compared

to the baseline (Figure 7-4A). Infusion of noradrenaline (0.003-0.3 µg/kg/min)

significantly increased mean arterial pressure to about 109 mmHg (P<0.05),

along with a significant decrease in heart rate in the supine position (P<0.05),

as shown in Figure 7-5A. Infusion of vasopressin 0.4-4 milliunits/kg/min (3-28.5

ng/kg/min) did not cause significant changes either in blood pressure or heart

rate (Figure 7-6A).

Page 235: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

211

The mean arterial pressure of the saline group significantly decreased as soon

as the patients were moved from the supine into the beach chair position

(P<0.05), requiring an intervention with an injection of metaraminol (500 µg),

but the heart rate was not immediately changed. The mean arterial pressure

gradually recovered after intervention to the values in the supine position. In

the noradrenaline treatment group, the mean arterial pressure was slightly but

insignificantly decreased by moving the patients to the upright position

(P>0.05). Noradrenaline maintained the post-beach chair mean arterial

pressure with similar values to those of pre-anaesthetic, but significantly

greater than the mean arterial pressure in the corresponding saline-treated

group (P>0.05) (Figure 7-5A). There was a further decrease in mean arterial

pressure without immediate change in heart rate in the vasopressin treatment

group after moving the patients into the beach chair position. The decrease in

blood pressure following the change to the beach chair position in patients

taking vasopressin was gradually reversed after intervention with metaraminol

injection (Figure 7-6A).

Page 236: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

212

Table 7-3: Summary of MAP, heart rate and ScO2 before and after

anaesthesia in the supine and beach chair positions.

Groups

Saline

(n=9)

Noradrenaline

(n=7)

Vasopressin

(n=8)

Basal (Air)

MAP (mmHg) 104±3.0 99±1.9 96±1.4

HR (bpm) 74±4.3 69±3.8 76±5.7

ScO2 AIR (%) 74±2.0 72±0.8 76±1.8

Baseline (BL) (anaesthesia)

MAP (mmHg) 64±2.3 70±3.8 69±2.1

HR (bpm) 74±3.7 66±2.6 75±4.6

ScO2 BL (%) 78±3.0 79±1.1 82±1.8

Post-infusion (INF)

MAP BL-INF (mmHg) -2±1.3 39±8.3* 1±2.1

HR BL-INF (bpm) -8±1.3 -17±3.0* -11±2.5

ScO2 BL-INF (%) -2±0.7 -6±1.1 -9±1.1

Beach chair position (BCP)

MAPBL-BCP (mmHg) -16±2.7 29±5.3* -17±2.8

HRBL-BCP (bpm) -6±3.7 -2±8.3 2±1.8

ScO2 BL-BCP (%) -6±1.3 -2±1.1 -11±1.8*

Intervention yesa yesb yesa

Data are expressed as mean±SEM. MAP, mean arterial pressure; HR, heart rate;

ScO2, cerebral oxygen saturation; and , change from baseline (BL) to post-

infusion (INF) or “beach chair” position (BCP). aAll patients received metaraminol

500 µg injection in BCP. bInfusion rate was decreased in 4 patients in the BCP.

*P<0.05 compared to saline, one-way ANOVA followed by Dunnett’s post hoc test.

Page 237: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

213

7.3.3. Effects of vasopressor agents on cerebral oxygen

saturation

The cerebral oxygen saturation (ScO2) was continuously monitored in the

control (saline), noradrenaline and vasopressin treatment groups both in supine

and beach chair positions. The ScO2 values obtained from the two optodes

placed on the right and left sides of the forehead overlying the prefrontal cortex

were averaged and compared with the pre-anaesthetic values. There was no

significant difference in the post-induction baseline ScO2 values between the

control and treatment groups (P>0.05) (Table 7-3). The ScO2 was stable in the

group receiving saline infusion while the patients were in the supine position,

but it was significantly decreased (ScO2 -6%, P<0.05) by placing the patient

into the beach chair position (Figure 7-4B). The decrease in ScO2 observed in

the saline group following beach chair positioning was gradually reversed after

intervention with metaraminol injection. In patients receiving noradrenaline

infusion, the ScO2 showed a slight but statistically insignificant decrease when

the patient was placed in the supine position. However, there was no further

desaturation upon placing the patients into the beach chair position, as shown

in Figure 7-5B. The post-beach chair ScO2 level, which was similar to the post-

anaesthetic baseline value, was maintained stable for the entire period of

surgery without requiring intervention. In the patient group treated with

vasopressin infusion, the ScO2 in the supine position demonstrated a progressive

decrease (Figure 7-6B). There was an additional decrease in the ScO2 after the

patients were seated into the beach chair position. The reduction in the ScO2

following beach chair positioning was gradually reversed with metaraminol

intervention.

Page 238: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

214

Figure 7-4: Effects of saline infusion on MAP and HR and ScO2 in

anaesthetised patients in supine and beach chair

positions.

The effects of saline infusion on mean arterial pressure (MAP; A) and heart rate

(HR; A) and cerebral oxygen saturation (ScO2; B) in anaesthetized human subjects

in supine and beach chair positions. After post-anaesthetic stabilization, the

normal saline solution was continuously infused. The infusion rate was increased

over 15 min (2.5 min allowed for each rate). The patient was then placed into the

beach chair position (BCP) and the infusion continued at the maximum rate

(shown by the horizontal arrow). Measurements for ScO2, MAP and HR were taken

before the infusion was started and after every dose was administered. Data are

expressed as changes compared to the post-induction baseline (BL) values before

treatment for MAP and HR ( baseline) and to the basal values (before induction)

for ScO2 ( ScO2 %). *P<0.05 compared to BL, one-way ANOVA followed by

Dunnett’s post hoc test. Number of subjects in the group =9.

Page 239: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

215

Figure 7-5: Effects of noradrenaline on MAP, HR and ScO2 in supine

and beach chair positions during anaesthesia.

The effects of noradrenaline infusion on mean arterial pressure (MAP; A) and

heart rate (HR; A) and cerebral oxygen saturation (ScO2; B) in anaesthetized

human subjects in supine and beach chair positions. After post-anaesthetic

stabilization, noradrenaline was continuously infused. The infusion rate was

increased over 15 min (2.5 min allowed for each rate). The patient was then

placed into the beach chair position (BCP) and the infusion continued at the

maximum rate (shown by the horizontal arrow). Measurements for ScO2, MAP and

HR were taken before the infusion was started and after every dose was

administered. Data are expressed as changes compared to the post-induction

baseline (BL) values before treatment for MAP and HR ( baseline) and to the

basal values (before induction) for ScO2 ( ScO2 %). Vertical bars are ±SEM.

Number of subjects in the group =7.

Page 240: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

216

Figure 7-6: Effects of vasopressin on MAP, HR and ScO2 in supine and

beach chair positions during anaesthesia.

The effects of vasopressin infusion on mean arterial pressure (MAP; A) and heart

rate (HR; A) and cerebral oxygen saturation (ScO2; B) in anaesthetized human

subjects in supine and beach chair positions. After post-anaesthetic stabilization,

vasopressin was continuously infused. The infusion rate was increased over 15 min

(2.5 min allowed for each rate). The patient was then placed into the beach chair

position (BCP) and the infusion continued at the maximum rate (shown by the

horizontal arrow). Measurements for ScO2, MAP and HR were taken before the

infusion was started and after every dose was administered. Data are expressed

as changes compared to the post-induction baseline (BL) values before treatment

for MAP and HR ( baseline) and to the basal values (before induction) for ScO2

( ScO2 %). Vertical bars are ±SEM. *P<0.05 compared to BL, one-way ANOVA

followed by Dunnett’s post hoc test. Number of subjects in the group =8.

Page 241: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

217

Discussion

This study has compared the effects of noradrenaline and vasopressin on blood

pressure and cerebral oxygen saturation in anaesthetised patients in supine and

beach chair positions. Hypotension was present in both positions, with

occurrence of cerebral desaturation upon placement of the patients in the

beach chair position. Continuous administration of noradrenaline increased the

blood pressure and prevented incidence of cerebral oxygen desaturation during

anaesthesia both in supine and beach chair positions, unlike vasopressin which

had minimal effects on blood pressure but led to further cerebral oxygen

desaturation. The findings from the current study suggest that vasopressor

therapy can maintain cerebral perfusion pressure and thus prevent occurrence

of cerebral oxygen desaturation during beach chair anaesthesia, but individual

agents may induce differential effects on the cerebral circulation causing either

beneficial or harmful outcomes.

General anaesthesia in the beach chair position has been known to be a

precarious practice because it is associated with significant hypotension and

decreased blood supply to the brain. A real-time monitoring of cerebral blood

flow along with blood pressure helps to detect episodes of cerebral ischaemia

associated with intraoperative hypotension. Cerebral oximetry is a rather direct

method of monitoring cerebral oxygenation and helps to detect any episode of

cerebral ischaemia. In the current study, NIRS oximetry technique was used to

continuously monitor regional cerebral oxygen saturation in anaesthetised

patients in supine and beach chair positions. Induction of anaesthesia

significantly decreased the patients’ mean arterial pressure from pre-

anaesthetic values of about 100 to under 70 mmHg (30%). Placing the patient

into the beach chair position further decreased the blood pressure by about 20

mmHg. Similar findings have previously been reported where positioning

anaesthetised patients in the upright position significantly decreased the mean

arterial pressure, but the blood pressure was not affected by placing sedated

patients (control) in the beach chair position (Soeding et al., 2011). Excessive

decrease in arterial blood pressure during beach chair anaesthesia compromises

Page 242: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

218

the cerebral perfusion pressure posing a risk of ischaemic brain damage in the

patients. Despite decreases in mean arterial pressure in anaesthetised patients

in the supine position, cerebral oxygen saturation was not significantly

decreased which may be due to the existence of cerebral autoregulation or

decreased oxygen extraction as a result of anaesthesia-induced decrease in

neural metabolism (Meng et al., 2013). It has been indicated that most

anaesthetic agents in clinical use, including sevoflurane, do not interfere with

the cerebral autoregulation (Summors et al., 1999). Thus, intraoperative

hypotension in supine position is as such may not have a great risk of ischaemic

brain damage. However, placing anaesthetised patients into the beach chair

position induces a further decrease in arterial blood pressure associated with a

significant cerebral oxygen desaturation. Severe and prolonged cerebral oxygen

desaturation or hypoxia can lead to ischaemic brain damage. Thus, severe

decreases in blood pressure should be prevented to maintain cerebral perfusion

while anaesthetised patients are seated in upright position, similar to the

recommended management of traumatic brain injury (Bratton et al., 2007).

Maintaining blood pressure with prophylactic administration of a vasopressor

agent can prevent the occurrence of cerebral oxygen desaturation during beach

chair anaesthesia. The right choice of a vasopressor agent may be of paramount

importance to obtain a favourable therapeutic outcome. Here, the ‘suitability’

of two vasopressor agents frequently used in neurocritical care, noradrenaline

and vasopressin, for prevention of cerebral oxygen desaturation during the

beach chair anaesthesia was investigated in patients. Infusion of noradrenaline

increased the blood pressure which was maintained both in the supine and

beach chair positions. Noradrenaline prevented cerebral oxygen desaturation

induced by placing the patient in the beach chair position.

The effects of noradrenaline on cerebral blood flow may be a consequence of

its effects on peripheral blood pressure. The direct effects of systemically

administered noradrenaline on cerebral arteries and arterioles is limited due to

the presence of blood-brain barrier in these vessels (MacKenzie et al., 1976).

In the peripheral circulation, noradrenaline increases arterial blood pressure by

Page 243: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

219

multiple mechanisms owing to its effects on both α- and β-adrenoceptors.

Activation of α1-adrenoceptors located on arteries causes constriction that

leads to increased peripheral resistance, while α1-adrenoceptor-mediated

constriction of the veins increases the venous return and improves cardiac

preload. Additionally, noradrenaline stimulates the heart by activating cardiac

β-adrenoceptors increases cardiac output. Overall, noradrenaline increases the

arterial blood pressure and maintains cerebral perfusion. Thus, continuous

infusion of noradrenaline has beneficial effects in preventing severe

hypotension and cerebral oxygen desaturation induced by seating the patient

in the upright beach chair position commonly used in shoulder surgery.

Noradrenaline had a tendency, though statistically insignificant, to decrease

ScO2 in supine position. Other studies have found similar decreases in cerebral

oxygen saturation after noradrenaline administration in healthy subjects

(Brassard et al., 2009; Ogoh et al., 2011). Another study reported that

noradrenaline increased mean arterial pressure in humans without affecting the

middle cerebral blood flow velocity, a surrogate measure of cerebral blood flow

(Moppett et al., 2008). The likelihood that noradrenaline may cause

constriction of the cerebral microcirculation is very low because it cannot

access the cerebrovascular smooth muscle unless the blood-brain barrier is

disrupted (Moller et al., 2004). Further, the constriction effect of noradrenaline

in human isolated cerebral arteries is very weak (Toda and Fujita, 1973). The

possible reason for the discrepancy between apparent cerebral desaturation

and unchanged cerebral blood flow observed after noradrenaline administration

may be related to interferences with the cerebral oximetry, such as constriction

of extracerebral arteries. Recent studies have indicated that constriction of

cutaneous arteries induced by noradrenaline influences the NIRS measurement

of cerebral oxygen saturation (Sorensen et al., 2012; Ogoh et al., 2014).

Infusion of vasopressin did not increase the blood pressure in the supine

position. Neither did vasopressin significantly affect the heart rate in

anaesthetised subjects. However, it decreased cerebral oxygen saturation in

the supine position. Vasopressin did not prevent a further decrease in blood

Page 244: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

220

pressure induced by placing the patient in the beach chair position. The

decrease in ScO2 was sustained in the beach chair position. This shows that

systemically infused vasopressin may cause direct vasoconstrictor effects in

human cerebral arteries. Similar results were recently reported wherein

vasopressin (0.07 unit/kg bolus) increased the mean arterial pressure by 26

mmHg (31%) without altering heart rate in supine position, which was reversed

in the beach chair position, and induced cerebral oxygen desaturation. The

cerebral desaturation induced in the beach chair position was more pronounced

in patients treated with vasopressin than in untreated (control) patients (Cho

et al., 2013).

The lack of effect on mean arterial pressure found in this study upon infusion

of vasopressin may be because relatively lower doses were used. Higher doses

of vasopressin were not considered for fear that certain serious cardiac adverse

effects such as myocardial ischaemia may develop (Maturi et al., 1991). Similar

findings were previously reported where a stepwise infusion of vasopressin at

0.2-5 pmol/kg/min (0.2-5 ng/kg/min) did not affect blood pressure in healthy

subjects, but induced a significant pressor response in patients with autonomic

insufficiency (Williams et al., 1986). Vasopressin (10-20 units bolus followed by

0.1 units/min) is recommended for the management of intraoperative

hypotension (Treschan and Peters, 2006), but its benefit in preserving cerebral

perfusion is uncertain. It appears that low doses of vasopressin have minimal

effects on mean arterial pressure in healthy subjects but may increase blood

pressure in hypotensive conditions that are associated with impaired

sympathetic nerve activity and low plasma vasopressin levels such as in septic

shock (Montani et al., 1980; Landry et al., 1997; Holmes et al., 2001; Dunser

et al., 2003; Holmes et al., 2004).

The prolonged decrease in ScO2 without significant change of mean arterial

pressure observed in patients receiving vasopressin infusion, however, is

interesting. A study in pig models of cardiopulmonary resuscitation found that

administration of vasopressin induced a sustained decrease in cerebral

oxygenation (Bein et al., 2005). Vasopressin may have caused selective

Page 245: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

221

constriction of the cerebral microcirculation without significantly affecting the

tone of peripheral resistance arteries. Peptides injected into the peripheral

circulation may cross the blood-brain barrier passively or via specific

transporters located on the endothelial cells (Banks, 2015). Whether significant

levels of vasopressin cross an intact blood-brain barrier and accumulate on the

interstitial side to cause constriction of cerebral arteries in healthy subjects is

not clearly known. An in situ brain perfusion study in pigs showed influx of

vasopressin into the brain indicating the existence of a carrier-mediated

transport located on the luminal side of the blood-brain barrier in cerebral

microcirculation (Zlokovic et al., 1990). However, the effects of vasopressin on

permeability of cerebral arteries and microcirculation have not been studies in

other species including humans.

Both vasodilator and vasoconstrictor effects of vasopressin have been

previously reported in cerebral arteries obtained from various species (Katusic

et al., 1984; Lluch et al., 1984; Martin de Aguilera et al., 1990; Suzuki et al.,

1993). The complexity of the cerebrovascular response to vasopressin has been

demonstrated by Takayasu et al. (1993) who showed that vasopressin induces a

triphasic response in rat isolated cerebral arterioles. Low concentrations of

vasopressin (10-12-10-11 M) dilate, but moderately higher concentrations of

vasopressin (10-10-10-8 M) constrict the arteries. A further increase in

vasopressin concentration (>10-8 M) completely dilated the arterioles. Systemic

administration of vasopressin can possibly cause constriction of cerebral

arteries owing to its carrier-mediated transportation across the blood-brain

barrier. Thus, the benefit of using vasopressin in the management of

hypotension induced by general anaesthesia is questionable. Rather,

vasopressin may increase the risk of cerebral desaturation in hypotensive

states.

In conclusion, maintaining arterial blood pressure during beach chair surgery

can minimize the risk of cerebral desaturation, however the choice of

vasopressor agent should be considered carefully. Noradrenaline can effectively

Page 246: Cerebrovascular effects of vasoactive drugs

Vasopressor agents and cerebral oxygenation

222

prevent cerebral desaturation but vasopressin has no benefit, or even increase

the risk of desaturation during general anaesthesia in the beach chair position.

Page 247: Cerebrovascular effects of vasoactive drugs

223

Summary and conclusions

Page 248: Cerebrovascular effects of vasoactive drugs

Summary and conclusions

224

The brain has a unique blood flow that is specially regulated in order to

maintain oxygen and nutrient supply over a range of peripheral blood pressure

(50-150 mmHg). This is achieved by networks of superficial and penetrating

arteries and arterioles that are controlled by the peripheral nerves, as well as

neural inputs from cortical and subcortical areas. The endothelial lining of the

blood vessels in the brain also plays an important role in regulating the tone of

cerebral arteries by releasing vasoconstrictor and vasodilator mediators.

Additionally, the vasogenic responses, pressure- or stretch-induced

constriction, of cerebral arteries, may also play important roles in the

regulation of cerebral blood flow. This study characterised the cerebrovascular

responses to in vitro pharmacological stimulation in rat isolated arteries, in

vivo effects of systemically administered vasoactive agents in rat models, as

well as effects of vasopressor administration on cerebral oxygen saturation

during anaesthesia in patients.

Rat cerebral arteries demonstrated variable responses to vasoactive agents

depending on their regional location in the rat cerebral circulation. Arteries

isolated from the anterior parts of the rat cerebral circulation were highly

sensitive to the constrictor mediators such as noradrenaline, vasopressin and

serotonin unlike arteries of the posterior cerebral circulation, which did not

respond or were only weakly constricted by many vasoconstrictor agents. On

the contrary, vasodilator agents relaxed arteries isolated from the posterior

cerebral circulation with greater potency than those isolated from anterior

regions of the rat cerebral circulation. The variations in the pharmacological

responses of cerebral arteries may be related to regional heterogeneity in the

distribution of the receptors mediating vasoconstriction and vasodilatation.

This study characterised the mechanisms involved in adrenoceptor-mediated

vascular responses in rat cerebral arteries. Stimulation of α1-adrenoceptors

induced a contractile response in arteries of the anterior rat cerebral

circulation, namely the anterior and middle cerebral arteries, but not in

posterior communicating and basilar arteries. However, the β-adrenoceptor-

mediated relaxant responses were found in all arteries taken from different

Page 249: Cerebrovascular effects of vasoactive drugs

Summary and conclusions

225

regions of the rat cerebral circulation. The relaxation response is mediated by

both β1- and β2-adrenoceptors that are distributed differentially in smooth

muscle or endothelial cells depending on the location of the arteries. β-

adrenoceptors are located on smooth muscle of the middle cerebral artery,

while they are located on the endothelium of the basilar artery. Activation of

the endothelial β-adrenoceptors in the rat basilar artery induces release either

NO or EDH depending on the type of agonist used. Noradrenaline primarily

induces release of NO, but it may also release a small proportion of EDH.

Isoprenaline, however, favours the release of EDH along with a small proportion

of NO. These variations may be related to differences in the intrinsic activity

of the agonists on adrenoceptors.

Some of the vasoactive mediators including endothelin-1 and noradrenaline

have been implicated in the pathogenesis of cerebrovascular diseases such as

ischaemia or vasospasm that often occurs following subarachnoid haemorrhage.

In light of this, the link between endothelin receptor stimulation and activation

of different calcium channels was analysed in this study. The potent contraction

of rat cerebral arteries induced by endothelin-1 was found to be largely

dependent on influx of Ca2+ from the extracellular space. Endothelin-1 activates

both voltage-operated and non-voltage operated calcium channels to induce

contraction of cerebral arteries. The non-voltage-operated calcium channels

play an important role in induction of the contraction by low endothelin-1

concentrations. T-type (low-voltage-operated) calcium channels are also

activated by low concentrations of endothelin-1 during the early phase of

contraction, but L-type (high-voltage-operated) calcium channels are activated

by higher concentrations of endothelin-1 and are involved in the maintenance

phase of the contraction.

The effects of systemically administered vasoactive agents on the diameter of

cerebral arteries were investigated in vivo in the rat using the cranial window

technique to visualize the cerebral arteries. Phenylephrine (an α1-adrenoceptor

vasoconstrictor agonist) and vasopressin (a vasoconstrictor peptide) increased

arterial blood pressure with simultaneous constriction of the cerebral arteries,

Page 250: Cerebrovascular effects of vasoactive drugs

Summary and conclusions

226

whereas formoterol (a β2-adrenoceptor agonist vasodilator) decreased blood

pressure and dilated the cerebral arteries. The extent and time-course of the

blood pressure and cerebral arterial responses were similar when the drugs

were systemically administered either through the femoral vein, or directly into

the cerebral circulation via the carotid artery. Thus, the cerebrovascular

effects of systemic vasoactive drug administration are indirect autoregulatory

responses induced by changes in the arterial pressure. Nonetheless,

phenylephrine and vasopressin may also elicit negligible effects on cerebral

arteries. Under normal conditions, the blood-brain barrier limits most blood-

borne substances from accessing the vascular smooth muscle, but its

permeability may be affected when the blood pressure is elevated.

Finally, the clinical benefit of vasopressor therapy in protecting cerebral

oxygen desaturation during anaesthesia in the ‘beach chair’ position was

investigated in patients. Excessive hypotension associated with placing an

anaesthetised patient in the beach chair position poses a risk of cerebral oxygen

desaturation. Noradrenaline prevented cerebral oxygen desaturation by

maintaining arterial pressure in the beach chair position. However, vasopressin

did not prevent hypotension during anaesthesia in the beach chair position, but

exacerbated the cerebral oxygen desaturation. A further cerebral oxygen

desaturation may result from the direct constrictor effect of vasopressin on the

cerebral microcirculation. The findings demonstrate that some vasopressor

agents may have deleterious effects on cerebral blood flow possibly leading to

ischaemic brain damage. Thus, the effects of vasopressor drugs on the cerebral

circulation should be rigorously investigated before they are applied to provide

haemodynamic support under conditions where the cerebral blood flow is

compromised.

In summary, this study found variable effects of vasoactive drugs in cerebral

arteries and demonstrated the need to understand the mechanisms behind the

effects induced by individual drugs. Future studies are required to test more

drugs and investigate their effects under different cerebrovascular diseases.

This will help to identify the systems affected by pathological conditions, as

Page 251: Cerebrovascular effects of vasoactive drugs

Summary and conclusions

227

well as to design rational therapy for effective treatment of cerebral blood flow

disorders including stroke.

Page 252: Cerebrovascular effects of vasoactive drugs
Page 253: Cerebrovascular effects of vasoactive drugs

229

References

Page 254: Cerebrovascular effects of vasoactive drugs

References

230

Abbott NJ, Ronnback L, Hansson E (2006). Astrocyte-endothelial interactions at

the blood-brain barrier. Nat Rev Neurosci 7: 41-53.

Abd El-Rahman RR, Harraz OF, Brett SE, Anfinogenova Y, Mufti RE, Goldman D,

et al. (2013). Identification of L- and T-type Ca2+ channels in rat cerebral

arteries: role in myogenic tone development. Am J Physiol Heart Circ Physiol

304: H58-71.

Abott NJ, Bundgaard M, Hughes CCW (1990). Cerebral microcirculation Progress

in applied microcirculation. Hammersen F, Messmer K (ed). Volume 16. Karger.

New York.

Agnoli A, Fieschi C, Bozzao L, Battistini N, Prencipe M (1968). Autoregulation of

cerebral blood flow. Studies during drug-induced hypertension in normal

subjects and in patients with cerebral vascular diseases. Circulation 38: 800-

812.

Aguilera P, Chanez-Cardenas ME, Floriano-Sanchez E, Barrera D, Santamaria A,

Sanchez-Gonzalez DJ, et al. (2007). Time-related changes in constitutive and

inducible nitric oxide synthases in the rat striatum in a model of Huntington's

disease. Neurotoxicology 28: 1200-1207.

Al-Tamimi YZ, Orsi NM, Quinn AC, Homer-Vanniasinkam S, Ross SA (2010). Peer-

Review Report: a review of delayed ischemic neurologic deficit following

aneurysmal subarachnoid hemorrhage: historical overview, current treatment,

and pathophysiology. World Neurosurg 73: 654-667.

Alborch E, Salom JB, Torregrosa G (1995). Calcium channels in cerebral

arteries. Pharmacol Ther 68: 1-34.

Alexander RW, Brock TA, Gimbrone MA, Jr., Rittenhouse SE (1985). Angiotensin

increases inositol trisphosphate and calcium in vascular smooth muscle.

Hypertension 7: 447-451.

Page 255: Cerebrovascular effects of vasoactive drugs

References

231

Ando K, Arai S, Kawamura K (1990). Comparative study on the innervation of

nerves with calcitonin gene-related peptide, substance P and neurokinin A

immunoreactivity in the walls of the cerebral arteries of small bats (Mammalia:

Microchiroptera). Neuroscience 36: 521-533.

Ando K, Ishikawa A, Okura N (1994). Nature and origin of cerebrovascular nerves

with substance P immunoreactivity in bats (Mammalia: Microchiroptera), with

special reference to species differences. J Comp Neurol 347: 357-368.

Andreas S, Daniela F, Lennart W, Johann B (2010). Guanylyl cyclase activator

ataciguat improves vascular function and reduces platelet activation in heart

failure. Pharmacol Res 62: 432-438.

Angus JA, Wright CE (2000). Techniques to study the pharmacodynamics of

isolated large and small blood vessels. J Pharmacol Toxicol Methods 44: 395-

407.

Ansar S, Vikman P, Nielsen M, Edvinsson L (2007). Cerebrovascular ETB, 5-HT1B,

and AT1 receptor upregulation correlates with reduction in regional CBF after

subarachnoid hemorrhage. Am J Physiol Heart Circ Physiol 293: H3750-H3758.

Arnold WP, Mittal CK, Katsuki S, Murad F (1977). Nitric oxide activates

guanylate cyclase and increases guanosine 3′-5′-cyclic monophosphate levels in

various tissue preparations. Proc Natl Acad Sci U S A 74: 3203-3207.

Asano M, Masuzawa-Ito K, Matsuda T, Suzuki Y, Oyama H, Shibuya M, et al.

(1993). Increased Ca2+ influx in the resting state maintains the myogenic tone

and activates charybdotoxin-sensitive K+ channels in dog basilar artery. J Cereb

Blood Flow Metab 13: 969-977.

Atochin DN, Huang PL (2011). Role of endothelial nitric oxide in cerebrovascular

regulation. Curr Pharm Biotechnol 12: 1334-1342.

Auer LM, Pucher R, Leber K, Ishiyama N (1987). Autoregulatory response of pial

vessels in the cat. Neurol Res 9: 245-248.

Page 256: Cerebrovascular effects of vasoactive drugs

References

232

Austin SA, Santhanam AV, Hinton DJ, Choi DS, Katusic ZS (2013). Endothelial

nitric oxide deficiency promotes Alzheimer's disease pathology. J Neurochem

127: 691-700.

Ayajiki K, Toda N (1990). Isolated bovine cerebral arteries from rostral and

caudal regions: distinct responses to adrenoceptor agonists. Eur J Pharmacol

191: 417-425.

Ayajiki K, Toda N (1992). Regional difference in the response mediated by beta

1-adrenoceptor subtype in bovine cerebral arteries. J Cereb Blood Flow Metab

12: 507-513.

Baeres FM, Moller M (2004). Origin of PACAP-immunoreactive nerve fibers

innervating the subarachnoidal blood vessels of the rat brain. J Cereb Blood

Flow Metab 24: 628-635.

Baker JG (2010). The selectivity of beta-adrenoceptor agonists at human beta1-

, beta2- and beta3-adrenoceptors. Br J Pharmacol 160: 1048-1061.

Ballabh P, Braun A, Nedergaard M (2004). The blood-brain barrier: an overview:

structure, regulation, and clinical implications. Neurobiol Dis 16: 1-13.

Banks WA (2015). Peptides and the blood-brain barrier. Peptides 2015/03/26

10.1016/j.peptides.2015.03.010.

Barbanti P, Egeo G, Aurilia C, Fofi L, Della-Morte D (2014). Drugs targeting nitric

oxide synthase for migraine treatment. Expert Opin Investig Drugs 23: 1141-

1148.

Barton M, Yanagisawa M (2008). Endothelin: 20 years from discovery to therapy.

Can J Physiol Pharm 86: 485-498.

Baumbach GL, Heistad DD (1985). Regional, segmental, and temporal

heterogeneity of cerebral vascular autoregulation. Ann Biomed Eng 13: 303-

310.

Page 257: Cerebrovascular effects of vasoactive drugs

References

233

Bayliss WM (1902). On the local reactions of the arterial wall to changes of

internal pressure. J Physiol 28: 220-231.

Bein B, Cavus E, Dorges V, Stadlbauer KH, Tonner PH, Steinfath M, et al. (2005).

Arginine vasopressin reduces cerebral oxygenation and cerebral blood volume

during intact circulation in swine - a near infrared spectroscopy study. Eur J

Anaesthesiol 22: 62-66.

Bell RM, Hannun YA, Loomis CR (1986). Mechanism of regulation of protein

kinase C by lipid second messengers. Symp Fundam Cancer Res 39: 145-156.

Bellien J, Joannides R, Iacob M, Arnaud P, Thuillez C (2006). Evidence for a

basal release of a cytochrome-related endothelium-derived hyperpolarizing

factor in the radial artery in humans. Am J Physiol Heart Circ Physiol 290:

H1347-1352.

Bentsen N, Larsen B, Lassen NA (1975). Chronically impaired autoregulation of

cerebral blood flow in long-term diabetics. Stroke 6: 497-502.

Bergsman JB, Wheeler DB, Tsien RW (2000). Classification and function of

voltage-gated calcium channels. Handbook Exp Pharmacol 147: 55-86.

Berridge MJ, Lipp P, Bootman MD (2000). The versatility and universality of

calcium signalling. Nat Rev Mol Cell Biol 1: 11-21.

Bevan J, Osher J (1972). A direct method for recording tension changes in the

wall of small blood vessels in vitro. Agents & Actions 2: 257.

Biondi A, Ricciardi GK, Puybasset L, Abdennour L, Longo M, Chiras J, et al.

(2004). Intra-arterial nimodipine for the treatment of symptomatic cerebral

vasospasm after aneurysmal subarachnoid hemorrhage: preliminary results.

AJNR Am J Neuroradiol 25: 1067-1076.

Bohr D, Goulet P, Taquini A (1961). Direct tension recording from smooth

muscle of resistance vessels from various organs. Angiology 12: 478.

Page 258: Cerebrovascular effects of vasoactive drugs

References

234

Bootman MD, Collins TJ, Peppiatt CM, Prothero LS, MacKenzie L, De Smet P, et

al. (2001). Calcium signalling - an overview. Semin Cell Dev Biol 12: 3-10.

Brassard P, Seifert T, Secher NH (2009). Is cerebral oxygenation negatively

affected by infusion of norepinephrine in healthy subjects? Br J Anaesth 102:

800-805.

Bratton SL, Chestnut RM, Ghajar J, McConnell Hammond FF, Harris OA, Hartl R,

et al. (2007). Guidelines for the management of severe traumatic brain injury.

XV. Steroids. J Neurotrauma 24 Suppl 1: S91-95.

Brian JE, Jr. (1998). Carbon dioxide and the cerebral circulation.

Anesthesiology 88: 1365-1386.

Brown MM, Pickles H (1982). Effect of epoprostenol (prostacyclin, PGI2) on

cerebral blood flow in man. J Neurol Neurosurg Psychiatry 45: 1033-1036.

Bryan NS, Bian K, Murad F (2009). Discovery of the nitric oxide signaling pathway

and targets for drug development. Front Biosci 14: 1-18.

Budohoski KP, Czosnyka M, Kirkpatrick PJ, Smielewski P, Steiner LA, Pickard JD

(2013). Clinical relevance of cerebral autoregulation following subarachnoid

haemorrhage. Nat Rev Neurol 9: 152-163.

Buhre W, Weyland A, Buhre K, Kazmaier S, Mursch K, Schmidt M, et al. (2000).

Effects of the sitting position on the distribution of blood volume in patients

undergoing neurosurgical procedures. Br J Anaesth 84: 354-357.

Bulley S, Neeb Z, Burris S, Bannister J, Thomas-Gatewood C, Jangsangthong W,

et al. (2012). TMEM16A/ANO1 channels contribute to the myogenic response in

cerebral arteries. Circ Res 111: 1027-1045.

Busija DW (1993).Cerebral autoregulation. Phillis JW (ed). ed. CRC Press. Boca

Raton, Florida. pp: 45-65.

Page 259: Cerebrovascular effects of vasoactive drugs

References

235

Buus NH, VanBavel E, Mulvany MJ (1994). Differences in sensitivity of rat

mesenteric small arteries to agonists when studied as ring preparations or as

cannulated preparations. Br J Pharmacol 112: 579-587.

Cain AE, Tanner DM, Khalil RA (2002). Endothelin-1--induced enhancement of

coronary smooth muscle contraction via MAPK-dependent and MAPK-

independent [Ca(2+)](i) sensitization pathways. Hypertension 39: 543-549.

Campbell WB, Gebremedhin D, Pratt PF, Harder DR (1996). Identification of

epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ

Res 78: 415-423.

Capra N, Kapp J (1987). Anatomical and physiological aspects of venous systems

In: Wood J (ed). ed. Cerebral blood flow: Physiology and clinical aspect.

McGraw-Hill. NY. pp:37-58.

Carey RM, Wang ZQ, Siragy HM (2000). Update: role of the angiotensin type-2

(AT(2)) receptor in blood pressure regulation. Curr Hypertens Rep 2: 198-201.

Carnevale D, Lembo G (2010). Hypertension and cerebrovascular dysfunction:

acute and chronic brain pathological alterations. Wolters Kluwer Health. pp:

191.

Castanares-Zapatero D, Hantson P (2011). Pharmacological treatment of

delayed cerebral ischemia and vasospasm in subarachnoid hemorrhage. Ann

Intensive Care 1: 12.

Catterall WA (2000). Structure and regulation of voltage-gated Ca2+ channels.

Annu Rev Cell Biol 16: 521.

Catterall WA, Perez-Reyes E, Snutch TP, Striessnig J (2005). International Union

of Pharmacology. XLVIII. Nomenclature and structure-function relationships of

voltage-gated calcium channels. Pharmacol Rev 57: 411-425.

Page 260: Cerebrovascular effects of vasoactive drugs

References

236

Chang HH, Lee YC, Chen MF, Kuo JS, Lee TJ (2012). Sympathetic activation

increases basilar arterial blood flow in normotensive but not hypertensive rats.

Am J Physiol Heart Circ Physiol 302: H1123-1130.

Chen JJ, Salat DH, Rosas HD (2012). Complex relationships between cerebral

blood flow and brain atrophy in early Huntington's disease. Neuroimage: 1043.

Cho SY, Kim SJ, Jeong CW, Jeong CY, Chung SS, Lee J, et al. (2013). Under

general anesthesia arginine vasopressin prevents hypotension but impairs

cerebral oxygenation during arthroscopic shoulder surgery in the beach chair

position. Anesth Analg 117: 1436-1443.

Cipolla MJ (2009).The Cerebral Circulation. (ed). ed. Morgan & Claypool Life

Sciences. San Rafael, CA. pp: 1-43.

Cipolla MJ, Godfrey JA (2010). Effect of hyperglycemia on brain penetrating

arterioles and cerebral blood flow before and after ischemia/reperfusion.

Transl Stroke Res 1: 127-134.

Cipolla MJ, Lessov N, Hammer ES, Curry AB (2001). Threshold duration of

ischemia for myogenic tone in middle cerebral arteries: effect on vascular

smooth muscle actin. Stroke 32: 1658-1664.

Cipolla MJ, Smith J, Kohlmeyer MM, Godfrey JA (2009). SKCa and IKCa Channels,

myogenic tone, and vasodilator responses in middle cerebral arteries and

parenchymal arterioles: effect of ischemia and reperfusion. Stroke 40: 1451-

1457.

Cipolla MJ, Sweet J, Chan SL, Tavares MJ, Gokina N, Brayden JE (2014).

Increased pressure-induced tone in rat parenchymal arterioles vs. middle

cerebral arteries: role of ion channels and calcium sensitivity. J Appl Physiol

Journal of applied physiology 117: 53-59.

Cohen Z, Bonvento G, Lacombe P, Hamel E (1996). Serotonin in the regulation

of brain microcirculation. Prog Neurobiol 50: 335-362.

Page 261: Cerebrovascular effects of vasoactive drugs

References

237

Cohen Z, Molinatti G, Hamel E (1997). Astroglial and vascular interactions of

noradrenaline terminals in the rat cerebral cortex. J Cereb Blood Flow Metab

17: 894-904.

Cole DJ, Matsumura JS, Drummond JC, Schultz RL, Wong MH (1991). Time- and

pressure-dependent changes in blood-brain barrier permeability after

temporary middle cerebral artery occlusion in rats. Acta neuropathologica 82:

266-273.

Coney AM, Marshall JM (1998). Role of adenosine and its receptors in the

vasodilatation induced in the cerebral cortex of the rat by systemic hypoxia. J

Physiol 509 ( Pt 2): 507-518.

Cowen T, Alafaci C, Crockard HA, Burnstock G (1986). 5-HT-containing nerves

to major cerebral arteries of the gerbil originate in the superior cervical

ganglia. Brain Res 384: 51-59.

Coyle P (1994). Collateral pial arteries In: RD. B, Bevan J (ed). ed. The human

cerebral circulation:Functional changes in disease. Humana Press. Totowa, NJ.

pp:237-246.

Crowley RW, Medel R, Kassell NF, Dumont AS (2008). New insights into the

causes and therapy of cerebral vasospasm following subarachnoid hemorrhage.

Drug Discov Today 13: 254-260.

Cullen D, Kirby R (2007). Beach chair position may decrease cerebral perfusion:

Catastrophic outcomes have occurred. APSF Newsletter 22: 25-27.

Dagal A, Lam AM (2009). Cerebral autoregulation and anesthesia. Curr Opin

Anaesthesiol 22: 547-552.

Dauphin F, Hamel E (1990). Muscarinic receptor subtype mediating vasodilation

feline middle cerebral artery exhibits M3 pharmacology. Eur J Pharmacol 178:

203-213.

Page 262: Cerebrovascular effects of vasoactive drugs

References

238

Dauphin F, Linville DG, Hamel E (1994). Cholinergic dilatation and constriction

of feline cerebral blood vessels are mediated by stimulation of phosphoinositide

metabolism via two different muscarinic receptor subtypes. J Neurochem 63:

544-551.

Dauphin F, Ting V, Payette P, Dennis M, Hamel E (1991). Vasocontractile

muscarinic M1 receptors in cat cerebral arteries: pharmacological identification

and detection of mRNA. Eur J Pharmacol 207: 319-327.

Davenport AP (2002). International Union of Pharmacology. XXIX. Update on

endothelin receptor nomenclature. Pharmacol Rev 54: 219-226.

Davis MJ (2012). Perspective: Physiological Role(s) of the Vascular Myogenic

Response. Microcirculation 19: 99-114.

Davis MJ, Hill MA (1999). Signaling mechanisms underlying the vascular

myogenic response. Physiol Rev 79: 37.

Dawson DA, Sugano H, McCarron RM, Hallenbeck JM, Spatz M (1999). Endothelin

receptor antagonist preserves microvascular perfusion and reduces ischemic

brain damage following permanent focal ischemia. Neurochem Res 24: 1499-

1505.

Day A (1987). Arterial distributions and variants. In: Wood J (ed). ed. Cerebral

blood flow: Physiologic and clinical aspects. McGraw-Hill. NY. pp:19-55.

De Vasconcelos A, Baldwin R, Wasterlain C (1995). Nitric oxide mediates the

increase in local cerebral blood flow during focal seizures. Proc Natl Acad Sci U

S A. 92: 3175-3179.

De Vasconcelos A, Riban V, Wasterlain C, Nehlig A (2006). Role of endothelial

nitric oxide synthase in cerebral blood flow changes during kainate seizures: A

genetic approach using knockout mice. Neurobiol Dis: 219.

Page 263: Cerebrovascular effects of vasoactive drugs

References

239

Deckert V, Angus JA (1992). Evidence that 5-HT2 receptors predominantly

mediate the contraction of the rat basilar artery to 5-hydroxytryptamine. Eur

J Pharmacol 221: 17-25.

Delpy DT, Cope M (1997). Quantification in Tissue Near-Infrared Spectroscopy.

The Royal Society. pp: 649.

Domer FR, Sankar R, Cole S, Wellmeyer D (1980). Dose-dependent,

amphetamine-induced changes in permeability of the blood-brain barrier of

normotensive and spontaneously hypertensive rats. Exp Neurol 70: 576-585.

Douds M, Straub E, Kent A, Bistrick C, Sistino J (2014). A systematic review of

cerebral oxygenation-monitoring devices in cardiac surgery. Perfusion 29: 545-

552.

Doughty JM, Plane F, Langton PD (1999). Charybdotoxin and apamin block EDHF

in rat mesenteric artery if selectively applied to the endothelium. Am J Physiol

276: H1107-1112.

Drummond JC (1997). The lower limit of autoregulation: time to revise our

thinking? Anesthesiology 86: 1431-1433.

Duckles SP (1980). Functional activity of the noradrenergic innervation of large

cerebral arteries. Br J Pharmacol 69: 193-199.

Ducruet AF, Grobelny BT, Zacharia BE, Hickman ZL, Yeh ML, Connolly ES (2009).

Pharmacotherapy of cerebral ischemia. Expert Opin Pharmacother 10: 1895-

1906.

Dumont AS, Dumont RJ, Chow MM, Lin C-l, Calisaneller KF, Ley T, et al. (2003).

Cerebral vasospasm after subarachnoid hemorrhage: putative role of

inflammation. Neurosurgery 53: 123-134.

Dunatov S, Antoncic I, Bralic M (2011). Hemodynamic changes in the posterior

cerebral circulation triggered by insufficient sympathetic innervation--cause of

primary intracerebral hemorrhage? Med Hypotheses 76: 668-669.

Page 264: Cerebrovascular effects of vasoactive drugs

References

240

Dunser MW, Mayr AJ, Ulmer H, Knotzer H, Sumann G, Pajk W, et al. (2003).

Arginine vasopressin in advanced vasodilatory shock: a prospective,

randomized, controlled study. Circulation 107: 2313-2319.

Eames P, Blake M, Dawson S, Panerai R, Potter J (2002). Dynamic cerebral

autoregulation and beat to beat blood pressure control are impaired in acute

ischaemic stroke. J Neurol Neurosurg Psychiatry 72: 467-472.

Earley S, Brayden JE (2010). Transient receptor potential channels and vascular

function. Clin Sci (Lond) 119: 19-36.

Edvinsson L (1975). Neurogenic mechanisms in the cerebrovascular bed.

Autonomic nerves, amine receptors and their effects on cerebral blood flow.

Acta Physiol Scand Suppl 427: 1-35.

Edvinsson L, Elsås T, Suzuki N, Shimizu T, Lee TJ (2001). Origin and Co-

localization of nitric oxide synthase, CGRP, PACAP, and VIP in the cerebral

circulation of the rat. Microsc Res Tech 53: 221-228.

Edvinsson L, Falck B, Owman C (1977). Possibilities for a cholinergic action on

smooth musculature and on sympathetic axons in brain vessels mediated by

muscarinic and nicotinic receptors. J Pharmacol Exp Ther 200: 117-126.

Edvinsson L, Fredholm BB, Hamel E, Jansen I, Verrecchia C (1985). Perivascular

peptides relax cerebral arteries concomitant with stimulation of cyclic

adenosine monophosphate accumulation or release of an endothelium-derived

relaxing factor in the cat. Neurosci Lett 58: 213-217.

Edvinsson L, Hardebo JE, Owman C (1979). Effects of angiotensin II on cerebral

blood vessels. Acta Physiol Scand 105: 381-383.

Edvinsson L, Mackenzie E, McCulloch J (1993).Cerebral blood flow and

metabolism. (ed). 1st ed. Raven Press. New York. pp: 183-230.

Edvinsson L, MacKenzie ET (1976). Amine mechanisms in the cerebral

circulation. Pharmacol Rev 28: 275-348.

Page 265: Cerebrovascular effects of vasoactive drugs

References

241

Edvinsson L, Owman C (1974). Pharmacological characterization of adrenergic

alpha and beta receptors mediating the vasomotor responses of cerebral

arteries in vitro. Circ Res 35: 835-349.

Edvinsson L, Povlsen GK (2011). Late cerebral ischaemia after subarachnoid

haemorrhage: Is cerebrovascular receptor upregulation the mechanism behind?

Acta Physiol 203: 209-224.

Eftekhar B, Dadmehr M, Ansari S, Ghodsi M, Nazparvar B, Ketabchi E (2006). Are

the distributions of variations of circle of Willis different in different

populations? - Results of an anatomical study and review of literature. BMC

Neurol 6: 22.

Ehrreich S, Clopper E (1970). Physiologic and pharmacologic responses of

mammalian vascular smooth muscles during electric field stimulation.

Experientia 26: 1229.

Ekström-Jodal B (1970). On the relation between blood pressure and blood flow

in the canine brain with particular regard to the mechanism responsible for

cerebral blood flow autoregulation. Acta Physiol Scand Suppl 350: 1-61.

Ellis EF, Nies AS, Oates JA (1977). Cerebral arterial smooth muscle contraction

by thromboxane A2. Stroke 8: 480-483.

Ellis EF, Oelz O, Roberts LJ, 2nd, Payne NA, Sweetman BJ, Nies AS, et al. (1976).

Coronary arterial smooth muscle contraction by a substance released from

platelets: evidence that it is thromboxane A2. Science 193: 1135-1137.

Enoki T, Miwa S, Sakamoto A, Minowa T, Komuro T, Kobayashi S, et al. (1995).

Long-lasting activation of cation current by low concentration of endothelin-1

in mouse fibroblasts and smooth muscle cells of rabbit aorta. Br J Pharmacol

115: 479–485.

Evgenov OV, Pacher P, Schmidt PM, Haskó G, Schmidt HHHW, Stasch J-P (2006).

NO-independent stimulators and activators of soluble guanylate cyclase:

discovery and therapeutic potential. Nat Rev Drug Discov 5: 755-768.

Page 266: Cerebrovascular effects of vasoactive drugs

References

242

Faraci FM (1994). Nitric oxide and the cerebral circulation. Stroke 25: 692-703.

Faraci FM, Heistad DD (1990). Regulation of large cerebral arteries and cerebral

microvascular pressure. Circ Res 66: 8-17.

Fassbender K, Hodapp B, Rossol S, Bertsch T, Schmeck J, Schütt S, et al. (2001).

Inflammatory cytokines in subarachnoid haemorrhage: association with

abnormal blood flow velocities in basal cerebral arteries. J Neurol Neurosurg

Psychiatry 70: 534-537.

Fernandez N, Martinez MA, Garcia-Villalon AL, Monge L, Dieguez G (2001).

Cerebral vasoconstriction produced by vasopressin in conscious goats: role of

vasopressin V(1) and V(2) receptors and nitric oxide. Br J Pharmacol 132: 1837-

1844.

Ferrari M, Mottola L, Quaresima V (2004). Principles, Techniques, and

Limitations of Near Infrared Spectroscopy. Can J Appl Physiol 29: 463-487.

Fleming I (2001). Cytochrome p450 and vascular homeostasis. Circ Res 89: 753-

762.

Fleming I, Busse R (2006). Endothelium-derived epoxyeicosatrienoic acids and

vascular function. Hypertension 47: 629-633.

Fukata Y, Amano M, Kaibuchi K (2001). Rho-Rho-kinase pathway in smooth

muscle contraction and cytoskeletal reorganization of non-muscle cells. Trends

Pharmacol Sci 22: 32-39.

Furukawa F, Ito H, Nitta J, Tsujino M, Adachi S, Hiroe M, et al. (1992).

Endothelin-1 enhances calcium entry through T-type calcium channels in

cultured neonatal rat ventricular myocytes. Circ Res 71: 1242-1253.

Ganong WF (2000). Circumventricular organs: definition and role in the

regulation of endocrine and autonomic function. Clin Exp Pharmacol Physiol

27: 422-427.

Page 267: Cerebrovascular effects of vasoactive drugs

References

243

Garcia-Villalon AL, Garcia JL, Fernandez N, Monge L, Gomez B, Dieguez G

(1996). Regional differences in the arterial response to vasopressin: role of

endothelial nitric oxide. Br J Pharmacol 118: 1848-1854.

Garcia-Villalon AL, Krause DN, Ehlert FJ, Duckles SP (1991). Heterogeneity of

muscarinic receptor subtypes in cerebral blood vessels. J Pharmacol Exp Ther

258: 304-310.

Giller CA, Bowman G, Dyer H, Mootz L, Krippner W (1993). Cerebral arterial

diameters during changes in blood pressure and carbon dioxide during

craniotomy. Neurosurgery 32: 737-741.

Golias C, Charalabopoulos A, Stagikas D, Charalabopoulos K, Batistatou A

(2007). The kinin system--bradykinin: biological effects and clinical

implications. Multiple role of the kinin system--bradykinin. Hippokratia 11:

124-128.

Görlach C, Wahl M (1996). Bradykinin dilates rat middle cerebral artery and its

large branches via endothelial B2 receptors and release of nitric oxide. Peptides

17: 1373-1378.

Goto K, Fujii K, Abe I (2001). Impaired beta-adrenergic hyperpolarization in

arteries from prehypertensive spontaneously hypertensive rats. Hypertension

37: 609-613.

Goto K, Kasuya Y, Matsuki N, Takuwa Y, Kurihara H, Ishikawa T, et al. (1989).

Endothelin activates the dihydropyridine-sensitive, voltage-dependent Ca2+

channel in vascular smooth muscle. Proc Natl Acad Sci U S A 86: 3915-3918.

Guimarães S, Moura D (2001). Vascular adrenoceptors: an update. Pharmacol

Rev 53: 319-356.

Gustafsson F, Andreasen D, Salomonsson M, Jensen BL, Holstein-Rathlou NH

(2001). Conducted vasoconstriction in rat mesenteric arterioles: role for

dihydropyridine-insensitive Ca2+ channels. Am J Physiol- Heart C 280: 582-590.

Page 268: Cerebrovascular effects of vasoactive drugs

References

244

Hamel E (2004). Cholinergic modulation of the cortical microvascular bed. Prog

Brain Res 145: 171-178.

Hamel E (2006). Perivascular nerves and the regulation of cerebrovascular tone.

J Appl Physiol 100: 1059-1064.

Hamner JW, Tan CO, Lee K, Cohen MA, Taylor JA (2010). Sympathetic control

of the cerebral vasculature in humans. Stroke 41: 102-109.

Hamner JW, Tan CO, Tzeng Y-C, Taylor JA (2012). Cholinergic control of the

cerebral vasculature in humans. J Physiol: 6343.

Hardebo JE, Owman C (1980). Barrier mechanisms for neurotransmitter

monoamines and their precursors at the blood-brain interface. Ann Neurol 8:

1-31.

Harik SI, Hritz MA, LaManna JC (1995). Hypoxia-induced brain angiogenesis in

the adult rat. J Physiol 485 ( Pt 2): 525-530.

Harraz OF, Welsh DG (2013). Protein kinase A regulation of T-type Ca2+

channels in rat cerebral arterial smooth muscle. J Cell Sci 126: 2944-2954.

Hashemi SM, Mahmoodi R, Amirjamshidi A (2013). Variations in the Anatomy of

the Willis' circle: A 3-year cross-sectional study from Iran (2006-2009). Are the

distributions of variations of circle of Willis different in different populations?

Result of an anatomical study and review of literature. Surg Neurol Int 4: 65.

Haynes WG, Strachan FE, Webb DJ (1995). Endothelin ETA and ETB receptors

cause vasoconstriction of human resistance and capacitance vessels in vivo.

Circulation 92: 357-363.

Hedberg A, Carlsson E, Fellenius E, Lundgren B (1982). Cardiostimulatory

effects of prenalterol, a beta-1 adrenoceptor partial agonist, in vivo and in

vitro. Correlation between physiological effects and adenylate cyclase activity.

Naunyn Schmiedebergs Arch Pharmacol 318: 185-191.

Page 269: Cerebrovascular effects of vasoactive drugs

References

245

Heistad DD, Marcus ML (1978). Evidence that neural mechanisms do not have

important effects on cerebral blood flow. Circ Res 42: 295-302.

Hempelmann RG, Ziegler A (1993). Endothelium-dependent noradrenaline-

induced relaxation of rat isolated cerebral arteries: pharmacological

characterization of receptor subtypes involved. Br J Pharmacol 110: 1321-1328.

Highton D, Elwell C, Smith M (2010). Noninvasive cerebral oximetry: is there

light at the end of the tunnel? Curr Opin Anaesthesiol 23: 576-581.

Hirano K, Hirano M (2010). Current perspective on the role of the thrombin

receptor in cerebral vasospasm after subarachnoid hemorrhage. J Pharmacol

Sci 114: 127 – 133.

Hogestatt ED, Andersson KE (1984). On the postjunctional α-adrenoceptors in

rat cerebral and mesenteric arteries. J Auton Pharmacol 4: 161.

Holmes CL, Landry DW, Granton JT (2004). Science Review: Vasopressin and the

cardiovascular system part 2 - clinical physiology. Critical care (London,

England) 8: 15-23.

Holmes CL, Walley KR, Chittock DR, Lehman T, Russell JA (2001). The effects

of vasopressin on hemodynamics and renal function in severe septic shock: a

case series. Intensive Care Med 27: 1416-1421.

Huk I, Nanobashvili J, Neumayer C, Punz A, Mueller M, Afkhampour K, et al.

(1997). L-arginine treatment alters the kinetics of nitric oxide and superoxide

release and reduces ischemia/reperfusion injury in skeletal muscle. Circulation

96: 667-675.

Hunter JM, Kwan J, Malek-Ahmadi M, Maarouf CL, Kokjohn TA, Belden C, et al.

(2012). Morphological and pathological evolution of the brain microcirculation

in aging and Alzheimer's disease. PloS one 7: e36893.

Page 270: Cerebrovascular effects of vasoactive drugs

References

246

Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G (1987). Endothelium-

derived relaxing factor produced and released from artery and vein is nitric

oxide. Proc Natl Acad Sci U S A 84: 9265-9269.

Ihara E, Hirano K, Derkach DN, Nishimura J, Nawata H, Kanaide H (2000). The

mechanism of bradykinin-induced endothelium-dependent contraction and

relaxation in the porcine interlobar renal artery. Br J Pharmacol 129: 943-952.

Imai H, Okuno T, Wu JY, Lee TJ (1991). GABAergic innervation in cerebral blood

vessels: an immunohistochemical demonstration of L-glutamic acid

decarboxylase and GABA transaminase. J Cereb Blood Flow Metab 11: 129-134.

Immink RV, van Montfrans GA, Stam J, Karemaker JM, Diamant M, van Lieshout

JJ (2005). Dynamic cerebral autoregulation in acute lacunar and middle

cerebral artery territory ischemic stroke. Sroke 36: 2595-2600.

Ito U, Ohno K, Yamaguchi T, Takei H, Tomita H, Inaba Y (1980). Effect of

hypertension on blood-brain barrier. Change after restoration of blood flow in

post-ischemic gerbil brains. An electronmicroscopic study. Stroke 11: 606-611.

IUPHAR/BPS Guide to Pharmacology. http://www.guidetopharmacology.org/

GRAC/FamilyDisplayForward?familyId=4. Accessed on: 21/01/2015.

Iwamuro Y, Miwa S, Minowa T, Enoki T, Zhang X, Ishikawa M, et al. (1998).

Activation of two types of Ca2+-permeable nonselective cation channel by

endothelin-1 in A7r5 cells. Br J Pharmacol 124: 1541-1549.

Iwamuro Y, Miwa S, Zhang X, Minowa T, Enoki T, Okamoto Y, et al. (1999).

Activation of three types of voltage-independent Ca2+ channel in A7r5 cells by

endothelin-1 as revealed by a novel Ca2+ channel blocker LOE 908. Br J

Pharmacol 126: 1107-1114.

Jackson W (2000). Ion channels and vascular tone. Hypertension 35: 173-178.

Johansson BB (1980). The blood-brain barrier in acute and chronic

hypertension. Adv Exp Med Biol 131: 211-226.

Page 271: Cerebrovascular effects of vasoactive drugs

References

247

Jordan JD, Powers WJ (2012). Cerebral autoregulation and acute ischemic

stroke. Am J Hypertens 25: 946-950.

Kamitani T, Little MH, Ellis EF (1985). Evidence for a possible role of the brain

kallikrein-kinin system in the modulation of the cerebral circulation. Circ Res

57: 545-552.

Kanaide H, Ichiki T, Nishimura J, Hirano K (2003). Cellular mechanism of

vasoconstriction induced by angiotensin II: it remains to be determined. Circ

Res 93: 1015-1017.

Kardile PB, Ughade JM, Pandit SV, Ughade MN (2013). Anatomical variations of

anterior communicating artery. J Clin Diagn Res 7: 2661-2664.

Kästner S, Oertel MF, Scharbrodt W, Krause M, Böker DK, Deinsberger W (2005).

Endothelin-1 in plasma, cisternal CSF and microdialysate following aneurysmal

SAH. Acta Neurochir 147: 1271-1279.

Katusic ZS, Shepherd JT, Vanhoutte PM (1984). Vasopressin causes

endothelium-dependent relaxations of the canine basilar artery. Circ Res 55:

575-579.

Kaumann AJ, Levy FO (2006). 5-hydroxytryptamine receptors in the human

cardiovascular system. Pharmacol Ther 111: 674-706.

Kawanabe Y, Hashimoto N, Masaki T (2002). Characterization of Ca2+ channels

involved in endothelin-1-induced contraction of rabbit basilar artery. J

Cardiovasc Pharm 40: 438-447.

Kawanabe Y, Nauli SM (2005). Involvement of extracellular Ca2+ influx through

voltage-independent Ca2+ channels in endothelin-1 function. Cell Signal 17:

911-916.

Kedzierski RM, Yanagisawa M (2001). Endothelin system: the double-edged

sword in health and disease. Annu Rev Pharmacol Toxicol 41: 851-876.

Page 272: Cerebrovascular effects of vasoactive drugs

References

248

Kety S, Schmidt C (1948). The effects of altered arterial tensions of carbon

dioxide and oxygen on cerebral blood flow and cerebral oxygen consumption of

normal young men. J Clin Invest 27: 484-492.

Khalil RA (2010).Regulation of vascular smooth muscle function. [electronic

resource]. (ed). ed. Morgan & Claypool. San Rafael, USA. pp:

Khalil Z, Poliviou H, Maynard CJ, Beyreuther K, Masters CL, Li QX (2002).

Mechanisms of peripheral microvascular dysfunction in transgenic mice

overexpressing the Alzheimer's disease amyloid Abeta protein. J Alzheimers Dis

4: 467-478.

Kidd GA, Hong H, Majid A, Kaufman DI, Chen AF (2005). Inhibition of brain GTP

cyclohydrolase I and tetrahydrobiopterin attenuates cerebral infarction via

reducing inducible NO synthase and peroxynitrite in ischemic stroke. Stroke 36:

2705-2711.

Kikuchi K, Tanaka E, Murai Y, Tancharoen S (2014). Clinical Trials in Acute

Ischemic Stroke. CNS Drugs 28: 929-938.

Kilic T, Akakin A (2008). Anatomy of cerebral veins and sinuses. Front Neurol

Neurosci 23: 4-15.

Kim EC, Choi SK, Lim M, Yeon SI, Lee YH (2013). Role of endogenous ENaC and

TRP channels in the myogenic response of rat posterior cerebral arteries. PloS

one 8: e84194.

Kimura M, White RP, Wolf EW, Robertson JT (1998). Responses of human basilar

and other isolated arteries to novel nitric oxide donors. J Cardiovasc Pharmacol

32: 695-701.

Kirkman MA, Smith M (2014). Intracranial pressure monitoring, cerebral

perfusion pressure estimation, and ICP/CPP-guided therapy: a standard of care

or optional extra after brain injury? Brit J Anaesth 112: 35-46.

Page 273: Cerebrovascular effects of vasoactive drugs

References

249

Kobayashi H, Ide H, Ishii H, Kabuto M, Handa Y, Kubota T (1995). Endothelin-1

levels in plasma and cerebrospinal fluid following subarachnoid haemorrhage.

J Clin Neurosci 2: 252-256.

Komuro T, Miwa S, Zhang XF, Minowa T, Enoki T, Kobayashi S, et al. (1997).

Physiological role of Ca2+-permeable nonselective cation channel in endothelin-

1-induced contraction of rabbit aorta. J Cardiovasc Pharmacol 30: 504-509.

Kontos HA, Raper AJ, Patterson JL, Jr. (1971). Mechanisms of action of CO 2 on

pial precapillary vessels. Eur Neurol 6: 114-118.

Kontos HA, Wei EP, Navari RM, Levasseur JE, Rosenblum WI, Patterson JL, Jr.

(1978). Responses of cerebral arteries and arterioles to acute hypotension and

hypertension. Am J Physiol 234: H371-383.

Kovacs A, Harsing LG, Jr., Szenasi G (2012). Vasoconstrictor 5-HT receptors in

the smooth muscle of the rat middle cerebral artery. Eur J Pharmacol 689: 160-

164.

Küng CF, Tschudi MR, Noll G, Clozel JP, Lüscher TF (1995). Differential effects

of the calcium antagonist mibefradil in epicardial and intramyocardial coronary

arteries. J Cardiovasc Pharm 26: 312-318.

Kuo IY, Ellis A, Seymour VA, Sandow SL, Hill CE (2010). Dihydropyridine-

insensitive calcium currents contribute to function of small cerebral arteries. J

Cereb Blood Flow Metab 30: 1226-1239.

Kuo IYT, Wölfle SE, Hill CE (2011). T-type calcium channels and vascular

function: the new kid on the block? J Physiol 589: 783-795.

Labadia A, Costa G, Jimenez E, Triguero D, Garcia-Pascual A (1997). Endothelin

receptor-mediated Ca2+ mobilization and contraction in bovine oviductal

arteries: comparison with noradrenaline and potassium. Gen Pharmacol 29:

611-619.

Page 274: Cerebrovascular effects of vasoactive drugs

References

250

Laflam A, Joshi B, Brady K, Yenokyan G, Brown C, Everett A, et al. (2015).

Shoulder Surgery in the Beach Chair Position Is Associated with Diminished

Cerebral Autoregulation but No Differences in Postoperative Cognition or Brain

Injury Biomarker Levels Compared with Supine Positioning: The Anesthesia

Patient Safety Foundation Beach Chair Study. Anesth Analg 120: 176-185.

Lambert G, Naredi S, Eden E, Rydenhag B, Friberg P (2002). Sympathetic

nervous activation following subarachnoid hemorrhage: Influence of

intravenous clonidine. Acta Anaesthesiol Scand 46: 160-165.

Landry DW, Levin HR, Gallant EM, Seo S, D'Alessandro D, Oz MC, et al. (1997).

Vasopressin pressor hypersensitivity in vasodilatory septic shock. Crit Care Med

25: 1279-1282.

Langton PD, Standen NB (1993). Calcium currents elicited by voltage steps and

steady voltages in myocytes isolated from the rat basilar artery. J Physiol 469:

14.

Lassen NA (1959). Cerebral blood flow and oxygen consumption in man. Physiol

Rev 39: 183-238.

Lassen NA (1974). Control of cerebral circulation in health and disease. Circ Res

34: 749-760.

Leanne LC (2006). T-type Ca2+ channels in vascular smooth muscle: Multiple

functions. Cell Calcium 40: 221-230.

Ledoux J, Werner ME, Brayden JE, Nelson MT (2006). Calcium-activated

potassium channels and the regulation of vascular tone. Physiology 21: 69-78.

Lee RM (1995). Morphology of cerebral arteries. Pharmacol Ther 66: 149-173.

Lee TJ (1982). Cholinergic mechanism in the large cat cerebral artery. Circ Res

50: 870-879.

Lee TJ, Su C, Bevan JA (1976). Neurogenic sympathetic vasoconstriction of the

rabbit basilar artery. Circ Res 39: 120-126.

Page 275: Cerebrovascular effects of vasoactive drugs

References

251

Lee TJ, Zhang W, Sarwinski S (2000). Presynaptic beta(2)-adrenoceptors

mediate nicotine-induced NOergic neurogenic dilation in porcine basilar

arteries. Am J Physiol Heart Circ Physiol 279: H808-816.

Lee TJF, Kinkead LR, Sarwinski S (1982). Norepinephrine and acetylcholine

transmitter mechanisms in large cerebral arteries of the pig. J Cereb Blood

Flow Metab 2: 439-450.

Lew M, Angus J (1992). Wall thickness to lumen diameter ratios of arteries from

SHR and WKY: comparison of pressurised and wire-mounted preparations. J Vasc

Res 29: 435-442.

Lincoln J (1995). Innervation of cerebral arteries by nerves containing 5-

hydroxytryptamine and noradrenaline. Pharmacol Ther 68: 473-501.

Linfante I, Delgado-Mederos R, Andreone V, Gounis M, Hendricks L, Wakhloo AK

(2008). Angiographic and hemodynamic effect of high concentration of intra-

arterial nicardipine in cerebral vasospasm. Neurosurgery 63: 1080-1086;

discussion 1086-1087.

Lluch S, Conde MV, Diéguez G, López de Pablo AL, González MC, Estrada C, et

al. (1984). Evidence for the direct effect of vasopressin on human and goat

cerebral arteries. J Pharmacol Exp Ther 228: 749-755.

Logallo N, Kvistad CE, Nacu A, Naess H, Waje-Andreassen U, Asmuss J, et al.

(2014). The Norwegian tenecteplase stroke trial (NOR-TEST): randomised

controlled trial of tenecteplase vs. alteplase in acute ischaemic stroke. BMC

Neurology 14: 1-16.

Macdonald R, Higashida R, Keller E, Mayer S, Molyneux A, Raabe A, et al. (2012).

Randomized trial of clazosentan in patients with aneurysmal subarachnoid

hemorrhage undergoing endovascular coiling. Stroke 43: 1463-1469.

MacGregor DA, Prielipp RC, Butterworth JF, James RL, Royster RL (1996).

Relative efficacy and potency of beta-adrenoceptor agonists for generating

cAMP in human lymphocytes. Chest 109: 194-200.

Page 276: Cerebrovascular effects of vasoactive drugs

References

252

MacKenzie ET, Farrar JK, Fitch W, Graham DI, Gregory PC, Harper AM (1979).

Effects of hemorrhagic hypotension on the cerebral circulation. Cerebral blood

flow and pial arteriolar caliber. Stroke 10: 711-718.

MacKenzie ET, McCulloch J, O'Kean M, Pickard JD, Harper AM (1976). Cerebral

circulation and norepinephrine: relevance of the blood-brain barrier. Am J

Physiol 231: 483-488.

Maddahi A, Edvinsson L (2010). Cerebral ischemia induces microvascular pro-

inflammatory cytokine expression via the MEK/ERK pathway. J

Neuroinflammation 7: 1-13.

Marcus AJ, Weksler BB, Jaffe EA, Broekman MJ (1980). Synthesis of prostacyclin

from platelet-derived endoperoxides by cultured human endothelial cells. J

Clin Invest 66: 979-986.

Marrelli S, Eckmann M, Hunte M (2003). Role of endothelial intermediate

conductance KCa channels in cerebral EDHF-mediated dilations. Am J Physiol

Heart Circ Physiol 285: H1590-1599.

Marrelli SP (2001). Mechanisms of endothelial P2Y(1)- and P2Y(2)-mediated

vasodilatation involve differential [Ca2+]i responses. Am J Physiol Heart Circ

Physiol 281: H1759-1766.

Martin de Aguilera E, Vila JM, Irurzun A, Martinez MC, Martinez Cuesta MA, Lluch

S (1990). Endothelium-independent contractions of human cerebral arteries in

response to vasopressin. Stroke 21: 1689-1693.

Maturi MF, Martin SE, Markle D, Maxwell M, Burruss CR, Speir E, et al. (1991).

Coronary vasoconstriction induced by vasopressin. Production of myocardial

ischemia in dogs by constriction of nondiseased small vessels. Circulation 83:

2111-2121.

Mayhan WG (1991). Disruption of the blood-brain barrier in open and closed

cranial window preparations in rats. Stroke 22: 1059-1063.

Page 277: Cerebrovascular effects of vasoactive drugs

References

253

Meng L, Cannesson M, Alexander BS, Yu Z, Kain ZN, Cerussi AE, et al. (2011).

Effect of phenylephrine and ephedrine bolus treatment on cerebral oxygenation

in anaesthetized patients. Br J Anaesth 107: 209-217.

Meng L, Gelb AW (2015). Regulation of cerebral autoregulation by carbon

dioxide. Anesthesiology 122: 196-205.

Meng L, Gelb AW, McDonagh DL (2013). Changes in cerebral tissue oxygen

saturation during anaesthetic-induced hypotension: an interpretation based on

neurovascular coupling and cerebral autoregulation. Anaesthesia 68: 736-741.

Merlini M, Davalos D, Akassoglou K (2012). In vivo imaging of the neurovascular

unit in CNS disease. IntraVital 1: 87-94.

Micieli G, Tassorelli C, Bosone D, Cavallini A, Viotti E, Nappi G (1994).

Intracerebral vascular changes induced by cold pressor test: a model of

sympathetic activation. Neurol Res 16: 163-167.

Min J, Jin YM, Moon JS, Sung MS, Jo SA, Jo I (2006). Hypoxia-induced endothelial

NO synthase gene transcriptional activation is mediated through the tax-

responsive element in endothelial cells. Hypertension 47: 1189-1196.

Mitsis GD, Zhang R, Levine BD, Tzanalaridou E, Katritsis DG, Marmarelis VZ

(2009). Autonomic neural control of cerebral hemodynamics. IEEE Eng Med Biol

Mag 28: 54-62.

Miyamoto A, Ishiguro S, Nishio A (1999). Stimulation of bradykinin B2-receptors

on endothelial cells induces relaxation and contraction in porcine basilar artery

in vitro. Br J Pharmacol 128: 241-247.

Moller K, Qvist T, Tofteng F, Sahl C, Sonderkaer S, Dethloff T, et al. (2004).

Cerebral blood flow and metabolism during infusion of norepinephrine and

propofol in patients with bacterial meningitis. Stroke 35: 1333-1339.

Mombouli JV, Vanhoutte PM (1995). Kinins and endothelial control of vascular

smooth muscle. Annu Rev Pharmacol Toxicol 35: 679-705.

Page 278: Cerebrovascular effects of vasoactive drugs

References

254

Montani JP, Liard JF, Schoun J, Mohring J (1980). Hemodynamic effects of

exogenous and endogenous vasopressin at low plasma concentrations in

conscious dogs. Circ Res 47: 346-355.

Moppett IK, Sherman RW, Wild MJ, Latter JA, Mahajan RP (2008). Effects of

norepinephrine and glyceryl trinitrate on cerebral haemodynamics: transcranial

Doppler study in healthy volunteers. Br J Anaesth 100: 240-244.

Moraine JJ, Berre J, Melot C (2000). Is cerebral perfusion pressure a major

determinant of cerebral blood flow during head elevation in comatose patients

with severe intracranial lesions? J Neurosurg 92: 606-614.

Moseley BD, Britton JW, So E (2014). Increased cerebral oxygenation precedes

generalized tonic clonic seizures. Epilepsy Res 108: 1671-1674.

Motte S, McEntee K, Naeije R (2006). Endothelin receptor antagonists.

Pharmacol Ther 110: 386-414.

Moulds RF (1983). Techniques for testing isolated blood vessels. Gen Pharmacol

14: 47-53.

Moussouttas M, Huynh TT, Khoury J, Lai EW, Dombrowski K, Pello S, et al.

(2012). Cerebrospinal fluid catecholamine levels as predictors of outcome in

subarachnoid hemorrhage. Cerebrovasc Dis 33: 173-181.

Mulvany M (2004). Procedures for Investigation of small vessels using small

vessel myograph, Technology DM (ed). Denmark.

Mulvany MJ, Halpern W (1977). Contractile properties of small arterial

resistance vessels in spontaneously hypertensive and normotensive rats. Circ

Res 41: 19-26.

Mulvany MJ, Halpern W (1976). Mechanical properties of vascular smooth

muscle cells in situ. Nature 260: 617-619.

Murphy GS, Szokol JW (2011). Blood pressure management during beach chair

position shoulder surgery: What do we know? Can J Anaesth: 977.

Page 279: Cerebrovascular effects of vasoactive drugs

References

255

Myburgh JA (2005). Driving cerebral perfusion pressure with pressors: how,

which, when? Crit Care Resusc 7: 200-205.

Näbauer M, Callewaert G, Cleemann L, Morad M (1989). Regulation of calcium

release is rated by calcium current, not gating charge, in cardiac myocytes.

Science 244: 800-803.

Naredi S, Lambert G, Eden E, Zall S, Runnerstam M, Rydenhag B, et al. (2000).

Increased sympathetic nervous activity in patients with nontraumatic

subarachnoid hemorrhage. Stroke 31: 901-906.

Navarro-Gonzalez MF, Grayson TH, Meaney KR, Cribbs LL, Hill CE (2009). Non-

L-typr voltage-dependent calcium channels control vascular tone of the rat

basilar artery. Clin Exp Pharmacol P 36: 55-66.

Näveri L, Strömberg C, Saavedra JM (1994). Angiotensin II AT1 receptor

mediated contraction of the perfused rat cerebral artery. Neuroreport 5: 2278-

2280.

Neylon CB (1999). Vascular biology of endothelin signal transduction. Clin Exp

Pharmacol P 26: 149-153.

Nikitina E, Zhang Z-D, Kawashima A, Jahromi BS, Bouryi VA, Takahashi M, et al.

(2007). Voltage-dependent calcium channels of dog basilar artery. J Physiol

580: 523-541.

Nilsson D, Wackenfors A, Gustafsson L, Ugander M, Paulsson P, Ingemansson R,

et al. (2008). Endothelin receptor-mediated vasodilatation: effects of organ

culture. Eur J Pharmacol 579: 233-240.

Nilsson T, Cantera L, Edvinsson L (1996). Presence of neuropeptide Y Y1

receptor mediating vasoconstriction in human cerebral arteries. Neurosci Lett

204: 145-148.

Page 280: Cerebrovascular effects of vasoactive drugs

References

256

Nishimura N, Schaffer CB, Friedman B, Lyden PD, Kleinfeld D (2007).

Penetrating arterioles are a bottleneck in the perfusion of neocortex. Proc Natl

Acad Sci U S A 104: 365-370.

Nobler MS, Mann JJ, Sackeim HA (1999). Serotonin, cerebral blood flow, and

cerebral metabolic rate in geriatric major depression and normal aging. Brain

Res Brain Res Rev 30: 250-263.

Nosko M, Schulz R, Weir B, Cook DA, Grace M (1988). Effects of vasospasm on

levels of prostacyclin and thromboxane A2 in cerebral arteries of the monkey.

Neurosurgery 22: 45-50.

Nozaki K, Moskowitz MA, Maynard KI, Koketsu N, Dawson TM, Bredt DS, et al.

(1993). Possible origins and distribution of immunoreactive nitric oxide

synthase-containing nerve fibers in cerebral arteries. J Cereb Blood Flow Metab

13: 70-79.

Ogoh S, Brothers RM, Eubank WL, Raven PB (2008). Autonomic neural control of

the cerebral vasculature: acute hypotension. Stroke 39: 1979-1987.

Ogoh S, Sato K, Fisher JP, Seifert T, Overgaard M, Secher NH (2011). The effect

of phenylephrine on arterial and venous cerebral blood flow in healthy subjects.

Clin Physiol Funct Imaging 31: 445-451.

Ogoh S, Sato K, Okazaki K, Miyamoto T, Secher F, Sorensen H, et al. (2014). A

decrease in spatially resolved near-infrared spectroscopy-determined frontal

lobe tissue oxygenation by phenylephrine reflects reduced skin blood flow.

Anesth Analg 118: 823-829.

Olesen J (1972). The effect of intracarotid epinephrine, norepinephrine, and

angiotensin on the regional cerebral blood flow in man. Neurology 22: 978-987.

Ozkor MA, Quyyumi AA (2011). Endothelium-derived hyperpolarizing factor and

vascular function. Cardiol Res Pract 2011: 156146.

Page 281: Cerebrovascular effects of vasoactive drugs

References

257

Palomares SM, Cipolla MJ (2013). Myogenic Tone as a Therapeutic Target for

Ischemic Stroke. Curr Vasc Pharmacol.

Palomares SM, Cipolla MJ (2011). Vascular Protection Following Cerebral

Ischemia and Reperfusion. J Neurol Neurophysiol 2011.

Pardridge WM (2012). Drug transport across the blood-brain barrier. J Cereb

Blood Flow Metab 32: 1959-1972.

Parsons AA, Schilling L, Wahl M (1991). Analysis of acetylcholine-induced

relaxation of rabbit isolated middle cerebral artery: effects of inhibitors of

nitric oxide synthesis, Na,K-ATPase, and ATP-sensitive K channels. J Cereb

Blood Flow Metab 11: 700-704.

Partington T, Farmery A (2014). Physiology: Intracranial pressure and cerebral

blood flow. Anasth Intesive Care 15: 189-194.

Paulson OB, Strandgaard S, Edvinsson L (1990). Cerebral autoregulation.

Cerebrovasc Brain Metab Rev 2: 161-192.

Paulson OB, Waldemar G, Schmidt JF, Strandgaard S (1989). Cerebral

circulation under normal and pathologic conditions. The American journal of

cardiology 63: 2c-5c.

Peterson EC, Wang Z, Britz G (2011). Regulation of cerebral blood flow. Int J

Vasc Med. 2011: 823525-823525.

Petzold GC, Murthy VN (2011). Role of astrocytes in neurovascular coupling.

Neuron 71: 782-797.

Pickard JD, Murray GD, Illingworth R, Shaw MDM, Teasdale GM, Foy PM, et al.

(1989). Effect of oral nimodipine on cerebral infarction and outcome after

subarachnoid haemorrhage: British aneurysm nimodipine trial. British Medical

Association. pp: 636.

Page 282: Cerebrovascular effects of vasoactive drugs

References

258

Pires PW, Dams Ramos CM, Matin N, Dorrance AM (2013). The effects of

hypertension on the cerebral circulation. Am J Physiol Heart Circ Physiol 304:

H1598-1614.

Pluta RM (2006). Dysfunction of nitric oxide synthases as a cause and

therapeutic target in delayed cerebral vasospasm after SAH. Neurol Res 28:

730-737.

Pluta RM, Oldfield EH, Boock RJ (1997). Reversal and prevention of cerebral

vasospasm by intracarotid infusions of nitric oxide donors in a primate model of

subarachnoid hemorrhage. J Neurosurg 87: 746-751.

Pohl A, Cullen DJ (2005). Cerebral ischemia during shoulder surgery in the

upright position: a case series. Journal of clinical anesthesia 17: 463-469.

Poulin MJ, Robbins PA (1998). Influence of cerebral blood flow on the

ventilatory response to hypoxia in humans. Exp Physiol 83: 95-106.

Poulsen CB, Al-Mashhadi RH, Cribbs LL, Skøtt O, Hansen PB (2011). T-type

voltage-gated calcium channels regulate the tone of mouse efferent arterioles.

Kidney Int 79: 443-451.

Pradilla G, Chaichana KL, Hoang S, Huang J, Tamargo RJ (2010). Inflammation

and cerebral vasospasm after subarachnoid hemorrhage. Neorosurg Clin N Am

21: 365-379.

Priviero FBM, Webb RC (2010). Heme-Dependent and Independent Soluble

Guanylate Cyclase Activators and Vasodilation. J Cardiovasc Pharm 56: 229.

Qi M, Hang C, Zhu L, Shi J (2011). Involvement of endothelial-derived relaxing

factors in the regulation of cerebral blood flow. Neurol Sci 32: 551-557.

QingHua Z, XingTing L, GouGan Z, WenJie Z, ChengWen S (2009). Endothelin-1

induces intracellular [Ca2+] increase via Ca2+ influx through the L-type Ca2+

channel, Ca2+-induced Ca2+ release and a pathway involving ETA receptors, PKC,

PKA and AT1 receptors in cardiomyocytes. Sci China Ser C-Life Sci 52: 360-370.

Page 283: Cerebrovascular effects of vasoactive drugs

References

259

Radnoti Glass Technology I (2010). Radnoti Tissue-Organ Bath Principles, Ltd AP

(ed).

Rahimi SY, Browns JH, Macomson SD, Jensen MA, Alleyne CH (2006). Evolution

of the treatment of cerebral vasospasm. Neurosurg Focus 20: E4.

Rahman AM, Murrow JR, Ozkor MA, Kavtaradze N, Lin J, De Staercke C, et al.

(2014). Endothelium-derived hyperpolarizing factor mediates bradykinin-

stimulated tissue plasminogen activator release in humans. J Vasc Res 51: 200-

208.

Raichle ME, Hartman BK, Eichling JO, Sharpe LG (1975). Central noradrenergic

regulation of cerebral blood flow and vascular permeability. Proc Natl Acad Sci

U S A 72: 3726-3730.

Rasmussen H, Takuwa Y, Park S (1987). Protein kinase C in the regulation of

smooth muscle contraction. FASEB J 1: 177-185.

Regoli D, Barabe J (1980). Pharmacology of bradykinin and related kinins.

Pharmacol Rev 32: 1-46.

Roatta S, Micieli G, Bosone D, Losano G, Bini R, Cavallini A, et al. (1998). Effect

of generalised sympathetic activation by cold pressor test on cerebral

haemodynamics in healthy humans. J Auton Nerv Syst 71: 159-166.

Ruskell GL, Simons T (1987). Trigeminal nerve pathways to the cerebral arteries

in monkeys. J Anat 155: 23-37.

Sabri M, Ai J, Knight B, Tariq A, Jeon H, Shang X, et al. (2011). Uncoupling of

endothelial nitric oxide synthase after experimental subarachnoid hemorrhage.

J Cereb Blood Flow Metab 31: 190-199.

Saema A, Carl L, Aida M, Lars E (2010). Research Report: Subarachnoid

hemorrhage induces enhanced expression of thromboxane A2 receptors in rat

cerebral arteries. Brain Res 1316: 163-172.

Page 284: Cerebrovascular effects of vasoactive drugs

References

260

Saleh NS, Alber AP, Large WA (2009). Activation of native TRPC1/C5/C6

channels by endothelin-1 is mediated by both PIP3 and PIP2 in rabbit coronary

artery myocytes. J Physiol 587: 5361-5375.

Salmon M (2014). Physiological salt solutions. In: (ed). 1st ed. Practical

Pharmacology for the Pharmaceutical Sciences John Wiley & Sons Ltd.

Chichester, UK.

Salom JB, Barbera MD, Centeno JM, Orti M, Torregrosa G, Alborch E (1999).

Relaxant effects of sodium nitroprusside and NONOates in goat middle cerebral

artery: delayed impairment by global ischemia-reperfusion. Nitric Oxide 3: 85-

93.

Salom JB, Barbera MD, Centeno JM, Orti M, Torregrosa G, Alborch E (1998).

Relaxant effects of sodium nitroprusside and NONOates in rabbit basilar artery.

Pharmacology 57: 79-87.

Salom JB, Torregrosa G, Miranda FJ, Alabadí JA, Alvarez C, Alborch E (1991).

Effects of endothelin-1 on the cerebrovascular bed of the goat. Eur J Pharmacol

192: 39-45.

Sanchez A, Fernandez N, Monge L, Salcedo A, Climent B, Luis Garcia-Villalon A,

et al. (2006). Goat cerebrovascular reactivity to ADP after ischemia-

reperfusion. Role of nitric oxide, prostanoids and reactive oxygen species. Brain

Res 1120: 114-123.

Sato A, Sato Y, Uchida S (2001). Regulation of regional cerebral blood flow by

cholinergic fibers originating in the basal forebrain. Int J Dev Neurosci 19: 327-

337.

Schäfer A, Flierl U, Kobsar A, Eigenthaler M, Ertl G, Bauersachs J (2006). Soluble

guanylyl cyclase activation with HMR1766 attenuates platelet activation in

diabetic rats. Arteriosclerosis, Thrombosis, And Vascular Biology 26: 2813-

2818.

Page 285: Cerebrovascular effects of vasoactive drugs

References

261

Schaller B (2004). Physiology of cerebral venous blood flow: from experimental

data in animals to normal function in humans. Brain Res Brain Res Rev 46: 243-

260.

Scherer EQ, Herzog M, Wangemann P (2002). Endothelin-1-induced vasospasms

of spiral modiolar artery are mediated by rho-kinase-induced Ca(2+)

sensitization of contractile apparatus and reversed by calcitonin gene-related

Peptide. Stroke 33: 2965-2971.

Scholkmann F, Kleiser S, Metz AJ, Zimmermann R, Mata Pavia J, Wolf U, et al.

(2014). A review on continuous wave functional near-infrared spectroscopy and

imaging instrumentation and methodology. Neuroimage 85 Pt 1: 6-27.

Schubert R, Krien U, Wulfsen I, Schiemann D, Lehmann G, Ulfig N, et al. (2004).

Nitric oxide donor sodium nitroprusside dilates rat small arteries by activation

of inward rectifier potassium channels. Hypertension 43: 891-896.

Semenas E, Sharma HS, Wiklund L (2014). Adrenaline increases blood-brain-

barrier permeability after haemorrhagic cardiac arrest in immature pigs. Acta

Anaesthesiol Scand 58: 620-629.

Shimizu T, Koto A, Suzuki N, Morita Y, Takao M, Otomo S, et al. (1999).

Occurrence and distribution of substance P receptors in the cerebral blood

vessels of the rat. Brain Res 830: 372-378.

Singh A, Hildebrand ME, Garcia E, Snutch TP (2010). The transient receptor

potential channel antagonist SKF96365 is a potent blocker of low-voltage-

activated T-type calcium channels. Br J Pharmacol 160: 1464-1475.

Soeding PF, Hoy S, Hoy G, Evans M, Royse CF (2013). Effect of phenylephrine

on the haemodynamic state and cerebral oxygen saturation during anaesthesia

in the upright position. Br J Anaesth 111: 229-234.

Soeding PF, Wang J, Hoy G, Jarman P, Phillips H, Marks P, et al. (2011). The

effect of the sitting upright or 'beachchair' position on cerebral blood flow

during anaesthesia for shoulder surgery. Anaesth Intensive Care 39: 440-448.

Page 286: Cerebrovascular effects of vasoactive drugs

References

262

Sorensen H, Secher NH, Siebenmann C, Nielsen HB, Kohl-Bareis M, Lundby C, et

al. (2012). Cutaneous vasoconstriction affects near-infrared spectroscopy

determined cerebral oxygen saturation during administration of

norepinephrine. Anesthesiology 117: 263-270.

Spierings EL (2003). Pathogenesis of the migraine attack. Clin J Pain 19: 255-

262.

Spiers A, Padmanabhan N (2005). A guide to wire myography. Methods Mol Med

108: 91-104.

Srikiatkhachorn A, Suwattanasophon C, Ruangpattanatawee U, Phansuwan-

Pujito P (2002). 2002 Wolff Award. 5 -HT2A receptor activation and nitric oxide

synthesis: a possible mechanism determining migraine attacks. Headache 42:

566-574.

Stasch JP, Pacher P, Evgenov OV (2011). Soluble guanylate cyclase as an

emerging therapeutic target in cardiopulmonary disease. Circulation 123:

2263-2273.

Stenman E, Edvinsson L (2004). Cerebral ischemia enhances vascular

angiotensin AT1 receptor-mediated contraction in rats. Stroke 35: 970-974.

Stephen CJ (2003). Nitric Oxide Synthase Inhibition Depresses the Height of the

Cerebral Blood Flow-Pressure Autoregulation Curve During Moderate

Hypotension. J Cereb Blood Flow Metab 23: 1085.

Strandgaard S, Olesen J, Skinhoj E, Lassen NA (1973). Autoregulation of brain

circulation in severe arterial hypertension. Br Med J 1: 507-510.

Strange K (1992). Regulation of solute and water balance and cell volume in the

central nervous system. Journal of the American Society of Nephrology : JASN

3: 12-27.

Suhardja A (2004). Mechanisms of disease: roles of nitric oxide and endothelin-

1 in delayed cerebral vasospasm produced by aneurysmal subarachnoid

Page 287: Cerebrovascular effects of vasoactive drugs

References

263

hemorrhage. Nat Clin Pract Cardiovasc Med 1: 110-116; quiz 112 p following

116.

Summors AC, Gupta AK, Matta BF (1999). Dynamic cerebral autoregulation

during sevoflurane anesthesia: a comparison with isoflurane. Anesth Analg 88:

341-345.

Suzuki Y, Satoh S, Kimura M, Oyama H, Asano T, Shibuya M, et al. (1992). Effects

of vasopressin and oxytocin on canine cerebral circulation in vivo. J Neurosurg

77: 424-431.

Suzuki Y, Satoh S, Oyama H, Takayasu M, Shibuya M (1993). Regional differences

in the vasodilator response to vasopressin in canine cerebral arteries in vivo.

Sroke 24: 1049-1053.

Suzuki Y, Satoh S, Oyama H, Takayasu M, Shibuya M, Sugita K (1994).

Vasopressin mediated vasodilation of cerebral arteries. J Auton Nerv Syst 49

Suppl: S129-132.

Taddei S, Ghiadoni L, Virdis A, Buralli S, Salvetti A (1999). Vasodilation to

bradykinin is mediated by an ouabain-sensitive pathway as a compensatory

mechanism for impaired nitric oxide availability in essential hypertensive

patients. Circulation 100: 1400-1405.

Takayasu M, Kajita Y, Suzuki Y, Shibuya M, Sugita K, Ishikawa T, et al. (1993).

Triphasic response of rat intracerebral arterioles to increasing concentrations

of vasopressin in vitro. J Cereb Blood Flow Metab 13: 304-309.

Tamargo J, Duarte J, Caballero R, Delpon E (2010). Cinaciguat, a soluble

guanylate cyclase activator for the potential treatment of acute heart failure.

Curr Opin Investig Drugs 11: 1039-1047.

Tanaka H, Fujita N, Enoki T, Matsumoto K, Watanabe Y, Murase K, et al. (2006).

Relationship between variations in the circle of Willis and flow rates in internal

carotid and basilar arteries determined by means of magnetic resonance

Page 288: Cerebrovascular effects of vasoactive drugs

References

264

imaging with semiautomated lumen segmentation: reference data from 125

healthy volunteers. AJNR Am J Neuroradiol 27: 1770-1775.

Tani E (2002). Molecular mechanism involved in development of cerebral

vasospasm. Neurosurg Focus 12: 1-8.

Tanishima T (1983). Cerebral vasospasm: comparison of contractile responses

in isolated human and canine basilar arteries. No To Shinkei 35: 323-329.

Tejerina T, Chulia T, Gonzalez P (1993). Effects of dotarizine on 45Ca2+

movements and contractile responses in vascular smooth muscle. Eur J

Pharmacol 239 7.

Tepper SJ, Rapoport AM, Sheftell FD (2002). Mechanisms of action of the 5-

HT1B/1D receptor agonists. Arch Neurol 59: 1084-1088.

ter Laan M, van Dijk JM, Elting JW, Staal MJ, Absalom AR (2013). Sympathetic

regulation of cerebral blood flow in humans: a review. Br J Anaesth 111: 361-

367.

Terpolilli NA, Moskowitz MA, Plesnila N (2012). Nitric oxide: considerations for

the treatment of ischemic stroke. J Cereb Blood Flow Metab 32: 1332-1346.

The Australian. http://www.theaustralian.com.au/news/latest-news/

anaesthetist-shocked-at-mans-op-death/story-fn3dxiwe-1226627095146.

Accessed on: 19/01/2015.

Toda N (1979). Acetylcholine-induced relaxation in isolated dog cerebral

arteries. J Pharmacol Exp Ther 209: 352-358.

Toda N (1984). Endothelium-dependent relaxation induced by angiotensin II and

histamine in isolated arteries of dog. Br J Pharmacol 81: 301-307.

Toda N, Ayajiki K, Okamura T (2009). Cerebral blood flow regulation by nitric

oxide in neurological disorders. Can J Physiol Pharm 87: 581-594.

Page 289: Cerebrovascular effects of vasoactive drugs

References

265

Toda N, Fujita Y (1973). Responsiveness of isolated cerebral and peripheral

arteries to serotonin, norepinephrine, and transmural electrical stimulation.

Circ Res 33: 98-104.

Toda N, Okamura T (1993). Cerebral vasoconstrictor mediators. Pharmacol Ther

57: 359-375.

Toda N, Okamura T (1998). Cerebral vasodilators. Jpn J Pharmacol 76: 349-367.

Toda N, Okamura T (1990). Endothelium-derived relaxing factor (EDRF). Nihon

yakurigaku zasshi. Folia pharmacologica Japonica 95: 295-308.

Tousoulis D, Kampoli A-M, Tentolouris C, Papageorgiou N, Stefanadis C (2012).

The role of nitric oxide on endothelial function. Curr Vasc Pharmacol 10: 4-18.

Touyz RM, Schiffrin EL (2000). Signal transduction mechanisms mediating the

physiological and pathophysiological actions of angiotensin II in vascular smooth

muscle cells. Pharmacol Rev 52: 639-672.

Trandafir CC, Nishihashi T, Wang A, Murakami S, Ji X, Kurahashi K (2004).

Participation of vasopressin in the development of cerebral vasospasm in a rat

model of subarachnoid haemorrhage. Clin Exp Pharmacol Physiol 31: 261-266.

Treschan TA, Peters J (2006). The vasopressin system: physiology and clinical

strategies. Anesthesiology 105: 599-612; quiz 639-540.

Tsai M, Lee C, Lin M (1989). Responses of cerebral circulation produced by

adrenoceptor agonists and antagonists in rats. Neuropharmacology 28: 1075-

1080.

Tsai S, Tew J, McLean J, Shipley M (1988). Cerebral arterial innervation by nerve

fibers containing calcitonin gene-related peptide (CGRP): I. Distribution and

origin of CGRP perivascular innervation in the rat. J Comp Neurol 271: 435-444.

Tsukahara T, Taniguchi T, Miwa S, Shimohama S, Fujiwara M, Nishikawa M, et

al. (1988). Presynaptic and postsynaptic alpha 2-adrenergic receptors in human

Page 290: Cerebrovascular effects of vasoactive drugs

References

266

cerebral arteries and their alteration after subarachnoid hemorrhage. Stroke

19: 80-83.

Tsukahara T, Taniguchi T, Shimohama S, Fujiwara M, Handa H (1986a).

Characterization of beta adrenergic receptors in human cerebral arteries and

alteration of the receptors after subarachnoid hemorrhage. Stroke 17: 202-207.

Tsukahara T, Usui H, Taniguchi T, Shimohama S, Fujiwara M, Handa H (1986b).

Characterization of muscarinic cholinergic receptors in human and dog cerebral

arteries. Stroke 17: 300-305.

Tuor UI (1992). Acute hypertension and sympathetic stimulation: local

heterogeneous changes in cerebral blood flow. Am J Physiol 263: H511-518.

Turner RC, Dodson SC, Rosen CL, Huber JD (2013). The science of cerebral

ischemia and the quest for neuroprotection: navigating past failure to future

success A review. J Neurosurg 118: 1075-1085.

Tykocki NR, Watts SW (2010). The interdependence of endothelin-1 and

calcium: a review. Clin Sci 119: 361-372.

University of Miami Health System http://surgery.med.miami.edu/

images/Circulation_of_brain.gif. http://surgery.med.miami.edu/images/

Circulation_of_brain.gif. Accessed on: 20/01/2015.

Uski T, Andersson KE, Brandt L, Edvinsson L, Ljunggren B (1983). Responses of

isolated feline and human cerebral arteries to prostacyclin and some of its

metabolites. J Cereb Blood Flow Metab 3: 238-245.

van Beek AH, Claassen JA, Rikkert MG, Jansen RW (2008). Cerebral

autoregulation: an overview of current concepts and methodology with special

focus on the elderly. J Cereb Blood Flow Metab 28: 1071-1085.

Vecchione C, Frati A, Di Pardo A, Cifelli G, Carnevale D, Gentile MT, et al.

(2009). Tumor necrosis factor-alpha mediates hemolysis-induced

Page 291: Cerebrovascular effects of vasoactive drugs

References

267

vasoconstriction and the cerebral vasospasm evoked by subarachnoid

hemorrhage. Hypertension 54: 150-156.

Vincent JM, Kwan YW, Chan SL, Perrin-Sarrado C, Atkinson J, Chillon JM (2005).

Constrictor and dilator effects of angiotensin II on cerebral arterioles. Stroke

36: 2691-2695.

Wabnitz H, Moeller M, Liebert A, Obrig H, Steinbrink J, Macdonald R (2010).

Time-resolved near-infrared spectroscopy and imaging of the adult human

brain. Adv Exp Med Biol 662: 143-148.

Wahl M, Gorlach C, Hortobagyi T, Benyo Z (1999). Effects of bradykinin in the

cerebral circulation. Acta Physiol Hung 86: 155-160.

Wahl M, Young AR, Edvinsson L, Wagner F (1983). Effects of Bradykinin on Pial

Arteries and Arterioles In Vitro and In Situ. J Cereb Blood Flow Metab 3: 231.

Wallis SJ, Firth J, Dunn WR (1996). Pressure-induced myogenic responses in

human isolated cerebral resistance arteries. Sroke 27: 2287-2290.

Wei EP, Kontos HA, Patterson JL, Jr. (1978). Vasoconstrictor effect of

angiotensin on pial arteries. Stroke 9: 487-489.

Weissmann N, Hackemack S, Dahal B, Pullamsetti S, Savai R, Mittal M, et al.

(2009). The soluble guanylate cyclase activator HMR1766 reverses hypoxia-

induced experimental pulmonary hypertension in mice. Am J Physiol Lung Cell

Mol Physiol 297: L658-665.

Wellman GC (2006). Ion channels and calcium signaling in cerebral arteries

following subarachnoid hemorrhage. Neurol Res 28: 690-702.

Wendling W, Harakal C (1987). Effects of calcium antagonists on isolated bovine

cerebral arteries: inhibition of constriction and calcium-45 uptake induced by

potassium or serotonin. Stroke 18: 591-598.

Page 292: Cerebrovascular effects of vasoactive drugs

References

268

Wendling WW, Harakal C (1991). Effects of prostaglandin-F2-alpha and

thromboxane-A2 analog on bovine cerebral arterial tone and calcium fluxes.

Stroke 22: 66-72.

Westcott EB, Segal SS (2013). Perivascular Innervation: A Multiplicity of Roles

in Vasomotor Control and Myoendothelial Signaling. Microcirculation: 217.

Whalley E, Fritz H, Geiger R (1983). Kinin receptors and angiotensin converting

enzyme in rabbits basilar arteries. Naunyn Schmiedebergs Arch Pharmacol 324:

296.

Whalley ET, Wahl M (1983). Analysis of bradykinin receptor mediating

relaxation of cat cerebral arteries in vivo and in vitro. Naunyn Schmiedebergs

Arch Pharmacol 323: 66-71.

Williams TD, Da Costa D, Mathias CJ, Bannister R, Lightman SL (1986). Pressor

effect of arginine vasopressin in progressive autonomic failure. Clin Sci 71: 173-

178.

Winquist RJ, Bohr DF (1982). Characterization of the rat basilar artery in vitro.

Experientia 38: 1187-1188.

Wolburg H, Neuhaus J, Kniesel U, Krauss B, Schmid EM, Ocalan M, et al. (1994).

Modulation of tight junction structure in blood-brain barrier endothelial cells.

Effects of tissue culture, second messengers and cocultured astrocytes. J Cell

Sci 107 ( Pt 5): 1347-1357.

Wynne BM, Chiao C, Webb RC (2009). Review Article: Vascular smooth muscle

cell signaling mechanisms for contraction to angiotensin II and endothelin-1.

Am J Hypertens 3: 84-95.

Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, et al.

(1988). A novel potent vasoconstrictor peptide produced by vascular

endothelial cells. Nature 332: 411-415.

Page 293: Cerebrovascular effects of vasoactive drugs

References

269

Yu DY, Su EN, Cringle SJ, Yu PK (2003). Isolated preparations of ocular

vasculature and their applications in ophthalmic research. Prog Retin Eye Res

22: 135-169.

Zaidi SM, Heller H (1974). Can neurohypophysial hormones cross the blood-

cerebrospinal fluid barrier? J Endocrinol 60: 195-196.

Zhang ET, Inman CB, Weller RO (1990). Interrelationships of the pia mater and

the perivascular (Virchow-Robin) spaces in the human cerebrum. J Anat 170:

111-123.

Zhang R, Zuckerman JH, Iwasaki K, Wilson TE, Crandall CG, Levine BD (2002).

Autonomic neural control of dynamic cerebral autoregulation in humans.

Circulation 106: 1814-1820.

Zhang W, Edvinsson L, Lee TJ (1998). Mechanism of nicotine-induced relaxation

in the porcine basilar artery. J Pharmacol Exp Ther 284: 790-797.

Zhang X, Iwamuro Y, Enoki T, Okazawa M, Lee A, Komuro T, et al. (1999).

Pharmacological characterization of Ca2+ entry channels in endothelin-1-

induced contraction of rat aorta using LOE 908 and SK&F 96365. Br J Pharmacol

127: 1388-1398.

Zhang X, Iwamuro Y, Okamoto Y, Kawanabe Y, Masaki T, Miwa S (2000).

Endothelin-1-induced contraction of rat thoracic aorta depends on calcium

entry through three types of calcium channel. J Cardiovasc Pharm 36: S105-

S106.

Zheng F, Sheinberg R, Yee MS, Ono M, Zheng Y, Hogue CW (2013). Cerebral

near-infrared spectroscopy monitoring and neurologic outcomes in adult

cardiac surgery patients: a systematic review. Anesth Analg 116: 663-676.

Zhong F, Pan J, Liu X, Wang H, Ying T, Su J, et al. (2011). A novel insight into

the heme and NO/CO binding mechanism of the alpha subunit of human soluble

guanylate cyclase. J Biol Inorg Chem 16: 1227-1239.

Page 294: Cerebrovascular effects of vasoactive drugs

References

270

Zlokovic BV, Hyman S, McComb JG, Lipovac MN, Tang G, Davson H (1990).

Kinetics of arginine-vasopressin uptake at the blood-brain barrier. Biochimica

et biophysica acta 1025: 191-198.

Page 295: Cerebrovascular effects of vasoactive drugs

Minerva Access is the Institutional Repository of The University of Melbourne

Author/s:MAMO, YOHANNES AYELE

Title:Cerebrovascular effects of vasoactive drugs: in vitro, in vivo and clinical investigations

Date:2015

Persistent Link:http://hdl.handle.net/11343/55348