a biophysically detailed mathematical model of a …

243
A BIOPHYSICALLY DETAILED MATHEMATICAL MODEL OF A SINGLE LATE PREGNANT RAT MYOMETRIAL CELL A thesis submitted to the University of Manchester for the degree of Doctor of Philosophy in the Faculty of Engineering and Physical Sciences 2010 By Cecilia Choi School of Physics and Astronomy

Upload: others

Post on 17-Mar-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

A BIOPHYSICALLY DETAILED

MATHEMATICAL MODEL OF A

SINGLE LATE PREGNANT RAT

MYOMETRIAL CELL

A thesis submitted to the University of Manchester

for the degree of Doctor of Philosophy

in the Faculty of Engineering and Physical Sciences

2010

By

Cecilia Choi

School of Physics and Astronomy

Contents

Abstract 11

Declaration 12

Copyright 13

Acknowledgements 14

1 Introduction 15

1.1 Structure of the uterus . . . . . . . . . . . . . . . . . . . . . . . . 16

1.2 Electrical activity and action potential . . . . . . . . . . . . . . . 18

1.3 Cell membrane properties and ionic currents . . . . . . . . . . . . 25

1.4 Ionic channels in myometrium . . . . . . . . . . . . . . . . . . . . 30

1.5 Ionic pumps and exchanger . . . . . . . . . . . . . . . . . . . . . 31

1.6 Calcium handling and uterine contractility . . . . . . . . . . . . . 31

1.7 Patch-clamp experiment . . . . . . . . . . . . . . . . . . . . . . . 37

2 Single uterine cell model development 41

2.1 Membrane current model in myometrium . . . . . . . . . . . . . . 44

2.1.1 Inward currents . . . . . . . . . . . . . . . . . . . . . . . . 48

2.1.1.1 Hyperpolarisation-activated current – Ih . . . . . 48

2.1.1.2 L-type calcium current – ICaL . . . . . . . . . . . 52

2.1.1.3 T-type calcium current – ICaT . . . . . . . . . . 56

2.1.1.4 Fast sodium current – INa . . . . . . . . . . . . . 57

2.1.1.5 Store-operated non-selective cation current – ISOC 60

2.1.2 Outward currents . . . . . . . . . . . . . . . . . . . . . . . 63

2.1.2.1 Delayed rectifying potassium current – IK1 . . . 63

2.1.2.2 Delayed rectifying potassium current – IK2 . . . 67

2

2.1.2.3 A-type transient potassium current – IKA . . . . 70

2.1.2.4 Calcium-activated K+ current – IKCa . . . . . . . 73

2.1.2.5 Background K+ leakage current – IKleak . . . . . 75

2.1.3 Other currents . . . . . . . . . . . . . . . . . . . . . . . . 76

2.1.3.1 Non-selective cation current – INSCC . . . . . . . 76

2.1.3.2 Calcium-activated chloride current – ICl . . . . . 78

2.1.4 Pumps and exchanger currents . . . . . . . . . . . . . . . . 81

2.1.4.1 Calcium pump current – ICapump . . . . . . . . . 81

2.1.4.2 Sodium/Potassium exchanger current – INaK . . 84

2.1.4.3 Sodium/Calcium exchanger current – INaCa . . . 85

2.1.4.4 Sodium-potassium-chloride co-transport current –

INaKCl . . . . . . . . . . . . . . . . . . . . . . . . 88

2.2 Intracellular dynamics and SR . . . . . . . . . . . . . . . . . . . . 89

2.2.1 SR currents, Iup , Itr and Irel . . . . . . . . . . . . . . . . . 89

2.2.2 Ionic balances . . . . . . . . . . . . . . . . . . . . . . . . . 92

2.3 Force production in myometrium . . . . . . . . . . . . . . . . . . 94

2.3.1 Kinetic cross-bridge cycling model in smooth muscles . . . 94

2.3.2 Mechanical production in smooth muscles . . . . . . . . . 99

2.3.2.1 Passive force . . . . . . . . . . . . . . . . . . . . 101

2.3.2.2 Cross-bridge elastic force . . . . . . . . . . . . . . 101

2.3.2.3 Active force . . . . . . . . . . . . . . . . . . . . . 101

2.3.2.4 Series viscoelastic force . . . . . . . . . . . . . . . 103

2.3.2.5 Total force produced by the cell . . . . . . . . . . 103

2.4 Solutions of the model equations . . . . . . . . . . . . . . . . . . . 104

3 Results I – Electrophysiological response 107

3.1 Whole cell membrane current and AP in control condition . . . . 107

3.1.1 [ Ca2+ ]i and ICa−tot responses on repetitive depolarisation . 108

3.2 Whole cell membrane current and AP under estradiol effect . . . . 112

3.3 Behaviour of all the membrane and SR currents during current-clamp114

3.3.1 Ca2+ -related currents . . . . . . . . . . . . . . . . . . . . 114

3.3.2 Na+ -related currents . . . . . . . . . . . . . . . . . . . . . 116

3.3.3 K+ -related currents . . . . . . . . . . . . . . . . . . . . . 116

3.3.4 Other currents . . . . . . . . . . . . . . . . . . . . . . . . 119

3.4 Investigation of AP and total membrane current while inhibiting

individual currents . . . . . . . . . . . . . . . . . . . . . . . . . . 119

3

3.4.1 Inhibiting the inward currents . . . . . . . . . . . . . . . . 119

3.4.2 Inhibiting the outward currents . . . . . . . . . . . . . . . 122

3.4.3 Inhibiting the other currents . . . . . . . . . . . . . . . . . 125

3.5 Different types of AP . . . . . . . . . . . . . . . . . . . . . . . . . 125

4 Results II – Mechanical production 129

4.1 Fractional stress production in myometrium . . . . . . . . . . . . 129

4.2 Single-, twin- and multiple-pulse simulation in myometrium . . . . 130

4.3 Other mechanical responses in a single uterine cell . . . . . . . . . 132

5 Drug actions on [ Ca2+ ]i and force 138

5.1 Properties of [ Ca2+ ]i transient in myometrial cells . . . . . . . . . 138

5.1.1 Depolarising pulse with varying magnitude . . . . . . . . . 139

5.1.2 Carboxyeosin, CE . . . . . . . . . . . . . . . . . . . . . . . 139

5.1.3 Nifedipine, NI . . . . . . . . . . . . . . . . . . . . . . . . . 141

5.1.4 Ryanodine, Ry . . . . . . . . . . . . . . . . . . . . . . . . 143

5.2 Properties of contractile force in myometrial cells . . . . . . . . . 145

5.2.1 Carbachol, CCh . . . . . . . . . . . . . . . . . . . . . . . . 147

5.2.2 Cyclopiazonic acid, CPA . . . . . . . . . . . . . . . . . . . 149

5.2.3 Nifedipine, NI . . . . . . . . . . . . . . . . . . . . . . . . . 151

5.2.4 Wortmannin, W . . . . . . . . . . . . . . . . . . . . . . . . 151

5.2.5 Calcium-free solution . . . . . . . . . . . . . . . . . . . . . 154

6 Sensitivity analysis 156

6.1 Cell area to volume ratio . . . . . . . . . . . . . . . . . . . . . . . 157

6.2 Length of the myometrial cell . . . . . . . . . . . . . . . . . . . . 158

6.3 Current density of ICa−tot . . . . . . . . . . . . . . . . . . . . . . 161

6.4 Current density of INa−tot . . . . . . . . . . . . . . . . . . . . . . 163

6.5 Current density of IK−tot . . . . . . . . . . . . . . . . . . . . . . . 165

6.6 Myosin light chain kinase . . . . . . . . . . . . . . . . . . . . . . . 169

7 1-D strand model in myometrium 171

7.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171

7.2 1-D strand model development . . . . . . . . . . . . . . . . . . . . 172

7.3 Results . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174

4

8 Discussion 178

8.1 Whole cell membrane, SR currents model . . . . . . . . . . . . . . 178

8.2 Electrophysiological responses . . . . . . . . . . . . . . . . . . . . 180

8.2.1 Cell dynamics in control condition . . . . . . . . . . . . . . 180

8.2.1.1 Repetitive depolarisation . . . . . . . . . . . . . . 181

8.2.2 Cell dynamics under estradiol condition . . . . . . . . . . . 181

8.2.3 Inhibiting individual current . . . . . . . . . . . . . . . . . 182

8.3 Cell mechanics and force generation . . . . . . . . . . . . . . . . . 183

8.4 Drug actions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184

8.5 Sensitivity Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . 186

8.6 1-D strand . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188

8.7 Model limitations . . . . . . . . . . . . . . . . . . . . . . . . . . . 189

8.7.1 Mixing species and tissues data . . . . . . . . . . . . . . . 189

8.7.2 Stretch-induced effects on myometrium . . . . . . . . . . . 189

8.7.3 Spatial heterogeneity . . . . . . . . . . . . . . . . . . . . . 190

8.7.4 Addition force generation pathways . . . . . . . . . . . . . 190

8.7.5 1-D strand . . . . . . . . . . . . . . . . . . . . . . . . . . . 191

9 Future plan 192

9.1 Single cell level . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192

9.2 Tissue level . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193

Bibliography 194

A Single cell uterine model 220

A.1 Model Equations – Membrane currents and Sarcoplasmic reticulum

control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220

A.2 Model Equations – Kinetic cross-bridge model and force generation 234

A.3 Parameter values for ionic currents . . . . . . . . . . . . . . . . . 236

A.4 Parameter values for force conctracion and 1-D strand model . . . 239

A.5 Other parameter values . . . . . . . . . . . . . . . . . . . . . . . . 241

A.6 Initial values . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242

5

List of Tables

1.1 Dimensional changes of the human uterus during pregnancy. . . . 16

1.2 Morphometric parameters and fibre dimensions of rat and rabbit

uterine cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

1.3 Ionic concentration inside and outside myometrial cells . . . . . . 27

1.4 Selected ionic channels expressed in myometrium with their prop-

erties. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32

1.5 [ Ca2+ ]i transient in pregnant human and rat myometrium . . . . 33

1.6 A structural and functional comparison of skeletal, cardiac and

smooth muscle in human. . . . . . . . . . . . . . . . . . . . . . . 39

2.1 Parameter values of Ih . . . . . . . . . . . . . . . . . . . . . . . . 51

2.2 Parameter values of ICaL . . . . . . . . . . . . . . . . . . . . . . . 54

2.3 Parameter values of ICaT . . . . . . . . . . . . . . . . . . . . . . . 57

2.4 The comparison of electrophysiological parameters between rat and

human myometrium for INa . . . . . . . . . . . . . . . . . . . . . . 59

2.5 Parameter values of INa . . . . . . . . . . . . . . . . . . . . . . . 60

2.6 Parameter values of ISOC . . . . . . . . . . . . . . . . . . . . . . 62

2.7 Parameter values of IK1 . . . . . . . . . . . . . . . . . . . . . . . 65

2.8 Parameter values of IK2 . . . . . . . . . . . . . . . . . . . . . . . 68

2.9 Parameter values of IKA . . . . . . . . . . . . . . . . . . . . . . . 71

2.10 Parameter values of IKCa . . . . . . . . . . . . . . . . . . . . . . . 74

2.11 Parameter values of IKleak . . . . . . . . . . . . . . . . . . . . . . 75

2.12 Parameter values of INSCC . . . . . . . . . . . . . . . . . . . . . . 78

2.13 Parameter values of ICl . . . . . . . . . . . . . . . . . . . . . . . 81

2.14 Characteristics of the major myometrial currents. . . . . . . . . . 83

2.15 Parameter values of ICapump . . . . . . . . . . . . . . . . . . . . . 84

2.16 Parameter values of INaK . . . . . . . . . . . . . . . . . . . . . . 85

2.17 Parameter values of INaCa . . . . . . . . . . . . . . . . . . . . . . 87

6

2.18 Parameter values of INaKCl . . . . . . . . . . . . . . . . . . . . . . 89

2.19 Parameter values of Iup , Itr , Irel . . . . . . . . . . . . . . . . . . 91

2.20 Parameter values of ryanodine receptor . . . . . . . . . . . . . . . 92

2.21 Parameter values of the ionic balances . . . . . . . . . . . . . . . 93

2.22 Parameter values of the kinetic cross-brodge cycling model . . . . 98

2.23 Parameter values of the mechanical model . . . . . . . . . . . . . 105

3.1 Changes made to stimulate estradiol effect. . . . . . . . . . . . . . 114

3.2 Parameters changed in producing various types of AP. . . . . . . 127

6.1 Values of peak [ Ca2+ ]i and total force, time to reach the peak of

[ Ca2+ ]i and total force and [ Ca2+ ]i and force in 3 s produced by

the model in control condition. . . . . . . . . . . . . . . . . . . . 157

6.2 Numerical changes of [ Ca2+ ]i transient and total force with chang-

ing cell area to volume ratio in the sensitivity analysis. . . . . . . 160

6.3 Numerical changes of total force with changing lengths of a my-

ometrial cell. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161

6.4 Numerical changes of both [ Ca2+ ]i transient and total force with

changing values of IK−tot . . . . . . . . . . . . . . . . . . . . . . . 163

6.5 Numerical changes of both [ Ca2+ ]i transient and total force with

changing values of INa−tot . . . . . . . . . . . . . . . . . . . . . . . 165

6.6 Numerical changes of [ Ca2+ ]i transient and total force with various

values of INa−tot . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167

6.7 Numerical changes of total force with changing level of MLCK in

the sensitivity analysis. . . . . . . . . . . . . . . . . . . . . . . . . 169

7.1 Parameter values of 1-D strand. . . . . . . . . . . . . . . . . . . . 174

7.2 Comparison of conduction velocities in rat and human from various

gestation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177

A.1 Parameter values on ionic currents . . . . . . . . . . . . . . . . . 238

A.2 Parameter values on force contraction and 1-D strand model. . . . 240

A.3 Other parameter used in the model . . . . . . . . . . . . . . . . . 241

A.4 Initial condition values . . . . . . . . . . . . . . . . . . . . . . . . 243

7

List of Figures

1.1 Freshly dissociated myocyctes from rat. . . . . . . . . . . . . . . . 17

1.2 Micro-anatomy of pregnant human myometrium and human fundal

myometrium at term. . . . . . . . . . . . . . . . . . . . . . . . . . 19

1.3 Two types of AP found in pregnant rat circular myometrium. . . 20

1.4 Membrane potentials and contractions of circular and longitudinal

cells in rat myometrium. . . . . . . . . . . . . . . . . . . . . . . . 22

1.5 Spontaneous mechanical and electrical activity of longitudinal and

circular cells in rat and human pregnant myometrium. . . . . . . 23

1.6 An effect of relaxin in longitudinal rat myometrial tissues. . . . . 24

1.7 A schematic diagram of the imbalance distribution of ions inside

and outside the cell. . . . . . . . . . . . . . . . . . . . . . . . . . 27

1.8 An illustration of a typical myometrial cell with the major ionic

currents pumps and exchangers. . . . . . . . . . . . . . . . . . . . 28

1.9 An illustration of a generic voltage-dependent channel (VOC). . . 30

1.10 Examples of ionic pumps and exchangers across a cellular membrane. 33

1.11 Representation of myosin and force generated by actin and myosin

interaction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35

1.12 A flow diagram of intracellular excitation-contraction coupling in

myometrium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36

1.13 Schematic diagrams of skeletal and smooth muscles showing their

differences in structural properties. . . . . . . . . . . . . . . . . . 38

1.14 A general whole-cell voltage clamp configuration in experiments. . 40

2.1 A block diagram of the myometrial cell model with the two coupled

subsystems. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43

2.2 The equivalent electrical circuit of a cell. . . . . . . . . . . . . . . 45

2.3 Schematic cell diagram for the myometrial model. . . . . . . . . . 49

2.4 Model of myometrial Ih . . . . . . . . . . . . . . . . . . . . . . . . 51

8

2.5 Model of myometrial ICaL . . . . . . . . . . . . . . . . . . . . . . . 55

2.6 Model of myometrial ICaT . . . . . . . . . . . . . . . . . . . . . . . 58

2.7 Model of myometrial INa . . . . . . . . . . . . . . . . . . . . . . . 61

2.8 Model of IK1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66

2.9 Myometrial model of IK2 . . . . . . . . . . . . . . . . . . . . . . . 69

2.10 Myometrial current model of IKA . . . . . . . . . . . . . . . . . . . 72

2.11 I-V relationship of IKCa , INSCC and IKleak . . . . . . . . . . . . . . 75

2.12 Myometrial current model of ICl . . . . . . . . . . . . . . . . . . . 82

2.13 Four-state kinetic model of RyR. . . . . . . . . . . . . . . . . . . 90

2.14 Two pathways leading to mechanical contraction in myometrium. 95

2.15 Four-state kinetic model of cross-bridge cycling in smooth muscle. 96

2.16 The mechanical model of a smooth muscle cell. . . . . . . . . . . 100

2.17 The contractile component describing the actin and myosin fila-

ment sliding action. . . . . . . . . . . . . . . . . . . . . . . . . . . 102

3.1 AP and [ Ca2+ ]i in control condition. . . . . . . . . . . . . . . . . 109

3.2 I-V and current traces of the whole cell in control condition. . . . 110

3.3 [ Ca2+ ]i and ICa−tot responses during repetitive depolarisation. . . 111

3.4 AP and [ Ca2+ ]i under effect of estradiol. . . . . . . . . . . . . . 113

3.5 I-V of and current traces for the whole cell under the effect of

estradiol. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

3.6 Behaviour of all Ca2+ -related currents during current-clamp. . . . 117

3.7 Behaviour of all Na+ -related currents during current-clamp. . . . 118

3.8 Behaviour of all K+ -related currents during current-clamp. . . . . 120

3.9 Behaviour of other currents during current-clamp. . . . . . . . . . 121

3.10 Effects of inhibiting the inward currents. . . . . . . . . . . . . . . 123

3.11 Effects of inhibiting the outward currents. . . . . . . . . . . . . . 124

3.12 Effects of inhibiting other currents. . . . . . . . . . . . . . . . . . 126

3.13 Other possible types of APs produced by the model. . . . . . . . . 128

4.1 Stress development in a single myometrial cell. . . . . . . . . . . . 131

4.2 Twin-pulse simulation in myometrium compared to experimental

recordings. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133

4.3 Ca2+ spikes and force production in a single myometrial cell with

single- and multiple-pulse stimulations. . . . . . . . . . . . . . . . 134

9

4.4 [ Ca2+ ]i transient, active force, Fa and the individual length com-

ponents in the myometrial cell. . . . . . . . . . . . . . . . . . . . 136

4.5 Comparison of [ Ca2+ ]i transient, total force and peak force rate. . 137

5.1 The effect of depolarisation with different magnitudes on [ Ca2+ ]i

transient and ICa−tot . . . . . . . . . . . . . . . . . . . . . . . . . . 140

5.2 The effect of carboxyeosin on [ Ca2+ ]i transient. . . . . . . . . . . 142

5.3 Effects of nifedipine on [ Ca2+ ]i transient. . . . . . . . . . . . . . 144

5.4 The effect of ryanodine on [ Ca2+ ]i and ICa−tot in myometrium. . 146

5.5 The effects of carbachol on contractile force in myometrium. . . . 148

5.6 The activity of contraction and [ Ca2+ ]i with presence of CPA. . . 150

5.7 The effects of nifedipine on [ Ca2+ ]i and contractile force in my-

ometrium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152

5.8 The effects of wortmannin on total force, [ Ca2+ ]i and ICa−tot in

myometrium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153

5.9 The effects of zero external calcium on contractile force and [ Ca2+ ]i

in myometrium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155

6.1 The effects of changing values of cell area to volume ratio on the

myometrial model. . . . . . . . . . . . . . . . . . . . . . . . . . . 159

6.2 The effects of changing values of cell length on the myometrial model.162

6.3 The effects of changing the current density of ICa−tot in [ Ca2+ ]i transient

and the total force. . . . . . . . . . . . . . . . . . . . . . . . . . . 164

6.4 The effects of changing the current density of INa−tot in [ Ca2+ ]i transient

and the total force. . . . . . . . . . . . . . . . . . . . . . . . . . . 166

6.5 The myometrial model’s physiological response to different values

of IK−tot . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168

6.6 The effects of various levels of MLCK on the total force. . . . . . 170

7.1 A chain of myometrial smooth muscles demonstrates parameters

in the 1-D strand model. . . . . . . . . . . . . . . . . . . . . . . . 173

7.2 AP conduction in the 1-D myometrial strand model. . . . . . . . . 176

10

Abstract

While premature birth is still one of the major clinical problems worldwide, the

exact physiological mechanisms underlying myometrium activity during preg-

nancy remain unclear. In this thesis, a novel biophysically detailed model was

constructed using available experimental data to simulate chemical, electrical and

mechanical activity in a late pregnant rat uterine myocycte. The developed model

has been used to elucidate the ionic mechanism underlying myometrium function-

ality, providing better insights in the function of the uterus during pregnancy. The

model consisted of 15 membrane currents, intracellular calcium handling process

coupled with a sliding actin-myosin filament mechanical model to describe uter-

ine behaviour and contractile activity at the single myocyte level. Each of the

ionic currents were modelled using Hodgkin-Huxley-type equations. The simu-

lated current traces and current-voltage curves were validated with experimental

recordings and the model was further validated by the ability to produce a burst-

ing action potential (AP) during an external stimulus. The model replicated the

effects of estradiol during pregnancy, modulating the amplitude and activation

properties of individual Ca2+ and K+ currents, therefore altering the AP config-

uration to a tonic-like plateau. The model also reproduced the actions of drugs

to inhibit certain channels to investigate their roles in myometrium. Sensitivity

analysis was performed to examine the model’s behaviour to changing parameters.

A simple 1-D study was conducted to investigate how electrical signals propagate

along strand of cells. Although the model successfully replicated results similar

to recordings seen in the experiments, limitations have to be addressed and more

studies have to be carried out to further improve the model.

11

Declaration

No portion of the work referred to in this thesis has been

submitted in support of an application for another degree

or qualification of this or any other university or other

institute of learning.

12

Copyright

i. The author of this thesis (including any appendices and/or schedules to this

thesis) owns any copyright in it (the “Copyright”) and s/he has given The

University of Manchester the right to use such Copyright for any adminis-

trative, promotional, educational and/or teaching purposes.

ii. Copies of this thesis, either in full or in extracts, may be made only in

accordance with the regulations of the John Rylands University Library of

Manchester. Details of these regulations may be obtained from the Librar-

ian. This page must form part of any such copies made.

iii. The ownership of any patents, designs, trade marks and any and all other

intellectual property rights except for the Copyright (the “Intellectual Prop-

erty Rights”) and any reproductions of copyright works, for example graphs

and tables (“Reproductions”), which may be described in this thesis, may

not be owned by the author and may be owned by third parties. Such Intel-

lectual Property Rights and Reproductions cannot and must not be made

available for use without the prior written permission of the owner(s) of the

relevant Intellectual Property Rights and/or Reproductions.

iv. Further information on the conditions under which disclosure, publication

and exploitation of this thesis, the Copyright and any Intellectual Property

Rights and/or Reproductions described in it may take place is available

from the Head of School of Physics and Astronomy (or the Vice-President).

13

Acknowledgements

I would like to thank my grandad who always inspired and encouraged me to

do great things and never stopped believing in me. I am forever indebted to my

grandma who raised me and supported me throughout everything.

I greatly appreciate and sincerely thank my supervisor Prof. Henggui Zhang

for giving me the opportunity to participate in this exciting project, and Dr.

Winnie Tong who helped and guided me during my study.

I am extremely grateful to my best friends Chris, Sarah, Penny and Judy who

have always been there when I needed them despite us living 6000 miles apart.

Finally I’d like to thank Philip Stewart who I met the first day of my Ph.D

and is now my husband. He helped me through my difficult times and put up

with my tantrums without ever complaining. This thesis would not have been

possible without his love and continuous support.

14

Chapter 1

Introduction

The control of myometrium excitability is vital during pregnancy. It is a highly

complex phenomenon which involves hormonal, structural, electrical and contrac-

tility changes throughout the gestation period. The activity of the myometrium

is relatively quiescent during early and mid pregnancy to accommodate develop-

ment and growth of the foetus. The contractions of the myometrium remains weak

and unsynchronised, however the contractions progressively increase in strength

towards the end of pregnancy. During labour, strong, powerful contractions occur

to dilate the cervix and expel the foetus and placenta [1]. Therefore, a successful

delivery almost entirely depends on the control of the myometrium towards the

latter stages of gestation. If these functions are not performed perfectly, it can

lead to important therapeutic implications since myometrial dysfunction is be-

lieved to be a cause of pre-term labour or dystocica [2], and premature labour is a

major cause of mortality, neonatal handicap and morbidity. It is a serious health

and financial burden to families and society. Despite extensive studies and ad-

vances in medical care developed in recent years, pre-term delivery has remained

constant at around 6-10 % of total live births in the UK [3, 4]. Myometrium

contraction failure occurs in 4-8 % of all labour and can lead to emergency cae-

sarean section which puts both the mother and the baby at risk [5]. It has

been estimated that neonatal intensive care for babies weighing less than 1500 g

costs the NHS between £42 and £72 million every year [2]. Understanding the

mechanisms and fundamental processes underlying myometrial function can help

to prevent or treat uterine dysfunction, therefore benefiting women, babies and

families worldwide.

15

1.1 Structure of the uterus

Mammalian uteri undergo enormous changes during and also after pregnancy.

The weight and the capacity of the organ can increase more than 10 and 500

fold, respectively at term [6]. Uterine cells hypertrophies during pregnancy to

adapt to the growth of the foetus and reduce back to their original size after

parturition. The details of the uterine changes occurring before the pregnancy

and at term in human are listed in table 1.1. The volume of a pregnant human

uterus at term is about 4000 to 4500 cm3. It is an elongated ellipsoid shape with

vertical and horizontal diameters of approximately 38 cm and 14 cm, respectively.

The surface area of the whole organ is about 11788 cm2 at term. However, when

the infant has descended only 2 cm, the surface area of the uterus decreases to

9330 cm2 (80 % of term value). The surface area of the organ massively decreases

to only 8000 cm2 (68 % of term value) immediately following delivery [7].

Gestation Weight Dimensions∗ Capacity Total intrauterine(g) (cm) (mL) volume (cm3)

Non-pregnant 50-70 7.5×5×2.5 10 <300 in earlypregnancy

At term 80-1200 20×25×22.5 5000 4500

Table 1.1: Dimensional changes of the human uterus during pregnancy.The uterus expands tremendously during pregnancy. The capacity of the organcan increase 500 times at term. ∗ Dimension represents: length x width x depth.The table was adopted from [6].

Although uterine cells share many similarities with other smooth muscle cells

such as gastro-intestinal, urethra and urinary bladder cells, etc., myometrial cells

are still very unique in many ways. The uterus is a hollow organ that is composed

of connective tissue, blood vessels, nerves and predominately, smooth muscle,

or myocytes. Myocyctes are interwoven in the uterus and form three distinct

layers: 1) an external layer composed of two sub-layers: circular muscle fibres

on the outside and longitudinal fibres inside; 2) an intermediate layer in which

the muscle fibres have a mixture of orientations; and 3) a non-excitable internal

layer of circular muscle fibres. The endometrium is rich in cytokines, hormones

and peptides that influence the contractile activity in pregnant or non-pregnant

myometrium [2]. Typical longitudinal uterine myocyctes found in pregnant and

non-pregnant rat are shown in figure 1.1.

16

Figure 1.1: Freshly dissociated myocyctes from rat. All images areadapted from Yoshino et al. [8]. Scale bar in (A) represents 40 µm; and 10µm in all other frames. (A) Late pregnant uterine myocycte in low magnifi-cation showing typical cell density. (B and C) Two typical cells from a latepregnant (18-21-d) uterus. (D) Myocyte from a non-pregnant estrus uterus (Thethickened part of the endometrium. During the period in the sexual cycle whenfemale mammals are in heat, the lining of the uterus thickens when they areready to accept a male to mate). (E) A mid-pregnant (14-d) uterine myocycte.(F to H) Myocytes showing marked pleomorphism (irregular and variant forms).These cells are most frequently encountered in late pregnancy, where they maycomprise 10% of the population (see A). These cells have complex geometriestherefore may pose problems for uniform voltage control and they are usuallyexcluded from experiments [8].

17

Action potentials (APs) are initiated in, and propagate rapidly from, the

fundus (top portion) of the uterus. The speed of AP propagation in myometrium

is about 13.5 cm/s in rat [9], 6.8 cm/s and 2.8 cm/s for longitudinal and circular

cells in pregnant guinea pig [10], 9.2 cm/s in late pregnant human cells and

10.5 cm/s at term [11]. In order to allow APs to propagate, micro-anatomy

communicates electrically throughout the whole organ along bundles of fibres.

An illustration of the micro-anatomy of a pregnant myometrium and a section of

fundus specimen obtained from a patient at term pregnancy is shown in figure

1.2.

1.2 Electrical activity and action potential

The uterine myocyctes are myogenic in nature - a single isolated pregnant or

non-pregnant uterus smooth muscle can produce spontaneous contraction with-

out external nervous or hormonal input. When an AP is initiated, a rhythmical

and regenerative AP is propagated along the wall of myometrium, known as a

slow wave, to trigger contractions. At present, the pacemaking cells in human my-

ometrium are unknown. Unlike cardiac muscle, pacemaker cells in myometrium

is not anatomically fixed or defined. It is still unclear whether all cells in the

myometrium have pacemaking ability or only certain types of cells within the

myometrium are responsible [13]. In some visceral smooth muscle, e.g. gastro-

intestinal, urethra and urinary bladder, interstitial cells of Cajal (ICC) have been

found and these cells constitute a pacemaking mechanism [14]. Similar cell types

are found in human and rat myometrium but these cells are unlikely to play a

part in pacemaking activity [15,16].

There are two main forms of APs with variable characteristics recorded with

different gestation in different regions of the uterus, 1) simple spikes, and 2)

plateau type shown in figure 1.3. The pattern of depolarisation leading to an AP

also changes from small, irregular spikes at early to mid pregnancy, to regular

sustained activity, in some cases, accompanied by a prolonged plateau phase [17].

The number of spikes in the AP, together with the number of cells involved,

determines the force of contraction, while the duration of the train of APs or the

depolarisation phase, corresponds to the period of the contraction.

There are also significant physiological differences between longitudinal and

circular cells, with regards to their changes in gestation, observed in uterine

18

Figure 1.2: Micro-anatomy of pregnant human myometrium and hu-man fundal myometrium at term. (A) Micro-anatomy of pregnant humanmyometrium. Myocyctes are densely packed together to form cylindric bundles,sheet-like bundles and communicating bundles, in turn organised into fascicu-lata. Fasciculi make up the majority of the wall of the uterus. (B) Human fundalmyometrium at term. The outer layer of the uterus can be seen with packedmyocycytes and is about 250 µm thick. The transitional layer is interwoven withbundles spanning from the outer layer to the fasciculata and is about 0.5 to 1mm thick. Images are adapted from Young et al. 1993 and 2000 [9, 12].

19

Figure 1.3: Two types of AP found in pregnant rat circular my-ometrium. During early to mid-pregnancy, circular myometrium display a singleplateau-type AP. The shape of the AP then changes to a repetitive, spike-like APat late pregnancy to term. Image adapted from Wilde and Marshall [18].

20

myocytes. In a study conducted by Bengsston et al. [19] using pregnant rat

myometrial cells, it was reported that in rat circular myocytes, single plateau-

type APs triggered small irregular contractions in 16-day pregnant cells (figure

1.4). Contractions become stronger and more regular with a longer duration and

repetitive spike trains at term. However in longitudinal myocytes, only repetitive

spike trains and strong contractions were recorded in 16-day gestation and at

term. It is clearly evident that the magnitudes of the contractions change in both

myocytes throughout the pregnancy [17].

The spontaneous electrical activity recordings show similar patterns of AP

in the same cell types (circular and longitudinal) even from different species.

Figure 1.5 shows the spontaneous AP and force production of both circular and

longitudinal in rat and human pregnant myometrial cells. It is clearly evident

that longitudinal cells in rat and human exhibit spike-like APs, while circular

cells exhibits a plateau-like AP in both species [11]. In the spike-like AP, each

spike triggers a small rise in tension which does not immediately decline to its

resting level, instead the tension accumulates and form a tetanus-like profile if the

spike intervals are small. Although the morphology and characteristics of both

human and rat spontaneous AP are very similar, the time courses are markedly

different with APs lasting about 60s in human but only 10s in rat.

A study carried out by Chamley and Parkington [20] compared the effect of

relaxin on the resting membrane potential, electrical and mechanical properties

of the longitudinal and circular in pregnant rat tissue shown in figure 1.6. Re-

laxin causes the release of noradrenaline, which is responsible for relaxations of

contraction in myometrium [21, 22]. In control condition, the resting membrane

potential was 62 ± 1 and 53 ± 1 mV in 15-17 day pregnant longitudinal and

circular rat tissues respectively. When relaxin was applied, little or no effect was

recorded on the resting membrane potential. However, the number of spikes in

a burst decreased dramatically but the amplitude of the AP remained the same

in longitudinal tissues. The results were changed when samples from 18-day to

term tissues was used. The number of spikes still decreased but after about 6-10

mins, the activity resumed despite the presence of the fresh relaxin.

In human myometrium, the separations between different types of electrical

patterns from different cells are more complex and often difficult to observe.

The action potentials found in human tend to have simple spikes followed by a

sustained depolarising plateau. This type of AP is found in human myometrium

21

Figure 1.4: Membrane potentials and contractions of circular and lon-gitudinal cells in rat myometrium. Membrane potentials (Lower traces) andcontractions (Upper traces) of circular (CIRC) and longitudinal (LONG) uterinesmooth muscle cells at 16-day pregnant and during delivery (DEL) on day 21.There is a clear difference of AP profile between circular and longitudinal cells inday 16 of the pregnancy. The difference becomes less apparent during deliveryapart from the magnitude of the AP. The AP from circular cells has a lower rest-ing membrane potential and a lower peak level of AP compared to longitudinalcells during delivery. The tension generated from longitudinal cells is about twiceas high as the circular cells. The contractions in both cells becomes more syn-chronised towards the end of the pregnancy. Also the magnitude and frequencyof the force severely increased during delivery in both cell types. Images wereadapted from Bengtsson et al. [19].

22

Figure 1.5: Spontaneous mechanical and electrical activity of longitu-dinal and circular cells in rat and human pregnant myometrium. Thereis a significant difference in the time course of AP between human and rat despitehaving similar shape and characteristics in both AP and force generation profile.Each contraction is well synchronised with the AP in both species. Note in ratcircular cells, the AP and contraction profiles are overlapped. AP is labelledwith the unit mV while force is labelled with the unit g. Figure adapted fromKawarbayashi [11].

23

Figure 1.6: An effect of relaxin in longitudinal rat myometrial tissues.(A) The AP, mechanical force profile (top and middle traces respectively) evokedby a 1.2 s depolarising current pulse (bottom trace) in control condition. It isclearly shown the duration of the contraction depends entirely on the durationof the bursting AP. (B) Introducing relaxin caused the late pregnant myometrialtissues to reduce the number of spikes in AP (top trace) and the magnitudes ofthe contraction (middle trace) even with various current pulses (bottom trace).However, the electrical and mechanical activities resumed after 8-15 mins. (C)The application of the same quantity of relaxin also initially reduced the numberof spikes in burst of AP from rat myometrium at term (top trace). The numberof spikes gradually increase and after a shorter time of 6-10 mins, Figure adaptedfrom Chamley and Parkington [20].

24

at term. Surprisingly, this does not occur in tissue from every woman. It also

does not seem to have any correlation with a particular region of the uterus where

the tissue samples were taken from, nor the gestation of the pregnancy [23]. The

morphometric parameters of the human, rat and rabbit in non-pregnant, pregnant

and estrodiol-treated myometrial cells are listed in table 1.2.

1.3 Cell membrane properties and ionic currents

The cell membrane is semi-permeable and selectively controls various ions move-

ment across the cell. The permeability of the cell depends on the ionic concentra-

tion gradients inside versus outside the cell as illustrated in figure 1.7. There are

unequal distribution of ions which leaves a net negative charge inside the cell. A

negative intracellular environment relative to the cell exterior creates a potential

difference across the cell membrane. Ions travel across the cell membrane through

ionic channels that have different regulated properties and selectivities. The di-

rection of the flux for a given ion is determined by the ionic equilibrium potential,

which is set by the concentration gradient for the particular ion and membrane

potential difference. If the membrane potential is more positive than the equilib-

rium potential, there will be an efflux of that ion from the cell and vice versa [23].

An estimated ionic concentration inside and outside a myometrial cell is listed in

table 1.3. In human, the resting membrane potential is around -40 to -70 mV in

pregnant myometrium. It becomes more negative, around -60 mV, during mid-

pregnancy and increases to a more positive potential of -45 mV near term. The

precise mechanisms causing these changes are not clearly understood. The resting

membrane potential is predominantly determined by the relative concentrations

of Na+ , K+ and Cl− , suggesting these ionic channels must have altered in some

way as pregnancy progresses. A simplified schematic cell diagram with selected

the ionic currents with directions of current flux is shown in figure 1.8.

When an AP is initiated in a mammalian myometrium, the cell membrane

depolarises and activates the L-type calcium channel current, ICaL at around -40

mV. This creates an influx of Ca2+ towards the inside of the cell and ultimately

leads to contraction. T-type calcium channel current, ICaT , on the other hand

activates at a more negative potentials (around -60 mV). This channel, which has

been shown to be present in human but not in rat myometrium, is associated with

AP transmission and pacemaker activity [1]. Hyperpolarising current, Ih is also

25

Anim

alan

dM

axim

um

dia

met

erA

vera

gedia

met

erLen

gth

Surf

ace

area

Mea

nce

llC

yto

pla

smic

volu

me

gest

atio

n(µ

m)

(µm

)(µ

m)

(µm

2)

volu

me

(µm

3)

Non

-pre

gnan

t(h

um

an)

N/A

550

N/A

N/A

N/A

At

term

N/A

1550

0N

/AN

/AN

/A(h

um

an)

Non

-pre

gnan

t(r

at)

7.9±

0.4

4.5±

0.2

129.

11.7

928.

235.

12.

0.4

pl

N/A

Mid

-pre

gnan

t(r

at)

15.2±0

.47.

5±0.

223

8.3

5193±

188.

411

.0±

0.5

pl

N/A

Lat

epre

gnan

t(r

at)

21.0±0

.710

.6±

0.5

255.

12.6

7599

.4±6

39.7

21.0±

2.5

pl

N/A

Non

-pre

gnan

t(r

abbit

)N

/A5.

56±

1.46

3523

.281

1µm

368

5

Est

radio

l-tr

eate

d(r

ab-

bit

)

N/A

7.92±

1.46

5246

.829

9629

12

Table

1.2

:M

orp

hom

etr

icpara

mete

rsand

fibre

dim

ensi

onsofra

tand

rabbit

ute

rine

cells.

Info

rmat

ion

was

bas

edon

repor

ted

exper

imen

taldat

afr

omB

lack

burn

etal

.[6

]in

hum

an,Y

oshin

oet

al.[8

]in

ratan

dG

illo

teau

xan

dSzc

zepan

ski[2

4]in

rabbit

myo

met

rium

.W

hen

par

amet

ers

wer

enot

repor

ted,N

/Aw

asnot

edin

the

cate

gory

.

26

Ions Outside Inside Ratio :(mmol/L) (mmol/L) Outside/Inside

Na+ 137 40 3.4Ca2+ 1.5 1.3×10−4 1×104

K+ 6 169 0.03Cl− 134 65 2.1

Table 1.3: Estimated ionic concentration inside and outside myometrialcells. Mean values of the ionic concentrations were obtained from rat, cat, guineapigs [25]. Table as described from Sanborn [23].

Figure 1.7: A schematic diagram of the imbalance distribution of ionsinside and outside the cell. Extracellular and intracellular ions are separatedby the semi-permeable cell membrane. Ions in larger fonts indicate a higherconcentrations and smaller fonts with lower concentrations. Since the majorityof the positive ions exist outside the cell, this results in the relatively negativepotential in cell interior [17].

27

Figure 1.8: An illustration of a typical myometrial cell with the majorionic currents pumps and exchangers. All the currents are shown with direc-tions of current flux leading to a control of [ Ca2+ ]i concentration in a simplifiedCa2+ store. The inward myometrial currents include the hyperpolarising current( Ih ), sodium current ( INa ), L-type calcium current ( ICaL ), T-type calcium cur-rent ( ICaT ) and store-operated cation current ( ISOC ). The outward currents arethe two delayed rectifying potassium currents ( IK1 and IK2 ), A-type potassiumcurrent ( IKA ) and calcium-activated potassium current ( IKCa ). Other currentswith both direction of flows are the calcium-activated chloride current ( ICl ) andnon-specific cation current ( INSCC ). The Na+ /Ca2+ exchanger, Na+ /K+ andCa2+ pump are also presented to maintain the ionic balance inside and outsidethe cell. Note this cell diagram is only a simplified illustration of a uterine celland does not represent the cell diagram for the myometrial model. More detailedschematic diagram for the myometrial model is shown in figure 2.3.

28

suggested to contribute to spontaneous pacemaking activity since this channel is

activated by hyperpolarisation at resting membrane potential [26]. It is also found

in cardiac cells, although the density of this current is much lower than pregnant

rat uterine cell. Another inward current identified in pregnant rat myometrium is

TTX-sensitive sodium current, INa [27]. This fast sodium channel current, INa is

not normally found in smooth muscle cells (but present in cardiac cells), but it

has been identified in the rat pregnant uterus. The role of this current includes

spreading the excitation and increasing spike frequency in myometrium. The

store-operated non-selective cation, ISOC is also identified in myometrium and it

is permeable to different types of ions [28], however no quantitative measurement

was recorded in experiments.

There are various types of outward potassium channel expressed in myometrium

with diverse properties and characteristics. Published data often uses various

names and formalisms for a particular ion channel according to different authors.

Therefore the definitions of different types of potassium channels are often very

poorly differentiated. In this model, definitions of different channels are classi-

fied according to the work by Khan and Knock et al. [2, 29, 30], which distin-

guish these channels by their inactivation dynamics and activators. This current

is sub-categorised into two delayed rectifying potassium currents, IK1 and IK2 ,

A-type potassium current, IKA and finally calcium-activated potassium current,

IKCa . Their main roles are relaxing the excitation cycle and restoring the resting

membrane potential following an AP. The calcium-activated potassium channel

exhibits a loss of sensitivity towards late pregnancy to increase excitation in

preparation of labour [29,31].

Other important currents identified in myometrium include the calcium-activated

chloride current, ICl [32] and the non-specific cation current, INSCC [33]. Acti-

vation of chloride channel leads to depolarisation and is therefore important in

governing excitability. The non-specific cation channel is permeable to a variety

of mono and divalent ions such as K+ , Cs+ , Na+ and Li+ and it is believed

that this channel is responsible for the pace-making activity and regulating the

myometrium contractility. Detailed descriptions of each ion channels, exchangers

and pump are given in section 2.

29

1.4 Ionic channels in myometrium

Ion channels are complex proteins which act as a barrier to control particular

ions entering or leaving the cell. Different ionic channels can be modulated by

different activators, hormonal state and gestation in pregnancy. Ionic movement

across the cell can also be controlled by blocking and opening the channels by

special activating and inhibiting agents. These agents change the conformation

of the ionic channel gating mechanisms and the gating factors are classified into

three main species (these groups are not mutually exclusive):

1. Voltage-operated channels (VOCs) - the channels have voltage sensors,

charges residues shift the position within the protein depending on the mem-

brane potential of the cell. An illustration of a generic voltage-dependent

channel is shown in figure 1.9.

2. Receptor-operated channels (ROCs) or ligand-gated channels - opening or

closing the channels depends on the binding of the extracellular factor, such

as a hormone or a neutrotransmitter.

3. Second-messenger-operated channels (SMOCs) - channels opening or clos-

ing are dependent to an intracellular factors, such as calcium ions or acti-

vated G protein subunits [34].

Figure 1.9: An illustration of a generic voltage-dependent channel(VOC). Plasma membrane is formed by phydriphilic heads and hydrophilic tailsand they are arranged in a bi-layer arrangement in a watery environment. Imageadapted from Molleman [34].

30

Different major ionic membrane channels identified in myometrium and their

general properties including their opening/blocking agents are listed in table 1.4.

1.5 Ionic pumps and exchanger

Apart from ionic channels, ionic pumps and exchangers across the cell membrane

also play an important part in maintaining the ion gradients and membrane

potential. There are extensive studies of ionic pumps and exchangers in cardiac

cell but detailed qualitative information in myometrium is lacking. Ionic pumps

transport molecules against their concentration gradient by active transport. The

energy required by this process is provided by the breaking down of ATP to ADP

on the cytosolic side of the membrane. Exchangers (or co-transporter) work

in the similar manner as pumps, but energy is derived from diffusion from the

other molecules. Since K+ has a much higher intracellular concentration than

Na+ , maintenance of this is done by an Na+ /K+ (or Na+ /K+ -ATPase) pump.

Each cycle transfers three sodium ions out of the cell and lets two potassium

ions enter the cell, therefore a net outward flux leaves the cell more negatively

charged. Ca2+ and Na+ concentrations on the other hand are controlled by a

Ca2+ /Na+ exchanger. One calcium ion is pumped out in each cycle and three

sodium ions are transferred into the cell to regulate the ionic gradients across the

myometrial cell.

1.6 Calcium handling and uterine contractility

When a myometrial cell is depolararised, Ca2+ entry into the cell through voltage-

gated channels (mainly L-type calcium channel) is initiated. This increases

the intracellular calcium concentration, [ Ca2+ ]i and allows [ Ca2+ ]i to bind with

calmodulin which activates the myosin light-chain kinase (MLCK), and thus phos-

phorylates a regulatory myosin light chain, consequently forming cross-bridges

between actin and myosin filaments to generate muscle contraction [35, 36]. The

entry current consists of two components which are fast Na+ ion current and

slow Ca2+ current (mainly L-type) [13, 37]. It has been demonstrated that the

level of intracellular concentration rises through Ca2+ channels and Ca2+ induced

Ca2+ released (CICR) from the sarcoplasmic reticulum (SR) when a myometrial

31

Ion

Chan

nel

Act

ivat

ors

Inhib

itor

sC

han

ges

inpre

gnan

cy

I hV

olta

geC

s+

I Na

Vol

tage

Sax

itox

in,te

trodot

oxin

(TT

X)

Incr

ease

snea

rte

rm

I CaL

Vol

tage

Dih

ydro

pyri

din

es(n

ifed

ipin

e),C

a2+

inac

tiva

tion

,M

n2+,M

g2+,

volt

age

inac

tiva

tion

Incr

ease

sin

mid

-pre

gnan

cy

I CaT

Vol

tage

Vol

tage

,m

iber

frad

ilI K

1V

olta

geT

EA

>50

mm

ol/L

I K2

Vol

tage

TE

A>

50m

mol

/L,4A

PI K

AV

olta

ge4-

amin

opyri

din

e(4

AP

)I K

Ca

Cal

cium

,ag

onis

t,re

laxin

Vol

tage

,D

ehydro

-yas

apon

inI,

Nifl

um

icac

id

TE

A,4-

AP,bar

ium

Los

sof

sensi

tivity

wit

hla

bou

r

I Cl

Vol

tage

Nifl

um

icac

idan

dan

thra

cene-

9-ca

rbox

ylic

acid

(9-A

C)

I NSC

CA

TP,H

yper

pol

aris

atio

nLa3

+,G

d3+

Dim

inis

hed

nea

rte

rmw

ith

anin

crea

sing

leve

lof

oes

trog

en

Table

1.4

:Sele

cted

ionic

channels

expre

ssed

inm

yom

etr

ium

wit

hth

eir

pro

pert

ies.

The

acti

vato

r/in

hib

itor

and

the

chan

ges

duri

ng

pre

gnan

cyof

the

chan

nel

sw

ere

also

incl

uded

.In

form

atio

nas

des

crib

edfr

omSan

bor

nan

dm

odifi

edfo

rad

dit

ional

chan

nel

s[1

7].

32

Figure 1.10: Examples of ionic pumps and exchangers across a cellu-lar membrane. Apart from ion channels, various mechanisms exist in cells tomaintain the cellular ionic balance by transporting ions across the cell membrane.These mechanisms include the Ca2+ pump (left), Na+ /K+ pump (middle) andNa+ /calcium transporter (right). Image adapted from Molleman [34].

cell undergoes repetitive stimulation with pulse trains. The decay of Ca2+ con-

centration, which is vital for relaxation of uterine mocyctes, is correlated to the

Ca2+ pump (30%) and Na+ /Ca2+ (60%) of the extraction of calcium from the

cell [38]. The [ Ca2+ ]i transient characteristics in pregnant rat and human my-

ometrium are listed in table 1.5. The [ Ca2+ ]i duration recorded in human is

markedly longer than rat (about 9 times longer) but the resting [ Ca2+ ]i and

peak [ Ca2+ ]i levels are both higher in rat than in human (about twice higher for

both levels).

Animal [ Ca2+ ]i duration Resting level Peak of [ Ca2+ ]i(s) (nM) (nM)

Human 120 94.12 423.3Rat 13-13.3 110-220 800

Table 1.5: [ Ca2+ ]i transient in pregnant human and rat myometrium.Measurements from human were obtained from Parkington et al. [25] and mea-surements from rat were obtained from Shmigol et al. [39].

There are three types of muscle cells existing in mammalian organs and bodies:

skeletal, cardiac and smooth muscle cells. The mechanisms responsible for the

generation of muscle contractions in all three types of muscles are actin and

33

myosin filaments. In myometrium, the myofilament proteins include actin, myosin

and intermediate. The myosin is the principal contractile protein which initiates

the development of tension in the cells. A schematic diagram of a smooth muscle

myosin is illustrated in figure 1.11(A) which shows a myosin filament made up of

two light chains arranged to form one helical chain. The chains are joined at one

end to form two globular heads (myosin heads) which stand out to interact with

the actin filament to generate force. Actin is a long thin filament that is found

between dense bodies. To optimise actin and myosin interaction, myosin is laid

down in thick myofilaments at a resting state as illustrated in figure 1.11(B).

To generate tension in smooth muscle, the rising level of intracellular calcium

binds with calmodulin to form Ca-calmodulin (figure 1.12). This in turns acti-

vates the myosin light chain kinase (MLCK) and phosphorylates light chains on

myosin (P-myosin or P-light chain) as shown in figure 1.11 (B-1). This causes the

binding of actin and activates myosin Mg-ATPase, and initiates the contraction

with the hydrolysis of ATP to ADP [40]. Binding of the myosin with F-actin is

then activated and ATP hydrolysis by ATPase (figure 1.11 (B-2 and 3)). The

myosin head changes conformation when it is bound to actin, it rotates and pull

on the actin filament generating force (power stroke). This movement creates

a relative spatial displacement or shortening and is powered by the release of

ADP when the myosin head changes its conformation (figure 1.11 (B-4)). Myosin

dephosphorylates by phosphatases stopping the binding of the myosin head and

F-actin, leading to relaxation of the cell (figure 1.11 (B5-7)). There are multiple

Ca2+ -regulatory mechanisms required in myometrial contraction, all these mech-

anisms work together to provide the ability of uterine cells to modulate contractile

force with different stimulations. This system is especially beneficial to the con-

trol in myometrium since the contractile performance can be affected by various

elements for example: stretch, hormones and metabolite concentrations [1,41–44].

The internal organisation in skeletal and cardiac muscle cells are very different

from a smooth muscle cell [45]. Actin and myosin proteins are organized in sar-

comeres, while in smooth muscle cells, the myosin filaments are not well ordered.

Smooth muscles cells are also shorter compared to skeletal cells and are grouped

into loose bundles as illustrated in figure 1.13. This arrangement increases the

maximum degree of shortening and generates 5 to 10 times greater shortening

than that produced by skeletal muscles [6]. Adjacent smooth muscle cells are

attached together at dense bodies, and contractile force is transmitted from cell

34

Figure 1.11: Representation of myosin and force generated by actin andmyosin interaction. (A) A schematic diagram of a myosin in smooth musclecells. Helical chains are united to create globular heads which bind with actinto develop tension in the myometrium. (B) (1-2) Myosin and actin filament in aresting state, when [ Ca2+ ]i binds with calmodulin, MLCK is activated and causesthe P-light chain phosphorylation (3) As a consequence, the myosin head binds toF-actin (4) The myosin head changes conformation when it is bound to actin, itrotates and pulls, creating cell shortening and contraction (5) Dephosphorylationof the P-light chain inhibits the binding of the myosin head with F-actin and (6-7) myosin head releases the F-actin and the original conformation of the myosinhead is restored as the cell finally relaxes. Figure adapted from Tucker-Blackburnet al. [6].

35

Figure 1.12: A flow diagram of intracellular excitation-contraction cou-pling in myometrium. Contraction is initiated in myometrium when a rise ofintracellular calcium causes the binding of calcium and calmodulin. This in turnactivates the myosin light chain kinase (MLCK) and the start of the cross-bridgeinteraction between actin and myosin generating tension in myometrium. Relax-ation is achieved when P-myosin is dephosphorylated by phosphatases and alsoby the reduction of intracellular calcium concentration [1].

36

to cell through out the tissue. The dense bodies in smooth muscles have the same

function as Z-disks in skeletal muscles [46] and comparisons of all three types of

human muscles are listed in table 1.6 [47].

1.7 Patch-clamp experiment

In order to construct a myometrial model which can be manipulated in a computa-

tional environment, data which describes the electrical and mechanical activities

must be obtained through experiments. One of the methods of studying the elec-

trophysiological properties of a cell is through the voltage-clamp experiment. It

is performed on individual cells in a biological laboratory. Since ions leaving and

entering the cell affect the membrane potential of a whole cell, measuring the ac-

tivity of the ion channels gives a good indicator of physiological significance of the

cell. To start with, the cell membrane of a single cell is held at a known potential

called the holding potential (voltage-clamp). A glass suction electrode that forms

a tight seal on an individual cell (patch-clamp) [23] is applied to measure the elec-

trical activity changes in the cell as shown in figure 1.14. The cell potential is

then stepped to a voltage protocol, the amount and the properties of the ionic

current resulting from flow of ions through channels can be calculated. In the

whole cell mode, the pipette with content solution can perforate the cell and have

direct access to the inside of the cell. The whole cell current measured represents

the sum contributions of the channels in the total membrane under conditions se-

lected. To achieve steady-state, the cell is clamped to a certain voltage for a long

period of time to ensure the cell is in equilibrium. Results from voltage-clamp

experiments are particularly useful when developing models for individual ionic

currents. Simulated results (activation and inactivation dynamics) of myometrial

currents were often compared to the recordings from the experiments to verify

whether good fits were achieved.

If the voltage is not clamped but instead, a fixed amount of external stimulus

current is injected, this is called current-clamp. Current-clamp technique are of-

ten used to study APs of a single cell. In computer simulations, the ‘virtual cell

is put in a voltage- or current-clamp under the same physiological conditions and

circumstances as described in experiments. Different voltage and current proto-

cols are then applied according to experimental conditions to see if the correct

results can be reproduced and predicted using purely computer simulations.

37

Figure 1.13: Schematic diagrams of skeletal muscles (A) and smooth muscles(B) showing their structural properties differences. Skeletal (striated) muscleis made up of individual muscle fibres called myofibre. The myofibres are longand cylindrical with multi-nucleate and consist of actin and myosin myofibrils.The muscles fibres are held together orderly by connective muscles. Smooth(non-striated) muscles on the other hand are shorter and are gathered into loosebundles. Myofilaments attached to the dense bodies (which serves as Z disks inskeletal muscle) at one end and connected to attachment plaques at the other endlocated at the plasma membrane of the smooth muscle cell. Image was adaptedfrom Lodish et al. [46].

38

Pro

per

tySke

leta

lm

usc

les

Car

dia

cm

usc

les

Sm

oot

hm

usc

les

Fib

redim

ensi

ons

100

mm

xup

to30

cm10

-20

mm

x50

-100

mm

5-10

mm

x30

-200

mm

*(d

iam

eter

xle

ngt

h)

Nucl

eiM

ult

iple

,nea

rsa

rcol

emm

aG

ener

ally

singl

e,ce

ntr

ally

Sin

gle,

centr

ally

loca

ted

loca

ted

Filam

ent

orga

nis

atio

nIn

sarc

omer

esal

ong

myo

fibri

lsIn

sacr

omer

esal

ong

myo

fibri

lsSca

tter

edth

rough

out

sarc

opla

smSR

Ter

min

alci

sten

aein

tria

ds

atzo

nes

ofov

erla

pSR

tubule

sco

nta

ctT

tubule

sat

Zlines

Dis

per

sed

thro

ugh

out

sarc

opla

sm,no

T-t

ubule

sC

ontr

olm

echan

ism

sN

eutr

al,

atsi

ngl

eneu

rom

us-

cula

rju

nct

ion

Auto

mat

icity

(pac

emak

ing

cells)

neu

ral

orhor

mon

alco

ntr

ol

Auto

mat

icity

(pac

emak

ing

cells)

Cal

cium

sourc

eR

elea

sefr

omSR

Extr

acel

lula

rfluid

and

rele

ase

from

SR

Extr

acel

lula

rfluid

Con

trac

tion

Rap

idon

set;

may

be

teta

nis

ed;

rapid

fati

gue

Slo

wer

onse

t;ca

nnot

be

teta

nis

ed;re

sist

ant

tofa

tigu

eSlo

won

set;

may

be

teta

nis

ed;

resi

stan

tto

fati

gue

Ener

gyso

urc

eA

erob

icm

etab

olis

mat

moder

-at

ele

vels

ofac

tivity;

glyco

ly-

sis

(anae

robic

)duri

ng

pea

k

Aer

obic

met

abol

ism

,usu

ally

lipid

orca

rboh

ydra

tesu

b-

stra

tes

Pri

mar

yae

robic

met

abol

ism

acti

vity

Table

1.6

:A

stru

ctura

land

funct

ional

com

pari

son

of

card

iac,

skele

tal

and

smooth

musc

lein

hum

an.

*As

pre

vio

usl

ydis

cuss

ed,th

ele

ngt

hs

ofnon

-pre

gnan

tm

yom

etri

alce

lls

and

cells

atte

rmar

e7.

5an

d20

cmre

spec

tive

ly.

Dat

aas

des

crib

edin

Mar

tiniel

al.[4

7].

39

Figure 1.14: A general whole-cell voltage clamp configuration in ex-periments. In control case, the micropipette is filled with solutions that matchthe intracellular concentration in the cell since the pipette electrode is in directcontact with the interior of the cell. On the other hand, the bath solution isreplaced by the extracellular solutions. Figure adapted from Molleman [34].

40

Chapter 2

Single uterine cell model

development

Although there are a few electrophysiological and force generation models for

single smooth muscle cells, at present, there is no mathematical single cell uterus

model that can predict the membrane potential, current and mechanical force

together. Gestrelius and Borgstrom [48] proposed a dynamical model of smooth

muscle contraction based on the sliding filament theory for the rat portal vein. In

2003, Yang et al. [49] developed a detailed model describing the myogenic response

of an isolated rat cerebrovascular arterial cell. Their model consisted of an elec-

trochemical and a chemomechanical subsystem. The electrochemical subsystem

includes ionic channels, an exchanger, pump and calcium-induced calcium release

mechanisms in the sacroplasmic reticulum. Calcium buffering and the balance

of intracellular Na+ and K+ were also taken into account. In chemomechanical

subsystem, myosin phosphorylation was explained using Hai and Murphy’s cross-

bridge model [50,51]. Active, passive and series viscoelastic force generation were

described using a modified Hill model [52]. The model was tested under iso-

metric and isotonic conditions and length-force and force-velocity relationships

were measured. Youm et al. (2006) [53] developed a mathematical model de-

scribing the pacemaking activity in small rat intestinal Cajal cells. Their model

included ionic channels, an exchanger, pumps, inositol 1,4,5-triphosphate (IP3)

production and IP3-mediated Ca2+ release activities. In the same year, Rihana

et al. [54] proposed an electrophysiological model for a single uterine cell using

general Hodgkin-Huxley type equations to describe the major currents. A sim-

ple intracellular calcium process was also incorporated. A bifurcation analysis

41

between the uterus membrane potential and current stimulation was carried out

and a repetitive firing behaviour in AP was shown using their model. Rihana

et al. further developed their model by including more ionic channels. Differ-

ent shapes of uterine electrical activities, action potential and the corresponding

[ Ca2+ ]i were investigated. Bursztyn et al. (2007) [38] proposed an excitation-

contraction model for a single uterine smooth muscle cell. This model accounts

for the voltage-gated calcium channels, pumps and the Na+ /Ca2+ exchanger.

Using the Hai and Murphy [50,51] cross-bridge model, stress was produced when

the calcium concentration was coupled with the rate constant of myosin phos-

phorylation. The Bursztyn model does not included other ionic channels (e.g

Na+ and K+ etc.). The ionic concentration of Na+ was set to a fixed value and

does not get updated during simulations.

In this thesis, a detailed novel mathematical model of a late pregnant (17-

19 days) rat longitudinal myometrial single cell was developed to aid designing

experiments in a laboratory research and provide better understanding of the

function of the uterus during pregnancy. The myometrial model reconstructed the

chemical, electrical and mechanical phenomena generated by the cell. Insufficient

data on human ionic currents was available, however data on the late pregnant

rat was more readily available, and therefore experimental data on pregnant rat

myometrial cells in both room and body temperature were used to construct the

model. When quantitative data for rat was not available, human data in similar

condition was substituted.

The model was constructed with two main parts. The first one is the electro-

chemical subsystem which consisted of all the inward and outward membrane

currents, electrogenic exchangers and pumps. The ionic currents were modelled

using Hodgkin-Huxley-type equations with a modified fluid compartment and

SR model developed by Kapela et al. for rat mesenteric smooth muscle [55] and

Bursztyn et al. for human myometrium [38]. The second part of the model, the

chemo-mechanical subsystem which was developed by Yang [49] described the

generation of the contractile force through the myosin phosphorylation (4-state

cross-bridge model), based on earlier development by Hai and Murphy [56, 57].

Figure 2.1 shows a functional block diagram of the myometrial cell model with

the two coupled subsystem.

The aim of the model was to reproduce the electrical and contractile activ-

ities, resting membrane, peak potential and spike frequency of a late pregnant

42

Figure 2.1: A block diagram of the myometrial cell model with the twocoupled subsystems. The myometrial mathematical model was divided intotwo subsystems: 1) Electrochemical and 2) Chemo-mechanical. The membranecurrents were modelled using Hodgkin-Huxlye type equations in the electrochem-ical subsystem, which described the electrical responses of the cell when the cellwas stimulated. The regulations of the influx of K+ , Na+ and Ca2+ (especiallythrough L-type Ca2+ channel) was described by the fluid compartment modeloriginally developed by Kapela et al. [55] and Bursztyn et al. [38]. The SR modeldescribed a calcium-induced Ca-release (CICR) process that controlled the releaseof Ca2+ from the SR store and back to the cytosol. The electrochemical subsys-tem was coupled with the chemo-mechanical one through the increase level of[ Ca2+ ]i . Hai and Murphy’s cross-bridge model was used in the myometriummodel to describe how [ Ca2+ ]i binds with calmodulin and activates the myosinlight-chain kinase (MLCK), and consequently forms cross-bridges between actinand myosin filaments, especially through the attached phosphorylated and de-phosphorylated non-cycling cross bridges (AM and AMp respectively) to generatemuscle contractions.

43

uterine cell. The model was validated by its ability to produce an action poten-

tial (AP) with bursts similar to those observed experimentally and intracellular

[ Ca2+ ]i transients in physiological ranges.

2.1 Membrane current model in myometrium

Cellular electrical activity can be explained in terms of electrical circuits. Since

the charges inside and outside a cell are imbalanced, which creates a potential

difference across the cell membrane. The phospholipid bilayer cell membranes

acts as a capacitor and accumulates charge as electrical potential across the cell

changes. Also, different ionic permeabilities of the membrane (ion channels) act

as resistors and the electrochemical driving forces in the cell act as batteries in the

electrical circuits. These ionic and capacitive currents are arranged in a parallel

circuit as shown in figure 2.2.

According to Ohms law, current, I is directly proportional to voltage, V and

conductance g. Since g is inversely proportional to resistance, R, I can be ex-

pressed as:

I = gV =V

R(2.1)

To understand and study the dynamics of the cell, equation 2.1 can be re-

arranged in a more useful way to investigate the time course and how various

currents flow in and out of the cell. Assuming the reversal potential, E stays

constant during the process, the current equation can be written as:

I = g(V − E) (2.2)

where V is the membrane potential, i.e. the potential difference between the

inside and the outside of the cell and (V - E) represents the driving force across

the membrane provided by the batteries. Since there are a number of ionic

currents in a cell, the sum of the total current, Itot can be presented as:

Itot =∑

IX =∑

gX (V − EX ) =

= gNa (V − ENa ) + gK (V − EK )− ... (2.3)

where gX and EX are the conductance and reversal potential of a particular ion,

44

Figure 2.2: The equivalent electrical circuit of a cell. The phospholipidbilayer cell membrane acts a capacitor separating ions inside and outside thecell while the electrochemical forces acts as batteries. Since charges inside andoutside the cell are imbalanced, electrical activity is generated when an externalstimulus, Ist is applied to the cell during experiments. This initiates APs andcontractions in the cell. The example ionic currents, Na+ and K+ channels areindicated by resistances (note the different direction of the flows). The membranepotential, V in the cell is determined by V = Vin − Vout and the total currentof the cell, Itot is the sum of all the ionic currents, IX in the cell where X is aparticular ion [58].

45

X, for example: sodium (Na+ ) and potassium (K+ ) etc. Since each circuit

element on the circuit diagram are in parallel, and assuming the cell membrane

acts as a capacitor, the capacitive current, Icap can be translated into an ordinary

differential equation (ODE):

Icap = CdV

dt(2.4)

where C is the capacitance of the cell. Also Kirchoffs current law states that

the sum of the currents flowing towards a certain point is equal to the sum of

currents flowing away from that point. Therefore the capacitive current must be

equal to the sum of the ionic currents and any currents that might be applied

during experimental manipulation, Ist . This can be expressed as:

Icap = −( Itot + Ist ) (2.5)

where the leading negative sign is necessary because of how current direction is

defined by convention. Combining equation 2.3 and equation 2.4, the expression

becomes:

CdV

dt= −

∑gX (V − EX )− Ist (2.6)

Since the conductances are generally not linearly dependent on the voltage (some-

times time), solving the differential equation for membrane voltage is not straight

forward. The relationships between conductances, voltage and time must be

found in order to study the activity of the cell. The ionic currents through ionic

channels of the cell may be computed from the transmembrane voltage according

to a Hodgkin-Huxley-type model [59] developed in 1952. In their model, ionic

currents are modelled as “gates”. Activation gates correspond to activation of

the ion channel and ions get transported across the cell membrane. Inactivation

gates on the other hand correspond to an inactivation of the channel (by confor-

mation changes in the channel protein) and stop ions getting transferred across.

The ionic conductance can be expressed as:

gX = gX ·ma · hb (2.7)

where gX is the maximal conductance, m and h are the activation and inactivation

46

gating variables respectively, a and b are integers chosen to fit the experimen-

tal data, and therefore an ionic current, IX can be described by the following

equation:

IX = gX ·ma · hb(V − EX ) (2.8)

The reversal potential of a particular ion is determined by the Nernst equation

and is given by:

EX =RT

zFln

([X]o

[X]i

)(2.9)

where F is the Faraday constant, R is the ideal gas constant, T is the absolute

temperature, and [X]o , [X]i are the extracellular and intracellular concentration

of the ion X. The gating variables for the ion channels were postulated to satisfy

linear differential equations where the variables “relax” to voltage-dependent val-

ues, or steady-state values m∞ and n∞. These values vary from one to zero with

voltage-dependent time constant τm or τh and are related as follows:

dm

dt=

m∞ −m

τm

(2.10)

dh

dt=

h∞ − h

τh

(2.11)

A total of 15 membrane currents in the myometrial model were developed as il-

lustrated in figure 2.3. The membrane currents are separated into 3 categories: 1)

Inward currents, currents which flow into the cell, i.e. hyperpolarisation-activated

current ( Ih ), L-type calcium current ( ICaL ), T-type calcium current ( ICaT ),

fast sodium current ( INa ) and store-operated non-selective current ( ISOC ). 2)

Outward currents, currents which flow out of the cell, i.e. two delayed rec-

tifying K+ currents ( IK1 and IK2 ), A-type transient K+ current ( IKA ), Ca2+ -

activated K+ current ( IKCa ) and a K+ background leakage current ( IKleak ). Fi-

nally 3) Other currents where the net flow of the current can be in both di-

rections, depending on the ionic concentration and the voltage of the cell, i.e.

Ca2+ -activated Cl− current ( ICl ) and non-specific cation current ( INSCC ). The

model also includes a calcium pump ( ICapump ), a Na+ -K+ pump ( INaK ), a

Na+ - Ca2+ exchanger ( INaCa ) and a sodium-potassium-chloride co-transporter

47

( INaKCl ).

The current model was constructed based on published experimental data

using information such as steady-state activation and inactivation, activation

and inactivation time constants, current-voltage (I-V) relationships and current

traces. Experimentally obtained figures were digitized into the computer us-

ing ScanIt or Engauge digitizer programs. Using graphing packages such as

SigmaPlot or gnuplot, an appropriate function was fitted to the data and the

variables of the function returned. The functions were chosen by inspection prior

to fitting. These were then be used to reconstruct an equation for the whole ion

channel. The simulated results were compared and validated to the experimen-

tally observed data when available. The following ionic current equations were

normalised in units of pA/pF while membrane voltages are in mV and conduc-

tances in nS/pF. The parameter values, initial values of the gating variables and

all the symbols used in the model including their definitions are all listed in the

appendix.

2.1.1 Inward currents

The single myometrial cell model consists of four inward membrane currents:

hyperpolarisation-activated current, Ih , L- and T-type calcium current, ICaL and

ICaT , fast sodium current, INa and store-operated non-selective cation current,

ISOC . ICaL and Ih were the only experiments conducted at body temperature

or which could be converted using the value of Q10 provided. When Q10 was

not provided for temperature correction, the dynamics of the currents were left

unchanged, assuming the currents behave similarly in both room and body tem-

perature. Also, there are no existing steady-state data on myometrial T-type

calcium channel, so data from rat genes α1G/Cav3.1 were substituted and mod-

ified to match the current density in human myometrium data.

2.1.1.1 Hyperpolarisation-activated current – Ih

Hyperpolarisation-activated current ( Ih ) has been identified in rat circular and

longitudinal myometrium [26, 60]. This current is also called If (funny current)

in cardiac cells and is found commonly in other types of myogenic cells. Ih is

activated by hyperpolarisaion of the membrane potential below -60 mV with a

48

Figure 2.3: Schematic cell diagram for the myometrial model. An il-lustration of all the ionic channels, pumps and exchangers in the myometrial cellmodel with directions of current flux leading to the control of [ Ca2+ ]i in the cal-cium store. A total of 15 membrane currents were presented in the myometrialmodel including five inward currents (hyperpolarisation-activated current ( Ih ),L-type and T-type calcium current ( ICaL , ICaT ), fast sodium current ( INa )) andstore-operated non-selective current ( ISOC ), five outward currents (1 and 2 de-layed rectifying K+ currents ( IK1 and IK2 ), A-type transient K+ current ( IKA ),Ca2+ -activated K+ current ( IKCa ) and a small K+ background leakage current( IKleak )) and other currents (Cl− current ( ICl ) and non-specific cation current( INSCC )). Also, a plasma membrane Ca2+ -ATPase (PMCA) or calcium pump( ICapump ), Na+ -K+ pump ( INaK ), Na+ - Ca2+ exchanger ( INaCa ) and sodium-potassium-chloride co-transporter ( INaKCl ) were also included in the model. Thecontrol of the Ca2+ entering and leaving the SR through SR Ca2+ -ATPase(SERCA) pump and ryanodine receptor (RyR) were described by the uptake cur-rent ( Iup ) and the release current ( Irel ). Ca2+ was diffused across from the uptakecompartment to the release compartment of the SR described by the transversecurrent ( Itr ) while a Ca2+ -binding protein, calsequestrin (CSQN) help storingthe Ca2+ in the SR.

49

half maximal activation around -85 mV and does not inactivate. The activa-

tion time of Ih is slow, taking around 1 s to fully activate. Ih is permeable to

K+ and Na+ with a permeability ratio of PNa /PK = 0.35 [61]. The reversal

potential of Ih , Eh is around -20 mV and is dependent on the extracellular con-

centration of K+ and Na+ , with excess K+ increasing the current amplitude and

low Na+ reducing the amplitude. Application of Cs+ inhibits and limits the fre-

quency of Ih . Since Ih is activated at the resting membrane potential, this current

is responsible in maintaining the resting potential and spontaneous activities in

myometrium.

There are two separate experimental studies to date which have reported

on Ih in pregnant myometrium. Okabe et al. [26] conducted their research with

17-19-day pregnant rat circular uterine cells at both 30◦C and 24◦C whereas

Satoh [60] performed an experiment on 18-day pregnant rat longitudinal cells at

room temperature, 22◦C. In order to construct the whole cell myometrial model

in body temperature, the activation time constant of Ih was corrected with a Q10

value of 3.5 [26]. Also, the single cell model was developed for a longitudinal cell,

the half-activation obtained in circular cells was modified and left-shifted by 15

mV to match the experimental I-V data in longitudinal cells [26]. The current

density of Ih in longitudinal myometrial cells is -1.03 pA/pF at -120 mV from a

holding potential of -50 mV at room temperature [60]. Steady-state activation

and time constant, I-V curve and current tracings of Ih are all shown in figure

2.4.

Hyperpolarisation-activated current – Ih

Ih = gh y(V − Eh ) (2.12)

Eh =RT

Fln

([ K+ ]o + ( PNa / PK ) [ Na+ ]o

[ K+ ]i + ( PNa / PK ) [ Na+ ]i

)(2.13)

y∞ =1

1 + exp

(V + 105.39

8.66

) (2.14)

τy =1

3.5x10−6 exp (−0.0497V) + 0.04 exp (0.0521V)(2.15)

dy

dt=

y∞ − y

τy

(2.16)

50

Parameters Values Definitiongh 0.0542 nS/pF Maximum conductance of IhPNa /PK 0.35 Na+ and K+ permeability ratio

Table 2.1: Parameter values of Ih

2.1.1.2 L-type calcium current – ICaL

L-type calcium current is found in both rat and human myometrium and ICaL is

the major contributor to the total inward current. It is also the main entry

route for calcium which triggers the myometrial contractile activities [25,62–64].

Studies suggested Ca2+ channel and cell density increases with up to gestation 18

days in pregnant rat [65] to prepare for synchronised contraction during labour at

term. This current is inhibited by Mg2+ and dihydropyridines such as verapamil,

nifedipine and israpidine and inactivated by increasing intracellular [ Ca2+ ]i [8,

17,66,67].

ICaL activates at around -40 to -30 mV and peaks at around -10 to 10 mV. It

also has a reversal potential, ECa of around 45 to 60 mV at 30 to 35 ◦C with 1.5 to

2.5 mM [ Ca2+ ]o [32,67–69]. ICaL is permeable to other ions [70] but quantitative

data specifically for myometrial cells is lacking, therefore the Goldman-Hodgkin-

Katz formulation was not used for calculating ECaL in the single myometrial cell

model. Instead the value of ECaL was fixed at 45 mV [68,69,71].

The conductance of ICaL was modelled by three gating variables: an activation

gating variable, d, and fast and slow inactivation gating variables, f1 and f2 .

Experiments performed by various groups at 30 - 35 ◦C reported different steady-

state values for activation and inactivation. This is due to different values of

[ Ca2+ ]o used experimentally in the bath solution and hormonal influence of the

myometrial cells. Yoshino et al. [8] showed that the half-activation and the I-

V relationship was right-shifted by about 15 mV when [ Ca2+ ]o decreased from

43 mM to 30 mM. However, when myometrial cells were exposed to estradiol,

the half-inactivation was left-shifted and the I-V relationship was reduced as

demonstrated by Yamamoto [68]. Data obtained from isolated myometrial cells

near term (19-21 days gestation) in rat showed the steady-state inactivation of

ICaL became more hyperpolarised, i.e. more negative [69]. This is because the level

of estradiol increases near term, and the cells have been exposed to this estradiol

naturally causing this effect. The activation of ICaL from the uterine cells near

term was also more hyperpolarised compared to other experimental recordings.

51

0

0.2

0.4

0.6

0.8

1

-120 -100 -80 -60 -40

I/Im

ax

V (mV)

A

Okabe 1999Best fit

y∞

0

0.4

0.8

1.2

1.6

-140 -100 -60 -20

τ (s

)V (mV)

B

Okabe 1999τih

-1.2

-1

-0.8

-0.6

-0.4

-0.2

0

-120 -100 -80 -60 -40

I/Im

ax

V (mV)

C

Okabe 1999Satoh 1995

Model

-1.4-1.2

-1-0.8-0.6-0.4-0.2

0

0 1 2 3

I/Im

ax

t (s)

D Okabe 1999Model

-140

-20

V (mV)

Figure 2.4: Model of myometrial Ih . (A) Steady-state activation of Ih .A modified curve was used instead of the best fit in our model to compensatethe differences in cell type. (B) Activation time constant of Ih corrected witha Q10 value of 3.5 with experimental data. The kinetics of both longitudinaland circular cells was assumed to be unchanged. (C) I-V curve from Satoh etal. [60] (cross, longitudinal cell) and Okabe et al. [26] (triangle, circular cell) withsimulated results. The current was normalised at -120 mV. (D) Experimentaland simulated current traces. The current was depolarised to -130 mV, -100 mV,-90 mV to -70 mV every 10 mV for 3 seconds from a holding potential of -50 mV(inset). The current was normalised at -130 mV.

52

The steady-state activation and inactivation of the model was obtained by fitting

to the experimental data obtained from Yamamoto in control condition at body

temperature [68] (figure 2.5 (A) and (B)). Other experimental data from both cell

and tissue at body temperature were also presented in the figure for comparison.

The activation time constant, τd of the model was obtained by fitting the

initial few seconds of time tracings obtained from available experimental data

prior to ICaL reaching its peak [32] (figure 2.5 (C)). The fast and slow inactivation

time constants, τ f1 and τ f2 were obtained from Amedee et al. [67] illustrated in

figure 2.5 (D). The calcium-dependent inactivation of ICaL , fCa was described by

a Hill equation with the half saturation concentration of ICaL , Km,CaL = 0.6 µM

and a Hill coefficient of 1. The simulated I-V relationship and current traces of

ICaL were compared to experimental data shown in figure 2.5 (E) and (F). The

current traces produced by the model shows ICaL reached its peak in about 12

ms before the current inactivated. The simulated I-V relationship shows that

ICaL first appears at a V of around -40 to -30 mV and peaks at mV, similar to

experimental reported data [32,67–69].

53

L-type calcium current – ICaL

ICaL = gCaL d2 fCa (0.8 f1 + 0.2 f2 )(V − ECaL ) (2.17)

fCa =1

1 +[ Ca2+ ]i

Km,CaL

(2.18)

d∞ =1

1 + exp

(− (V + 22)

7

) (2.19)

f∞ =1

1 + exp

(V + 33

7

) (2.20)

τd = 2.29 +5.7

1 +

(V + 29.97

9

)2 (2.21)

τ f1 = 12 ms (2.22)

τ f2 = 90.97− 90.97(

1 + exp

(V + 13.96

45.38

))(1 + exp

(− (V + 9.5)

3.39

)) (2.23)

dd

dt=

d∞ − d

τd

(2.24)

d f1

dt=

f∞ − f1

τ f1

(2.25)

d f2

dt=

f∞ − f2

τ f2

(2.26)

Parameters Values DefinitiongCaL 0.6 nS/pF Maximum conductance of ICaL

ECaL 45 mV Reversal potential of ICaL

Km,CaL 0.6 µM Half saturation concentration of ICaL

Table 2.2: Parameter values of ICaL

54

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50

I/Im

ax

V (mV)

A

(Jones et al. 2004)(Okabe et al. 1994)

(Yamamoto 1995)Shmigol et al. 1998

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50

I/Im

ax

V (mV)

A

Amdee et al. 1987Jmari et al. 1986

d∞

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50

I/Im

ax

V (mV)

B

Shmigol et al. 1998Yamamoto 1995

Amedee et al. 1987Jmari et al. 1986

f∞

1

3

5

7

9

-100 -50 0 50

τ (m

s)

V (mV)

C

Jones et al. 2004τd

0

25

50

75

100

-100 -50 0 50

τ (m

s)

V (mV)

D

Amedee et al 1987τf2

0

25

50

75

100

-100 -50 0 50

τ (m

s)

V (mV)

D

Amedee et al 1987τf1

-1.2

-1

-0.8

-0.6

-0.4

-0.2

0

0.2

-100 -50 0 50

I/Im

ax

V (mV)

E Jones et al 2004Okabe et al 1999

Shmigol et al 1998

-1.2

-1

-0.8

-0.6

-0.4

-0.2

0

0.2

-100 -50 0 50

I/Im

ax

V (mV)

E Yamamoto 1995Amedee et al 1987

Simulation

-1

-0.8

-0.6

-0.4

-0.2

0

0 30 60 90 120

I/Im

ax

t (ms)

F

-60

0

V (mV)

Figure 2.5: Model of myometrial ICaL . (A and B) Steady-state activationand inactivation of ICaL . Experimental data obtained from Yamamoto [68] wereused to obtain both d∞ and f∞ for the model. Other data from late pregnantrat myometrial cell [32, 67, 71] and tissue [66] were also shown for comparison.Data in brackets were extrapolated from their reported I-V relationships usingthe function d∞=ICaL /(V-ECaL ) and normalised to the maximum value. (C andD) Activation time constant, fast and slow inactivation time constant of ICaL .The fast inactivation shown in grey is voltage-independent with a fixed value of12 ms. (E and F) Peak I-V relationship with experimental data and simulatedcurrent tracings of ICaL . All data in (E and F) are normalised to the peak currentvalue at 0 mV.

55

2.1.1.3 T-type calcium current – ICaT

Transient, or T-type calcium current, ICaT is found in late pregnant human my-

ometrial cells but it has been reported that this channel is not found in late preg-

nant rat myometrial cells [63, 64, 72, 73]. However, the existence of this channel

in late pregnant rat has never been conclusively ruled out. It has been demon-

strated that mRNA for α1G and α1H subunits of the T-type calcium channel

were expressed and dynamically regulate in rat myometrium throughout the ges-

tation [74]. Furthermore, the contractions in pregnant rat myometrial tissue strips

were markedly reduced by mibefradil, a potent T-type calcium blocker [75], al-

though the results could also be explained by the inhibition effects of mibefradil

on L-type calcium channels [75–77]. It has been suggested ICaT plays an im-

portant role in generating low-threshold Ca2+ spikes and AP propagation rather

than a direct link to muscle contractions in the myometrium [78].

In the uterine model, the conductance of ICaT is modelled by two gating vari-

ables, an activation and inactivation gating variable, b and g respectively. Steady-

state activation and inactivation data of the channel on myometrium was unavail-

able, therefore recordings from cloned rat genes (α1G/Cav3.1) expressed in HEK

cells were substituted [79,80] (figure 2.6 (A)). Both of the steady-state activation

and inactivation values and the I-V relationships between the expressed rat α1G

behave very similarly to recordings observed in human myometrium [64, 81, 82].

The current density was then adjusted to match the human myometrium data.

Activation and inactivation time constants of the model with experimental data

are shown in figure 2.6. The equation of the activation time constant was chosen

to fit the simulated time-to-peak experimental data.

The reversal potential of the current is fixed at 45 mV in the model due

to the reason discussed previously for ICaL . The I-V relationship produced by

simulations with experimental data are illustrated in figure 2.6 (D). It showed

ICaT first activated at -60 mV and peaked between -20 to -30 V. The current

traces illustrated in figure 2.6 (E) showed ICaT activated rapidly and inactivation

of this channel can vary between 7 - 100 ms depending on the voltage [64,78–82].

56

T-type calcium current – ICaT

ICaT = gCaT b2g(V − ECaT ) (2.27)

b∞ =1

1 + exp

(− (V + 54.23)

9.88

) (2.28)

g∞ = 0.02 +0.98

1 + exp

(V + 72.98

4.64

) (2.29)

τb = 0.45 +3.9

1 +

(V + 66

26

)2 (2.30)

τg = 150− 150(

1 + exp

(V − 417.43

203.18

))(1 + exp

(− (V + 61.11)

8.07

)) (2.31)

db

dt=

b∞ − b

τb

(2.32)

dg

dt=

g∞ − g

τg

(2.33)

Parameters Values DefinitiongCaT 0.0174 nS/pF Maximum conductance of ICaT

ECaT 42 mV Reversal potential of ICaT

Table 2.3: Parameter values of ICaT

2.1.1.4 Fast sodium current – INa

Voltage activated inward fast sodium channel is found in pregnant human myome-

trial cells and tissues. It was first identified by Amedee et al. in myometrium [83]

and it was found that INa is expressed in some but not all late pregnant rat my-

ometrial cells [8,27,62]. This channel is tetrotodoxin- and sagitoxin- sensitive. In

rat INa activates at around -50 to -40 mV and its I-V relationship shows this cur-

rent peaks at -10 to 10 mV, with a magnitude of 0.86 to 3.67 pA/pF [8,27,72,84].

57

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50

I/Im

ax

V (mV)

AHering et al. 2004

(Serrano et al. 1994)Serrano et al. 1994

b∞

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50

I/Im

ax

V (mV)

AHering et al. 2004

Serrano et al. 1994g∞

0

5

10

15

20

25

-100 -50 0 50T

ime-

to-p

eak

(ms)

V (mV)

B

Serrano et al. 1994Simulation

0

40

80

120

160

-100 -50 0 50

τ (m

s)

V (mV)

C

Hering et al 2004Serrano et al 1999

τg

-1.2

-0.8

-0.4

0

0.4

-100 -50 0 50

I/Im

ax

V (mV)

D

Serrano et al 1999Hering et al 2004

Best fitModel -1

-0.8

-0.6

-0.4

-0.2

0

20 40 60 80 100

I/Im

ax

t (ms)

E

-80

20

V (mV)

Figure 2.6: Model of myometrial ICaT . (A) Steady-state activation andinactivation of ICaT compared with experimental data obtained from cloned ratgenes (α1G/Cav3.1) expressed in HEK cells. Data in brackets was extrapolatedfrom the I-V relationship using the function b∞=ICaT /(V-ECaT ) and normalisedto the maximum value. (B) Superimposed simulated and experimental time-to-peak at different voltage steps from a holding potential of -100 mV. A functionfor the voltage-dependent time constant was chosen to produce simulated time-to-peak matching the experimental data. (C) Voltage-dependent inactivationtime constant of ICaT with experimental data. (D) I-V relationship of ICaT . Thereversal potential of ICaL , ECaT was changed from 45 mV (solid line, indicated asmodel) to 25 mV to match the experimental data (dotted line, indicated as ”Bestfit”). (E) Current traces produced by the model during voltage-clamp protocolas shown in the inset. A voltage-clamp was applied from a holding potential of-80 mV and stepped up from -60 to 20 mV for 90 ms. All simulated data from(D) and (E) were normalised to the peak current value at -25 mV.

58

The comparison of electrophysiological parameters between rat and human my-

ometrium obtained from Young et al. [84] in INa is illustrated in table 2.1.1.4. Cur-

rent tracings obtained from experiments show INa activates very fast and reaches

its peak within ≈1 ms and then almost completely inactivated [8, 63,72,85].

Species Activation (mV) Inactivation (mV)Onset Peak V0.5 k

Rat -40 0 -64 7.1Human -40 -10 -67 8.5

Table 2.4: The comparison of electrophysiological parameters betweenrat and human myometrium for INa [84].

Studies suggested that this channel may be responsible for the fast upstroke

of AP generation. In rat, some studies found that the number density of sodium

channels seem to increase as term approaches [27]. Along with the formation of

gap junction, INa is responsible for a rapid and more complete excitation propa-

gation during parturition at term [86].

The conductance of INa was modelled by two gating variables: an activation

gating variable, m and an inactivation gating variable, h. Steady state values

for activation and inactivation are shown in figure 2.7 (A). Both time constants

of activation and inactivation shown in figure 2.7 (B) were obtained by fitting

the current time tracings from experimental data [8, 27, 63, 72]. Simulated time

tracings and the I-V relationship were compared to experimental recordings in

figure 2.7 (C) and (D). The current tracings produced by the model showed the

dynamics were similar to the experimental tracings [8, 27, 63, 72]. After reaching

its peak at 0 mV in ≈ 2 ms, INa inactivated within 10 ms. The simulated I-V

relationship revealed that INa first appeared at about 50 mV, which is consistent

with the experimental data.

59

Fast sodium current – INa

INa = gNa m3h(V − ENa ) (2.34)

ENa =RT

Fln

([Na]o

[Na]i

)(2.35)

m∞ =1

1 + exp

(− (V + 35.96)

9.24

) (2.36)

h∞ =1

1 + exp

(V + 57

8

) (2.37)

τm = 0.25 +7

1 + exp

(V + 38

10

) (2.38)

τh = 0.9 +1002.85

1 +

(V + 47.5

1.5

)2 (2.39)

dm

dt=

m∞ −m

τm

(2.40)

dh

dt=

h∞ − h

τh

(2.41)

Parameters Values DefinitiongNa 0.12 nS/pF Maximum conductance of INa

Table 2.5: Parameter values of INa

2.1.1.5 Store-operated non-selective cation current – ISOC

Apart from voltage-operated L-type and T-type Ca2+ channels, Ca2+ can also

enter the cell through a receptor-operated store-activated channel. It was first

proposed by Putnety [87] and this store-operated non-selective cation current,

ISOC is permeable to Ca2+ (capacitative calcium entry, CCE) as well as other ions

(non CCE) [28,45]. This channel is identified in pregnant human myometrium and

60

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50

I/Im

ax

V (mV)

A

Sperelakis et al. 1992Sperelakis et al. 1992

m∞h∞

0.1

1

10

100

1000

-100 -50 0 50τ

(ms)

V (mV)

B

Ohya and Sperelakis 1989Inoue and Sperelakis 1992

τhOhya and Sperelakis 1989Inoue and Sperelakis 1992

Yoshino et al. 1997τm

-1.4

-1.2

-1

-0.8

-0.6

-0.4

-0.2

0

-100 -50 0 50

I/Im

ax

V (mV)

C

SimulationOhya and Sperelakis 1989Inoue and Sperelakis 1992

-1

-0.8

-0.6

-0.4

-0.2

0

0 10 20 30 40 50

I/Im

ax

t (ms)

D

-90

0

V (mV)

Figure 2.7: Model of myometrial INa . (A) Steady-state activation, m∞and inactivation, h∞ of INa . Experimental data was obtained from Sperelakis etal. [63] from rat longitudinal cells. Both m∞ and h∞ are fitted with sigmoidalequations. (B) Activation and inactivation time constant recordings obtainedfrom experiments [8,27,72]. (C) I-V relationship of INa compared to experimentaldata. (D) INa current tracings produced by the model. The model was held at-90 and stepped up to 0 mV from -40 mV every 10 mV for 40 ms. Both simulatedresults in (C) and (D) are normalised to peak current value at 0 mV.

61

is responsible for replenishing the Ca2+ in the SR store [88–91]. In myometrium,

store-operated Ca2+ entry is elicited by thapsigargin and activated when inositol

1,4,5-trisphosphate (InsP3)-sensitive calcium store is depleted [28]. Babich et al.

found that rTrpC4 protein has an important role in Ca2+ entry regulation through

ISOC in rat myometrial cells during pregnancy [92]. ISOC through CCE and non-

CCE are important in oxytocin-induced Ca2+ responses [28, 49, 93]. The role of

this current includes the generation of calcium oscillations and prolongation of

agonist-induced calcium signal.

Quantitative experimental data describing this channel is lacking, so formula-

tions of ISOC were substituted with equations from rat mesenteric smooth muscle

cell (MSMC) by Kapela et al. [55]. In MSMC, ISOC has a linear I-V relationship

and showed a little inactivation. The maximum current of -10 to -5 pA [94] and

the permeability ratio of Ca2+ to K+ was 8.2 [95]. Base on these data, the con-

ductances of gSOCCa and gSOCNa were estimated. The open probability, PSOC of

this channel was formulated with a sigmoidal function and the unit of the ISOC for

the uterine model was converted from pA to pA/pF. This was done by dividing

the whole current by the capacitance of a late pregnant rat myometrial cell, esti-

mated to be 100 pF.

Store-operated non-selective cation current – ISOC

ISOC = ISOCCa + ISOCNa (2.42)

ISOCNa = gSOCNa PSOC (V − ENa ) (2.43)

ISOCCa = gSOCCa PSOC (V − ECa ) (2.44)

PSOC =

(1 +

[ Ca2+ ]uKm,SOC

)−1

(2.45)

Parameters Values DefinitiongSOCCa 8.3×10−5 nS/pF Maximum conductance of the

Ca2+ component of ISOC

gSOCNa 5.75×10−4 nS/pF Maximum conductance of theNa+ component of ISOC

Km,SOC 100 nM Half activation constant of ISOC

Table 2.6: Parameter values of ISOC

62

2.1.2 Outward currents

There are various types of outward potassium channel expressed in late preg-

nant rat myometrium, with diverse properties and characteristics. They include

the fast inactivation transient current, delayed rectifiers and calcium activation

potassium currents. Experimental recordings showed that there are two delayed

rectifiers [81] and two calcium activation potassium currents [96] but no inward

rectifying potassium current was found [31]. Interpretation of the various sub-

types of potassium currents are difficult, the classification of the currents are

categorised differently in published literatures depending on the research groups.

This is because there are no specific blockers to selectively block individual types

of voltage-dependent potassium currents in myometrial cells.

In this model, outward potassium currents are categorised into four different

potassium sub-currents and a small background leakage current. They are: de-

layed rectifying potassium current one and two, IK1 and IK2 , A-type transient

potassium current, IKA , calcium-activated K+ current, IKCa and background leak-

age K+ current, IKleak . Definitions of different channels are classified according

to the work by Khan et al. [2, 97] and Knock et al. [29, 30], which distinguish

potassium channels by their inactivation dynamics and activators. Khan et al.

focused their experiments on potassium uterine single channels while Knock et al

performed all their experiments on pregnant human single cells in myometrium.

The main function of the K+ current is to dampen cellular excitability by main-

taining the cell membrane potential close to the reversal potential of the K+ ion.

This produces repolarisation that brings the cell membrane potential to its resting

potential, and also renders the AP generation and reduces or stops contraction

eventually. The K+ current is particularly important to keep the myometrium

quiescent during the early and middle stage of the pregnancy. Interestingly,

the majority of human myometrial cells consisted of either IK1 (24/42 cells) or

IK2 (18/42 cells) as the dominant potassium current [81], and only a very small

number of human myometrial cells were reported to consist of both types of de-

layed rectifying K+ channels [30]. It is reported that the voltage-gated potassium

current accounted for almost 90% of total potassium current in late pregnant rat

myometrial cells [31].

63

2.1.2.1 Delayed rectifying potassium current – IK1

Voltage-gated potassium currents, IK1 is a 4-aminopyridine (4-AP)-insensitive

current with half inactivation around -63 mV and activation threshold around

-60 to -40 mV. This current is sensitive to tetraethylammonium and clofilium de-

pending on the concentrations of the chemical. IK1 activates fast, but inactivates

very slowly and it does not fully inactivate. The non-inactivation component

contributes around 30-43% of the total current. The conductance of current was

modelled by three gating variables: an activation gating variable, q and fast and

slow inactivation gating variables, r1 and r2 . The activation and inactivation

steady state values were obtained from rat myometrial cells [31] as shown in

figure 2.8 (A). The activation and inactivation time constants were obtained by

fitting published [29] and additional unpublished human myometrial current time

tracings provided by Knock, G and Aaronson, P through personal communica-

tion (figure 2.8 (B) and (C)). The simulated I-V curves from different holding

potentials shown in figure 2.8 revealed that 50% of the IK1 was activated at 20

mV similar to experimental recordings from Knock et al. [29]. Current tracings

produced by the model illustrated in figure 2.8 (E) show that this current reached

its peak value in about 50 ms and took about 10 s to inactivate.

64

Delayed rectifying potassium current – IK1

IK1 = gK1 q(0.38 r1 + 0.62 r2 )(V − EK ) (2.46)

EK =RT

Fln

([K]o

[K]i

)(2.47)

q∞ =1

1 + exp

(− (V − 7.7)

23.7

) (2.48)

r∞ =1

1 + exp

(V + 63

6.3

) (2.49)

τq =500

1 +

(V + 60.71

15.79

)2 (2.50)

τ r1 =5× 104

1 +

(V + 62.71

35.86

)2 (2.51)

τ r2 = 3× 104 +2.2× 105

1 + exp

(V + 22

4

) (2.52)

dq

dt=

q∞ − q

τq

(2.53)

d r1

dt=

r∞ − r1

τ r1

(2.54)

d r2

dt=

r∞ − r2

τ r2

(2.55)

Parameters Values DefinitiongK1 0.2275 nS/pF Maximum conductance of IK1

Table 2.7: Parameter values of IK1

65

0

0.2

0.4

0.6

0.8

1

-90 -60 -30 0 30 60

I/Im

ax

V (mV)

A

Wang et al 1998q∞r∞

1

10

100

1000

-90 -60 -30 0 30 60

τ (m

s)

V (mV)

B

Knock et al 1999τq

0.1

1

10

100

1000

-90 -60 -30 0 30 60

τ (s

)

V (mV)

C τr1Knock et al 1999 and upublished

(Knock and Aaronson)

0.1

1

10

100

1000

-90 -60 -30 0 30 60

τ (s

)

V (mV)

C τr2Knock et al 1999 and upublished

(Knock and Aaronson)

0

0.2

0.4

0.6

0.8

1

-90 -60 -30 0 30 60

I/Im

ax

V (mV)

D

Knock et al 1999Simulation

0

0.2

0.4

0.6

0.8

1

0 1 2 3 4 5 6 7 8

I/Im

ax

t (s)

E

SimulationKnock et al 1999

-80

10

V (mV)

Figure 2.8: Model of IK1 . (A) Experimental data and simulations of steady-state activation, q∞ (solid line) and inactivation, r∞ (dotted line) of IK1 . r∞was formulated with the half-inactivation of -63 mV and slope factor of 6.3 mVreported from Wang et al. [31]. (B) Activation time constant and (C) Fast (dottedline) and slow inactivation time constant (solid line) of IK1 . Experimental valueswere extracted by fitting and averaging the data of current tracings from Knocket al. [29] and unpublished data from Knock and Aaronson. (D) Experimentaland simulated I-V curve of IK1 . The current was elicited by voltage pulses from-80 to 60 mV every 10 mV (holding potential of -80 mV). The I-V curve wasmeasured at the peak value and was normalised at 60 mV. (E) Experimentalraw traces (cross point) and simulations (solid line) of IK1 were normalised to thepeak current at 10 mV.

66

2.1.2.2 Delayed rectifying potassium current – IK2

IK2 is a 4-AP-sensitve current that has a lot of similar characteristics as IK1 despite

having faster kinetics. This current has a more positive half-inactivation of -30

mV and activation threshold between -40 to -30 mV shown in figure 2.9 (A).

IK2 also does not fully inactivate and this component accounted for about 8-14%

of the total current. Both IK1 and IK2 are unevenly distributed between human

myometrial cells. The conductance of IK2 was modelled similarly to IK1 by an ac-

tivation gating variable, p and fast and slow inactivation gating variables, k1 and

k2 . In the same method as IK1 , the activation and inactivation time constants

were fitted to human myometrial data from Knock et al. [81] and unpublished

data provided by Knock, G and Aaronson, P (figure 2.9 (B) and (C)). Simulated

I-V relationship and current tracings illustrated in figure 2.9 (D) and (E) show

that IK2 took about 50 ms to reach the peak and inactivated at a faster rate than

IK1 .

67

Delayed rectifying potassium current – IK2

IK2 = gK2 p(0.75 k1 + 0.25 k2 )(V − EK ) (2.56)

EK =RT

Fln

([K]o

[K]i

)

p∞ =1

1 + exp

(− (V − 4.2)

23

) (2.57)

k∞ =1

1 + exp

((V + 21.2)

5.7

) (2.58)

τp =100

1 +

(V + 64.1

28.67

)2 (2.59)

τ k1 = 1× 106 − 1× 106

(1 + exp

(V − 315

50

))(1 + exp

(− (V + 74.9)

8

)) (2.60)

τ k2 = 2.5× 106 − 2.5× 106

(1 + exp

(V − 132.87

25.40

))(1 + exp

(− (V + 24.92)

2.68

))

(2.61)

dp

dt=

p∞ − p

τp

(2.62)

d k1

dt=

k∞ − k1

τ k1

(2.63)

d k2

dt=

k∞ − k2

τ k2

(2.64)

Parameters Values DefinitiongK2 0.07875 nS/pF Maximum conductance of IKA

Table 2.8: Parameter values of IK2

68

0

0.2

0.4

0.6

0.8

1

-90 -60 -30 0 30 60

I/Im

ax

V (mV)

A

Wang et al 1998p∞k∞

0

20

40

60

80

100

-90 -60 -30 0 30 60

τ (m

s)

V (mV)

B

Knock et al 1999τp

1

10

100

1000

10000

-90 -60 -30 0 30 60

τ (s

)

V (mV)

C τr1Knock et al 1999 and upublished

(Knock and Aaronson)

1

10

100

1000

10000

-90 -60 -30 0 30 60

τ (s

)

V (mV)

C τr2Knock et al 1999 and upublished

(Knock and Aaronson)

0

0.2

0.4

0.6

0.8

1

-90 -60 -30 0 30 60

I/Im

ax

V (mV)

D

Simulation

0

0.2

0.4

0.6

0.8

1

0 1 2 3 4 5 6 7 8

I/Im

ax

t (s)

E

-80

10

V (mV)

Figure 2.9: Myometrial model of IK2 . (A) Steady-state activation, p∞(solid line) and inactivation, k∞ (dotted line) of IK2 with experimental data. k∞was formulated with the reported half-inactivation value of -21.2 mV and slopefactor of 5.7 mV [31]. (B) Activation time constant of IK2 . (C) Fast (solid line)and slow inactivation (dotted line) of IK2 with experimental data. (D and E)I-V relationship and current tracings of IK2 . Both currents in (D) and (E) arenormalised to the peak current at 60 mV in the I-V curve and 10 mV in thecurrent tracings. Note there are no existing experimental data on both the I-Vcurve and current traces.

69

2.1.2.3 A-type transient potassium current – IKA

IKA is a 4-AP sensitive and TEA insensitive potassium transient current with very

fast activation and inactivation kinetics. It is found in various smooth muscles [98,

99] and non-pregnant rat uterine myocyctes, but IKA is rare in pregnant rat and

human myometrial cells [30, 62]. Experiments performed by Knock et al. [29, 30]

using human non-labour single cells at term (38-41 weeks) in room temperature

showed IKA activated at around -40 mV, with a half-inactivation of around -70

mV and a slope factor of around 5 mV. The current reached its peak value within

10 ms and almost completely inactivated within 50 ms [30,62]. In voltage-clamp

experiments, IKA caused the sudden changes in the initial time course of the

total potassium currents in late pregnant rat myometrial cells prior to the total

potassium current reaching its peak [31,62].

The conductance of IKA was modelled by an activation gating variable, s and

an inactivation gating variable, x. The Steady-state activation and inactivation

data was obtained using unpublished data provided by Knock, G and Aaronson, P

(figure 2.10(A)). The function of the activation time constant was chosen to fit the

time-to-peak experimental data shown in 2.10(B). The inactivation time constant

was measured by extracting an exponential equation from raw traces [30]. The I-V

relationship illustrated in 2.10 (C) shows IKA first appeared at -40 mV, similar to

experimental data. The simulated I-V curve was normalised to the peak current

value at 60 mV. The peak current of IKA reported from experimental recordings

was about 2 - 4 pA/pF in human myometrial cells [30] and about 18 pA/pF in rat

myometrial cells [62] at voltage steps of 40 - 60 mV from a holding potential of

-80 mV. Raw current traces of IKA illustrated in figure 2.10 (D) shows the current

rapidly activated and completely inactivated, unlike IK1 and IK2 .

70

A-type transient potassium current – IKA

IKA = gKA s x(V − EK ) (2.65)

EK =RT

Fln

([K]o

[K]i

)

s∞ =1

1 + exp

(− (V + 27.79)

7.57

) (2.66)

x∞ = 0.02 +0.98

1 + exp

(V + 69.5

6

) (2.67)

τ s =17

1 +

(V + 20.52

35

)2 (2.68)

τ x = 7.5 +10

1 +

(V + 34.18

120

)2 (2.69)

d s

dt=

s∞ − s

τ s

(2.70)

d x

dt=

x∞ − x

τ x

(2.71)

Parameters Values DefinitiongKA 0.189 nS/pF Maximum conductance of IKA

Table 2.9: Parameter values of IKA

71

-0.4

0

0.4

0.8

1.2

-90 -60 -30 0 30 60

I/Im

ax

V (mV)

AUnpublished, Knock and Aaronson

s∞x∞

4

6

8

10

12

14

16

18

-90 -60 -30 0 30 60

t (m

s)V (mV)

BKnock et al 2001

time to peakSimulation time to peak

4

6

8

10

12

14

16

18

-90 -60 -30 0 30 60

t (m

s)V (mV)

B τsτx

0

0.2

0.4

0.6

0.8

1

1.2

-90 -60 -30 0 30 60

I/Im

ax

V (mV)

C

Knock et al 2001Simulation

0

0.2

0.4

0.6

0.8

1

0 50 100 150

I/Im

ax

t (ms)

D

Knock et al 2001Simulation

-80

60

V (mV)

Figure 2.10: Myometrial current model of IKA . (A) Voltage-dependentsteady-state activation, s∞ and inactivation, x∞ of IKA with experimental data.s∞ was formulated using reported values of half-inactivation (-69.5 mV) and slopefactor (6 mV) from experiments [30]. (B) Simulated (empty points) and exper-imental (filled points) time-to-peak of IKA . Activation time constant, τ s (solidline) and inactivation time constants, τ x (dotted line) of the model are also pre-sented for comparison. (C and D) I-V relationship and current tracings of IKA .The current tracings show IKA activated and inactivated rapidly, this current alsofully inactivated unlike IK1 and IK2 . Both simulated and experimental data werenormalised to their peak current values.

72

2.1.2.4 Calcium-activated K+ current – IKCa

Calcium-activated K+ current, IKCa is a voltage- and calcium dependent current

with a large conductance of around 200 pS. It is inhibited by tetraethylammo-

nium (TEA), 4-AP, barium, scorpion toxic, paxiline, charybdotoxin and iberi-

toxin. Studies suggested that this channel is associated with setting the resting

membrane potential and regulating both the shape and duration of AP. It also

suppresses the excitability and contraction in myometrial cells throughout the

gestation [100, 101]. This channel is a predominate K+ channel in non-pregnant

rat [96] and human [102, 103] myometrial cells. IKCa was also found in mid-

pregnant (14 to 16 days) rat myometrial cells [100]. Although IKCa was also

found in late pregnant myometrial cells [104], the current density of IKCa was

found to be markedly reduced prior to labour [29, 31]. This is because the ex-

pression of IKCa is down-regulated to reduce the importance of this channel and

therefore increase the excitability at parturition. Experimental study reported

that the delayed rectifying potassium currents accounted for almost 90% of the

total K+ current in late pregnant rat myometrial cells and the remaining 10%

were sustained currents consisted of mostly IKCa with an activation threshold of

greater than 0 mV [31].

Quantitative data on this channel is lacking and the majority of the experi-

ments only concentrated on the behaviour in the level of single channels [2, 97].

The formulations of this current were substituted by equations developed for rat

mesenteric smooth muscle [55]. The [ Ca2+ ]i -dependency of this current was

modelled based on an empirical relationship. Studies in myometrium show the

content of cGMP increases with increasing level of estrogen [105], and cGMP

kinase contributes to uterine quiescence in pregnancy [101]. Despite the relax-

ant effects of nitric-oxide (NO) and cGMP on pregnant rat uterine longitudinal

smooth muscle [106], a cGMP-dependent pathway in IKCa was not included in the

model. This is because the descriptions of cGMP formation in experiments were

not sufficient to establish mathematical equations. For clarity, the NO-terms in

the equations describing IKA were removed (NO concentration in the equations

was set as zero and subsequently the NO-related terms in the equations have

no effect on the current). In vascular smooth muscles cells, cGMP is activated

by NO to promote relaxation in cells. The activation time constant and open

probability of IKCa was modelled based on small mesenteric artery from guinea

73

pig [107]. Since all the membrane currents were expressed in pA/pF, for consis-

tency, the units of IKCa was converted from pA which was originally given in the

model by Kapela et al. to pA/pF. This was done by estimating a late pregnant

rat myometrial cell to be 100 pF (a late pregnant rat uterine cell has a cell capac-

itance between 80 to 130 pF in published literatures [26, 72, 108]).The simulated

I-V relationship is shown in figure 2.11 (A).

Calcium-activated potassium current – IKCa

IKCa =Am NBKCa PKCa iKCa

100(2.72)

PKCa = 0.17 pf + 0.83 ps (2.73)

d pf

dt=

po − pf

τpf

(2.74)

d ps

dt=

po − ps

τ ps

(2.75)

po =

(1 + exp

(−V − V1/2KCa

18.25

))−1

(2.76)

V1/2KCa = −41.7log10( [ Ca2+ ]i )− 128.2 (2.77)

iKCa = 104 PBKCa VF2

RT

[K]o − [K]i exp(VF/RT)

1− exp(VF/RT)(2.78)

Parameters Values DefinitionNBKCa 5.94×106 cm−2 Channel densityτpf

0.84 ms Fast activation time constantτps 35.9 ms Slow activation time constantPBKCa 3.9×10−13 cm3/s Single channel permeability

Table 2.10: Parameter values of IKCa

74

2.1.2.5 Background K+ leakage current – IKleak

IKleak is a simple background potassium current with a linear I-V relationship.

This small current was included in the model to regulate and stabilise the resting

membrane potential. The equation and the parameters describing this current

are shown below and in the appendix. Figure 2.11 (B) shows the I-V relationship

of IKleak at different voltages.

Background K+ leakage current – IKleak

IKleak = gK−leak (V − EK ) (2.79)

Parameters Values DefinitiongK−leak 0.005 nS/pF Conductance of IKleak

Table 2.11: Parameter values of IKleak

-0.2

0

0.2

0.4

0.6

0.8

-90 -60 -30 0 30 60

I (pA

/pF

)

V (mV)

A

INSCaINSK

-0.2

0

0.2

0.4

0.6

0.8

-90 -60 -30 0 30 60

I (pA

/pF

)

V (mV)

A

INSNaIKleak

0

0.2

0.4

0.6

0.8

1

-90 -60 -30 0 30 60

I/Im

ax

V (mV)

B[Ca2+]i=800nM

500nM100nM

Figure 2.11: I-V relationship of IKCa , INSCC and IKleak in myometrium.(A) I-V relationships of IKleak and Ca2+ , K+ and Na+ components of INSCC . (B)I-V relationships of IKCa with different values of [ Ca2+ ]i . It shows how changingvalues of [ Ca2+ ]i has an effect on this current. Currents were normalised at thepeak value at 60 mV for [ Ca2+ ]i =800 nM. In both (A) and (B), a voltage-clamped was applied at a holding potential of -80 mV and stepped up to 60 mVevery 1 mV to obtain the I-V relationships.

75

2.1.3 Other currents

In the myometrial cell model, other currents are defined as the currents that

can be flown in both directions, depending on the ionic concentration and the

voltage of the cell. They are the non-specific cation current ( INSCC ) and the

Ca2+ -activated Cl− current ( ICl ). INSCC plays a role in the reconstruction of

pace-making activity while ICl has a functional role in the plateau state of the

AP . These two currents are small but significant to the myometrial cells.

2.1.3.1 Non-selective cation current – INSCC

Non-selective cation current, INSCC is a background linear current identified in late

pregnant rat myometrial cells [33]. INSCC plays an important role in reconstruct-

ing the pace-maker activity, regulating the contractility and is also considered to

be the alternative mechanism for automaticity bedsides Ih in the myometrium.

INSCC is time independent and has a linear I-V relationship. Experiments show ex-

tracellular Mg2+ (treatment for preterm labour) inhibited INSCC and suppressed

contraction. INSCC was inhibited by La3+ and Gd3+ and reduction of extra-

cellular Na+ decreases the amplitude of this current. It was also found that

INSCC diminished near term with an increasing level of estrogen in rat myometrium

[109]. The channel is permeable to a variety of ions such as K+ , Cs+ , Na+ and

Li+, and its relative permeability for K+ : Cs+ : Na+ :Li+ is 1.3:1:0.9:0.8. INSCC also

has a small but detectable permeability for Ca2+ .

Experiments performed by Miyoshi et al. [33] using 18-20 day pregnant rat

single longitudinal cells at room temperature reported the reversal potential was

around -5 mV. The current density under control physiological conditions (with

0.1 mM [ Mg2+ ]o) was 0.6± 0.46 pA/pF at a voltage step of -50 mV. Since INSCC is

sensitive to more than one type of ion (Ca2+ , Na+ and K+ ), the reversal poten-

tial, ENSCC of this current, was therefore dependent on all these ions. ENSCC in

the myometrium model was modified using the Goldman-Hodgkin-Katz (GHK)

equation [61], assuming the ions interact with each other and diffuse through the

channel.

In the myometrium model, the conductance of INSCC depends on the ex-

tracellular concentrations of permeable cations. Experiments recordings from

the voltage ramp I-V relationships presented a ratio of Ca2+ : Na+ : K+ : Cs+ =

0.5:1:1.19:1.6 [33]. Similar to INSCC found in guinea-pig endocardial endothelial

cells [110], the conductance of myometrial INSCC reduced with decreasing [ Na+ ]o .

76

This relationship was described by a Hill equation with a half-saturating concen-

tration of 150 mM and a Hill coefficient of 2. The Hill equation was normalised

with the Na+ conductance at [ Na+ ]o = 125 mM. For Ca2+ ions, the Hill equa-

tion was normalised to the Ca2+ conductance at [ Ca2+ ]o = 20 mM. Inhibition

by [ Mg2+ ]o was described by a Hill equation with a half-saturating concentration

of 0.28 mM and a Hill coefficient of 1.3 [33]. Blockage by La3+ and Gd3+ were

excluded in the current model. Since INSCC was also permeable to other cations, a

small leak component, gleak was needed to compensate and match the experimen-

tal voltage clamp data. Under physiological conditions with 0.1 mM [ Mg2+ ]o ,

the simulated INSCC consisted of mostly Na+ and leak components. Contribution

from K+ and Ca2+ were very small as illustrated in figure 2.11.

Non-selective cation current – INSCC

INSCC = INSCa + INSNa + INSK + INSleak (2.80)

INSCa = 0.15 fMg g( [ Ca2+ ]o ) gNS (V − ENS ) (2.81)

INSNa = fMg g( [ Na+ ]o ) gNS (V − ENS ) (2.82)

INSK = 1.19 fMg g( [ K+ ]o ) gNS (V − ENS ) (2.83)

INSleak = fMg gleak (V − ENS ) (2.84)

g( [X]o ) =1

g s

0.03

1 +

(150

[X]o + 10−8

)2 , g s =

0.000525 for Ca2+

0.0123 otherwise(2.85)

fMg = 0.1 +0.9

1 +

([Mg2+]o

Kd,Mg

)1.3 (2.86)

ENS =RT

F· ln

PNa

PCs

[ Na+ ]o +PK

PCs

[ K+ ]o + PCa′4 PCa

PCs

[ Ca2+ ]o

PNa

PCs

[ Na+ ]i +PK

PCs

[ K+ ]i + PCa′4 PCa

PCs

[ Ca2+ ]i

(2.87)

PCa′ =

PCa

1 + exp

(VF

RT

) (2.88)

77

Parameters Values DefinitiongNS 0.0123 Maximum conductance of INSCC

gleak 0.009685 Maximum conductance of leakagecomponent of INSCC

Kd,Mg 0.28 Half saturation concentrationPCa : PCs 0.89 Permeability ratio of Ca2+ and Cs+

PNa : PCs 0.9 Permeability ratio of Na+ and Cs+

PK : PCs 1.3 Permeability ratio of K+ and Cs+

Table 2.12: Parameter values of INSCC

2.1.3.2 Calcium-activated chloride current – ICl

ICl was identified in myometrium and this current is activated by Ca2+ through

the L-type Ca2+ channel [111]. This current is inhibited by Cl− blockers, fena-

mates (e.g. niflumic acid and flufenamic acid). When the channel blocker drugs

were applied, the frequency of spontaneous contractions significantly reduced.

Since the equilibrium potential for Cl− is close to the plateau of the AP in uter-

ine cell, this current is thought to have a functional role in the plateau state in

myometrium, contributing to the membrane potential and bursting frequency in

cells. Although ICl contributes up to 30% of the inward current in rat pregnant

cell [32], some other reports in contrast suggested the chloride channels are not

important in uterine electrophysiology [12]. Jones et al. [32] investigated ICl in

the myometrium using single longitudinal primary cultured cells from 18-21 days

pregnant rats at 35◦C. Their raw current tracings show ICl flows in both inward

and outward directions depending on the depolarisation voltages and the dy-

namics of both inward (with negative voltage range) and outward (with positive

voltage range) currents behave very differently. The inward component of ICl has

a fast activation and took 3 - 5 ms to reach its peak and a fast inactivation,

while the outward component has a slower activation of around 50 ms and did

not inactivate fully. Therefore, the formulations of this current were developed

by separating the current into positive and negative components of voltage.

Since voltages in positive range inactivate over a long range of time (more

than 200 ms), in the myometrial model, it was assumed that the current does not

inactivate, and only the activation variable was considered. Both activation and

inactivation variables were considered in the negative voltages. Steady-state acti-

vation and inactivation (negative range only) of the model are shown in figure 2.12

78

(A). This was obtained by extracting the raw current traces in the published ex-

perimental recordings [32]. Time constants for both positive and negative ranges

shown in figure 2.12 (B) were extracted by fitting the tail of the current traces

to a single exponential equation. The time constants which are illustrated in

figure 2.12 were then fitted with a sigmoidal function. Inactivation of the inward

current is rapid, and the inactivation time constant was estimated as 0.69 ms.

The Ca2+ dependency of ICl was confirmed in Jones et al. [32], but no quali-

tative data was measured in myometrium. Equations from the Luo-Rudy model

of mammalian ventriclular myocytes [112] were modified to substitute for this de-

pendency. The [ Ca2+ ]i required for half activation in the current, the Kd,Cl term

in equation 2.92 was 0.256×10−3 mM. This value was obtained based on available

experimental data of different cell types and species. Also this value agreed with

the range suggested by Greenwood et al. [86] and Hartzell et al. [113] (0.2×10−3

to 0.5×10−3 mM) in endothelial and epithelial cells, parotid acinar cell and other

types of smooth muscles. A further voltage-dependence conductance function,

g(V) illustrated in 2.12 (C) was introduced in the model for both positive and

negative voltages to compensate for the uncertainties of the Ca2+ dependencies

in ICl .

According to test simulations, the model works best when the activation vari-

able in the positive range, c1 is square-rooted. This might be due to the rapid

activation of the current. Previous test simulations replacing the activation term

with different indices were unsatisfactory (results not shown). Furthermore, ex-

periments [32] and simulations showed that a short pre-pulse (10 ms) greatly

reduced the amplitudes in the negative voltages, this might be due to the cell

not reaching its steady-state values. Therefore in the myometrial current model,

a long (2000 ms) pre-pulse was included to ensure gating variables have reached

steady-state and total current ( ICa and ICl ) reached zero before voltage-clamping

was applied (figure 2.12 (D)). The experimental and simulated ICl tracings are

shown in figure 2.12 (E) and the I-V relationship of ICl is shown in figure 2.12

(F). Both currents were normalised to their peak inward current values at -80

mV.

79

Calcium-activated chloride current – ICl

ICl =

gCl gfCa

√c1(V − ECl ) if V > 0

gCl gfCa c2 n(V − ECl ) otherwise(2.89)

ECl =RT

F zCl

ln

([Cl]i

[Cl]o

)(2.90)

gCl =

gCl (19.26 + 365.8 exp(0.0564V)) if V < −20.0

gCl (0.752 + 0.0349 exp(−0.414V)) otherwise(2.91)

gfCa =[ Ca2+ ]i

[ Ca2+ ]i + Kd,Cl

(2.92)

c∞ =1

1 + exp

(44.08− V

13.7

) (2.93)

n∞ =1

1 + exp

(V + 56.97

20.29

) (2.94)

τ c1 = 26.14− 16.88

1 + exp

(− (V − 48.14)

14.85

) (2.95)

τ c2 = 50.79 +81.54

1 + exp

(− (V + 43.51)

9.5

) (2.96)

τn = 0.69 ms (2.97)

d c1

dt=

c∞ − c1

τ c1

(2.98)

d c2

dt=

c∞ − c2

τ c2

(2.99)

d n

dt=

n∞ − n

τn

(2.100)

80

Parameters Values DefinitiongCl 0.1 pA/pF Maximal conductance of ICl

Kd,Cl 257 nM Calcium required for half activation in ICl

Table 2.13: Parameter values of ICl

Selected major uterine membrane currents and their current characteristics

are summarised in table 2.14. All the values were based on simulated results

obtained from the myometrial model.

2.1.4 Pumps and exchanger currents

Apart from the ionic membrane currents, ions can also be transported across the

cell membrane through other mechanisms for example pumps and exchangers.

In the pregnant myometrium, the Ca2+ pump, Na+ /K+ exchanger, Na+/Ca2+

exchanger and Na+ -K+ - Cl− cotransporter were identified to maintain and reg-

ulate the ionic balance in the cell. Quantitative descriptions of these transporters

are lacking and formulations of their currents were adopted and modified using

various cardiac and smooth muscle cell models. All the exchangers and pumps

are considered to be electrogenic apart from the Na+ -K+ - Cl− cotransporter in

this model. Details of this will be given in the subsequent sections.

2.1.4.1 Calcium pump current – ICapump

The calcium pump or plasma membrane Ca2+ ATP-ase has been characterised

in rat myometrial cells [114–116]. This ionic transporter maintains and regulates

the Ca2+ concentrations between the cytoplasm, sarcoplasmic and endoplasmic

reticulum (SR and ER) in the cells. In a typical cell, free cytosolic calcium

concentration is in the order of 0.1 µM while concentrations in the ER and SR

are in the range of 0.1 to 1 mM [58]. To surmount this three to four order of

magnitude of concentration difference, Ca2+ ions are actively pumped against

the concentration gradient. The energy required during this process is obtained

by utilising the chemical energy store in ATP. In our model, rapid equilibrium

was assumed and calcium pump was assumed to work instantaneously with quick

bindings. The equation describing this current was a empirical, using a Hill-

type formula for the calcium pump rate rather than a set of kinetic differential

equations. The Hill coefficient, hPCa and half saturation of [ Ca2+ ]i , Km,pCa were

81

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50 100

I/Im

ax

V (mV)

A

Jones et al. 2004c∞

0

0.2

0.4

0.6

0.8

1

-100 -50 0 50 100

I/Im

ax

V (mV)

A

Jones et al. 2004n∞

0

20

40

60

80

100

120

140

-100 -50 0 50 100

τ (m

s)

V (mV)

B

Jones et al 2004τc1

0

20

40

60

80

100

120

140

-100 -50 0 50 100

τ (m

s)

V (mV)

B

Jones et al 2004τc2

0

40

80

120

160

-100 -50 0 50 100

g(V

)

V (mV)

C

Jones et al 2004gCl(V≤-20)gCl(V>-20)

-1.2

-0.8

-0.4

0

0.4

0.8

0 0.2 0.4 0.6 0.8 1 1.2

I/Im

ax

t (s)

DSimulation

-1.2

-0.8

-0.4

0

0.4

0.8

1.2

0 50 100 150 200 250

I/Im

ax

t (ms)

E

Jones et al. 2004Simulation

-1.5

-1

-0.5

0

0.5

1

1.5

2

-100 -50 0 50 100

I/Im

ax

V (mV)

F

Jones et al 2004Simulation

-80

40

V (mV)

Figure 2.12: Myometrial current model of ICl . (A) Steady-state activation,c∞ (for both positive and negative ranges) and inactivation, n∞ (positive rangeonly) of ICl with experimental data. (B) Activation time constants for positive,τ c1 and negative ranges, τ c2 of ICl . Inactivation for the negative voltage rangewas fixed at 0.69 ms and is not shown on the figure. (C) Voltage-dependent con-ductance of ICl with experimental recordings. (D) Simulations of the total currentincluding ICa and ICl . A voltage-clamp was applied with a holding potential of0 mV and stepped from -80 to 40 mV every 20 mV. An inward Ca2+ currentwas clearly seen activating ICl and a small tail current was also presented whenthe voltage-clamp was released. (E) Current tracings of ICl with experimentaldata. (F) I-V relationship of ICl with experimental recordings. All the simulatedcurrents were normalised at the peak inward current value at -80 mV.

82

Curr

ent

Hal

fac

tiva

tion

(mV

)H

alfin

acti

vati

on(m

V)

Tim

eto

pea

k(m

s)T

ime

fully

inac

tiva

te(m

s)R

ole

inm

yom

etri

um

I h-8

5does

not

inac

tiva

te10

00does

not

inac

tiva

te

mai

nta

inre

stin

gm

embra

ne

pot

enti

alan

da

auto

mat

icity

mec

han

ism

I CaL

22-3

2.8

6-25

<50

0m

ajo

rco

ntr

ibuto

rto

the

tota

lin

war

dcu

rren

tan

dtr

igge

rsm

yom

etri

alco

ntr

acti

lity

I CaT

-47.

1-7

2.9

4-13

<50

0ge

ner

atio

nof

the

low

-thre

shol

dC

a2+

spik

esan

dA

Ppro

pag

atio

n

I Na

-23.

4-5

7.2

1-14

<12

0re

spon

sible

for

the

fast

upst

roke

ofA

Pge

ner

atio

n

I K1

7.8

-63.

110

0-90

0does

not

fully

inac

tiva

te

render

sth

eA

Pge

ner

atio

nan

dst

opth

eco

ntr

acti

onev

entu

ally

I K2

4.4

-21.

710

0-45

0does

not

fully

inac

tiva

tesa

me

asab

ove

I KA

-27.

6-6

9.4

8.6-

24ab

out

150

ms

todec

ayto

10%

ofit

spea

ksa

me

asab

ove

I Cl

44-5

6.5

pos

itiv

e:2-

4neg

ativ

e50

pos

itiv

e:does

not

inac

tiva

teneg

ativ

e:60

0

contr

ibute

topla

teau

stat

eof

AP

Table

2.1

4:

Chara

cteri

stic

sof

the

majo

rm

yom

etr

ialcu

rrents

.A

llth

ere

sult

sw

ere

obta

ined

bas

edon

sim

ula

tion

sfr

omth

em

yom

etri

alce

llm

odel

.

83

determined and modified based on experimentally obtained values by Grover et

al. [115].

Plasma membrane Ca2+ -ATPase current– ICapump

ICapump =zCa F Vc

Cm BCa Ac

· JpCa

1 +

(Km,pCa

[ Ca2+ ]i

) hPCa(2.101)

Parameters Values DefinitionJpCa 7×10−7 mM/ms Maximum flux of ICapump

Km,pCa 0.5 µN Half saturation of [ Ca2+ ]ihPCa 2 Hill coefficient

Table 2.15: Parameter values of ICapump

2.1.4.2 Sodium/Potassium exchanger current – INaK

The Na+ /K+ exchanger and its current, INaK is vital in regulating the Na+ and

K+ concentration extra- and intracellularly. It is identified in myometrial cells of

late pregnant rats [117–119] and human [25]. In each forward cycle, the Na+/K+

exchanger expels three Na+ ions and uptakes two K+ ions for each hydrolysed

ATP, creating a net outward flux which leaves the cell more negatively charged.

Reports showed INaK in the rat myometrium increased during pregnancy and

peaked at 17 days gestation [119].

Despite evidence of an electrogenic INaK in myometrial cells, there is little in-

formation about its current properties in myometrium. The myometrium model

adopted and modified the equations from the Luo-Rudy mammalian ventricu-

lar model [112] (based on guinea pig ventricular myocyctes data on Nakao and

Gadsby [120]). This current is [ Na+ ]o , [ Na+ ]i , [ K+ ]o and voltage dependent.

The simulated results were then used to validate experimental data from guinea

pig mesenteric resistance arteries cells [121] since smooth muscle cells are much

more similar to the myometrial cell than ventricular cells. The current model

for INaK was developed at 36◦C, a Q10 value of 1.87 for every 10◦C change (be-

tween 26 to 36◦C) was reported for vascular smooth muscle cells by Nakamura et

al. [121]. Simulations of the whole cell model showed this current was necessary to

84

produce a bursting AP since the inhibition of the exchanger dynamically changes

[ Na+ ]i and in turn affects the activities of K+ and K+ -dependent currents (e.g.

INSCC ).

Na+ / K+ exchanger current – INaK

INaK = gNaK fNaK kNaK nNaK (2.102)

fNaK =1

1 + 0.125 exp

(− 0.1VF

RT

)+

0.00219 exp

([ Na+ ]o

49.71

)exp

(− 1.9VF

RT

)

(2.103)

kNaK =1

1 +

(Kd,K

[ K+ ]o

)nK(2.104)

nNaK =1

1 +

(Kd,Na

[ Na+ ]i

)nNa(2.105)

Parameters Values DefinitiongNaK 1.7 nS/pF Maximum conductance of INaK

Kd,K 2 mM Half saturation of K+ in INaK

nK 1.5 Hill coefficient of K+ in INaK

Kd,Na 22 mM Half saturation of Na+ in INaK

nNa 2 Hill coefficient of Na+ in INaK

Table 2.16: Parameter values of INaK

2.1.4.3 Sodium/Calcium exchanger current – INaCa

Na+ /Ca2+ exchanger current, INaCa works in a similar way as INaK . It controls

reversibly and regulates the Na+ and Ca2+ concentration inside and outside the

cell. In each cycle, one Ca2+ ion is pumped out of the cell while three Na+ ions

are transferred in. Research has shown Na+ /Ca2+ exchangers are involved in

calcium translocation in pregnant rat myometrial cells [39, 69, 122, 123]. Studies

85

performed by Abe [124] showed the effects of changing [ Na+ ]o and [ Ca2+ ]o on

the membrane properties in pregnant rat myometrial tissues. It was found that

the prepotential and spontaneous spike discharge was abolished with low level of

[ Na+ ]o but the tissue was still excitable with larger and faster spikes evoked by

depolarising current stimuli. These results explained and confirmed the existence

of the electrogenic INaCa in rat myometrium.

Like the Na+ /K+ exchanger, there is no quantitative study to describe the dy-

namic of this this current in myometrium, but there are cardiac experimental data

and models available. Weber et al. [125] developed a model of Na+ /Ca2+ exchange

current for intact mammalian cardiac myocytes. This model, however does

not describe [ Ca2+ ]i in the cell fully. The INaCa in the myometrial cell model

was based on the description from Hund-Rudy canine ventricular model [126]

(which was developed based on Webers model). The current described by Hund-

Rudy has a more realistic approach and was also sensitive to the changes of the

[ Ca2+ ]i level. The current depends on membrane potential, intra- and extra-

cellular Ca2+ and Na+ concentrations and has a stoichiometry of 3 Na+ :1Ca2+ .

The Na+ /Ca2+ flux, JNaCa was composed of an electrochemical (∆E) and al-

losteric (Allo) factor, assuming the maximal flux was the same in both directions.

It was also assumed that the dissociation constants of [ Ca2+ ]i and [ Na+ ]i from

the catalytic sites are approximately equivalent to the apparent dissociation con-

stant, Km for [ Ca2+ ]i and [ Na+ ]i transport by Na+ /Ca2+ exchange when other

substrates are at their saturating concentrations ( Km,Cai , Km,Nai ) [126]. The

Hill coefficient in this current, nallo which measures the cooperativity is four, in-

dicating four ions were bound to the receptor in the process. Also according

to Weber et al. [125], the gamma factor, γ described the energy barrier of the

Na+ /Ca2+ exchanger in the membrane electric field, ksat controlled the satura-

tion of INaCa at negative potentials and Km,Allo was the Ca2+ activation deter-

mined by refitted with experimental results of calcium decay in late pregnant rat

myometrial cells [69, 123].

86

Sodium/Calcium exchanger current – INaCa

INaCa =JNaCa zCa Vc F

Ac Cm

(2.106)

JNaCa = (Allo)(∆E) (2.107)

∆E =E1

E2(E3 + E4)(2.108)

Allo =1

1 +

(Km,Allo

[ Ca2+ ]i

) nallo(2.109)

E1 = JNaCa

([ Na+ ]i

3 [ Ca2+ ]o expγVF

RT

)−

JNaCa

([ Na+ ]o

3 [ Ca2+ ]i exp(γ − 1)VF

RT

)(2.110)

E2 = 1 + ksat exp(γ − 1)VF

RT(2.111)

E3 = Km,Cao [ Na+ ]i3 + Km,Nao

3 [ Ca2+ ]i +

Km,Nai3 [ Ca2+ ]o

(1 +

[ Ca2+ ]iKm,Cai

)(2.112)

E4 = [ Ca2+ ]o [ Na+ ]i3 + [ Na+ ]o

3 [ Ca2+ ]i +

[ Na+ ]o3 Km,Cai

(1 +

[ Na+ ]iKm,Nai

3)

(2.113)

Parameters Values DefinitionJNaCa 3.5×10−6 mM/ms Maximum flux of Na+ /Ca2+ exchangerKm,Allo 0.3 µM Calcium activation of INaCa

nallo 4 Hill coefficient of INaCa

ksat 0.27 Saturation of INaCa at negative potentialsγ 0.35 Energy barrier of Na+ /Ca2+ exchanger

in the electric fieldKm,Nai 30 mM Saturating concentration of [ Na+ ]iKm,Cai 7 µM Saturating concentration of [ Ca2+ ]iKm,Nao 87.5 mM Saturating concentration of [ Na+ ]oKm,Cao 1.3 mM Saturating concentration of [ Ca2+ ]o

Table 2.17: Parameter values of INaCa

87

2.1.4.4 Sodium-potassium-chloride co-transport current – INaKCl

The Na+ - K+ - Cl− cotransporter (NKCC) exists in almost all types of mam-

malian cells and tissues and this cotransporter has been first identified in rabbit

non-pregnant uterus tissues by Johns and Cutshaw [127]. Further studies con-

firmed NKCC1 (dominate protein of NKCC) were expressed in the mouse my-

ometrium uterus [128]. The NCCC transports one Na+ and one K+ molecule (two

positively charged ions) along with two Cl−molecules (two negatively charged

ions) into or out of the cell. This keeps the cell electroneutral and the stoichiom-

etry of the NKCC is 1Na+ :1K+ :2Cl− . Since all the ions are moved in the same

direction, NKCC is often considered a symporters. Under normal physiological

condition, NKCC has a net influx into the cell since the Na+ and Cl− ions are

both inwardly directed.

Although NKCC is expressed in all types of cells in various organs, the specific

role and contribution of NKCC is largely undefined. However, the main physiolog-

ical function is to maintain the level of [ Cl− ]i above the electrochemical equilib-

rium [129]. In human pregnant myometrium, NKCC was identified [130] but there

is no quantitative description to date about this current in myometrium. There

are some experimental data available in smooth muscle, and formulations were

substituted using equations adopted from the smooth muscle cells model [55,131].

In the myometrial model, the NKCC fluxes depended purely on the concentration

gradients of the Na+ , K+ and Cl− ions. The value of the co-transport coefficient,

LNaKCl was estimated from the resting Cl− balance. The cGMP dependency in

this current was eliminated as previously discussed in IKCa .

Sodium-potassium-chloride co-transport – INaKCl

INaKCl = −RNaKCl zCl Am LNaKCl RFT×

ln

[ Na+ ]o

[ Na+ ]i

[ K+ ]o

[ K+ ]i

([ Cl− ]o

[ Cl− ]i

)2 (2.114)

INaKCl−Na = INaKCl−K = −0.5 INaKCl−Cl (2.115)

88

Parameters Values DefinitionLNaKCl 1.79×10−8 Cotransport coefficient

nmol−2/J · s · cm−2

RNaKCl 1 Cotransport regulation

Table 2.18: Parameter values of INaKCl

2.2 Intracellular dynamics and SR

In the myometrium model, a modified model of the rat mensenteric smooth mus-

cle from Kapela et al. [55] (based on the cerebrovascular smooth muscle model

from Yang et al. [49]) was adopted to describe the intracellular dynamics. Yang

et al. described the SR as a two-compartment fluid system with an uptake and a

release compartment. In the uptake compartment, Ca2+ from the cytosol is trans-

ferred to the SR via sarco/endoplasmic reticulum Ca2+ -ATPase or SERCA pump

( Iup , uptake current), Ca2+ is then transferred across to the release comportment

through diffusion ( Itr ) before releasing back to the cytosol space through the

ryanodine receptor (RyR) current ( Irel ). The release compartment which is de-

scribed by a four-state kinetic model of RyR is responsible for storing Ca2+ prior

its release when RyR is activated. A leakage component was also included in the

Kapela et al. model to prevent the overloading of Ca2+ in the SR.

2.2.1 SR currents, Iup , Itr and Irel

The Michaelis constant of the SERCA pump in Iup , Km, up remained as 1µM

(used in Kapela et al.) instead of 80µM (used in Yang et al.) since this value

is more physiological [55]. In Irel , a leakage parameter, Rleak was included in

the release compartment to measure the leakage of Ca2+ from the SR to prevent

excessive loading and cellular damage. The value of Rleak and Ca2+ release time

constant, τ rel were adjusted from the Kapela et al. model to match the myome-

trial cell data. A scaling factor, Frel of the whole current was also introduced

for the adjustment of the current. The Ca2+ release from the SR (or calcium-

induced calcium release, CICR) through the ryanodine receptor was described

by a four-state kinetic model similar to cardiac ventricular cells [49]. In the my-

ometrial model, a set of differential equations (equation 2.121 to 2.124) was used

to described the dynamics of the four states of RyR. The four-state of the dy-

namic model is shown graphically in figure 2.13. The states of the RyR fractions

89

were described as R00 (free receptors), R10 (receptors with Ca2+ bound to acti-

vation sites), R01 (receptors with Ca2+ bound to inactivation sites) and finally

R11 (receptors with both activation and inactivation sites bound by Ca2+ ) [49].

The system was constrained by the condition: R00 + R01 + R10 + R11 = 1. The

rates of the Ca2+ biding to activation and inactivation sites depends on the value

of rate constants, Kr1 and Kr2 respectively. These values did not depend on each

other and Ca2+ binds faster to activation sites than to inactivation sites in the

model. The unbinding of the activation/inactivation sites from Ca2+ were de-

pending on rate constants K−r1 and K−r2 respectively. Calcium is released from

the internal SR store through the release current ( Irel ) which is dependent on

R10 .

R R

R R

00 10

01 11

K -r2 K -r2K r2 K r2

K r1

K r1

K -r1

K -r1

[Ca ]2+

i

2

[Ca ]2+

i

2

[Ca ]2+

i [Ca ]2+

i

Figure 2.13: Four-state kinetic model of RyR. These states are describedas R00 (free receptors), R10 (receptors with Ca2+ bound to activation sites),R01 (receptors with Ca2+ bound to inactivation sites) and finally R11 (receptorswith both activation and inactivation sites bound by Ca2+ ) [49]. How fastCa2+ binds with the activation/inactivation sites is dependent on the rate con-stants Kr1 and Kr2 and the rates of Ca2+ unbinding the activation/inactivationsites are dependent on K−r1 and K−r2 .

In the myometrial model, a description of Ca2+ concentration in calsequestrin

(CSQN) was also provided in the release compartment. CSQN is a low-affinity/high-

capacity calcium-binding protein located in the SR. It is also the major Ca2+ -

binding protein in cardiac and skeletal muscles [132]. In smooth muscle, experi-

ments conducted by Milner et al. on smooth muscle SR and rat liver ER reported

90

calsequestrin is not uniformly distributed in smooth muscle cells. Calreticuli is

the major Ca2+ -protein and not calsequestrin [133]. Adjustments have been

made to take this factor into account in the myometrial cell model. For consis-

tency, the units all three of the SR currents Iup , Itr and Irel were converted from

pA to pA/pF.

Sarcoplasmic reticulum control and SR currents

Iup = Iup[ Ca2+ ]i

100 ( [ Ca2+ ]i + Km, up )(2.116)

Itr =([ Ca2+ ]u − [ Ca2+ ]r) zCa F volu

100 τ tr

(2.117)

Irel = Frel( R10

2 + Rleak )([ Ca2+ ]r − [ Ca2+ ]u ) zCa F volr100 τ rel

(2.118)

d[ Ca2+ ] u

dt=

Iup − ItrzCa F volu

(2.119)

d[ Ca2+ ]r

dt=

Itr − IrelzCa F volr

(1 +

[ CSQN ] KCSQN

( KCSQN + [ Ca2+ ]r)2

)−1

(2.120)

Parameters Values DefinitionIup 6.68 pA Maximum SR uptake currentKm, up 1 µM Michaelis constant of the SERCA pumpvolr 0.07 pL Volume of the uptake compartmentτ tr 1000 ms Time constant for the internal diffusion in ItrFrel 0.2 Scaling factor of IrelRleak 75.07×10−4 Leakage parameterτ rel 0.015 ms Time constant of IrelKCSQN 0.8 mM Binding affinity of calsequestrin[CSQN ] 15 mM Concentration of calsequestrin in the release

compartment

Table 2.19: Parameter values of Iup , Itr , Irel

91

Ryanodine receptor

R00 = 1− R01 − R10 − R11 (2.121)

d R10

dt= Kr1 [ Ca2+ ]i

2 R00 − ( K−r1 + Kr2 [ Ca2+ ]i ) R10 + K−r2 R11 (2.122)

d R01

dt= Kr2 [ Ca2+ ]i R00 + K−r1 R11 − ( K−r2 + Kr1 [ Ca2+ ]i

2) R01 (2.123)

d R11

dt= Kr2 [ Ca2+ ]i R10 − ( K−r1 + K−r2 ) R11 + Kr1 [ Ca2+ ]i

2R01 (2.124)

Parameters Values DefinitionKr1 2500 mM−2ms−1 Activation rate constantKr2 1.05 mM−1ms−1 Inactivation rate constantK−r1 0.00076 ms−1 Unbinding rate constant from activationK−r2 0.084 ms−1 Unbinding rate constant from inactivation

Table 2.20: Parameter values of ryanodine receptor

2.2.2 Ionic balances

The changes in the intracellular ionic concentration of Ca2+ , Na+ , K+ and

Cl−with time and voltage were considered in the myometrial model. The equa-

tions describing the ionic balances of these four ions are shown in the appendix

(equation 2.125 to 2.132). The calcium balance in the model also accounted for

buffering by calmodulin and other buffers intracellularly and calsequestrin in the

store. Parameters used were based on cardiac Ca2+ dynamic values obtained from

Jafri et al. [134] and the values were modified to fit the experimental [ Ca2+ ]i data

from myometrium. A new factor, FBCa was introduced to dampen the strength

of the buffer and was set to 0.3% of its original value.

92

Ionic balances

ICa−tot = ICaL + ICaT + INSCa + ISOCCa − 2 INaCa + ICapump (2.125)

d [ Ca2+ ]i

dt= −( ICa−tot + Iup − Irel )

Ac Cm BCa

Vc zCa F×

FBCa

(1 +

[ SCM ]Kd

(Kd + [ Ca2+ ]i )2+

[ BF ] Kd,B

( Kd,B + [ Ca2+ ]i )2

)−1

(2.126)

INa−tot = −0.5 INaKCl−Cl + INa + ISOCNa +

3( INaK + INaCa ) + INSNa + PNa · Ih (2.127)

d [ Na+ ]i

dt= − INa−tot

Ac Cm

Vc F(2.128)

IK−tot = INaKCl−K + IK1 + IK2 + IKA + IKCa + INSK + IKleak − 2 INaK

(2.129)

d [ K+ ]i

dt= − IK−tot

Ac Cm

Vc F(2.130)

ICl−tot = INaKCl−Cl + ICl (2.131)

d [ Cl− ]i

dt= − ICl−tot

Ac Cm

Vc F zCl

(2.132)

Itot = ICaL + ICaT + INa + IK1 + IK2 + IKA + IKCa + Ih + ICl +

INSCC + INaK + INaCa + IKleak + ISOC + ICapump (2.133)

dV

dt=− Itot + Ist

Cm

(2.134)

Parameters Values DefinitionFBCa 0.003 Fractional Ca2+ buffer[ SCM ] 0.3 mM Total concentration of calmodulin sites

for Ca2+

[ BF ] 0.2 mM Total concentration of other buffer sitesfor Ca2+

Kd 2.6× 10−4 mM Dissociation constantKd,B 5.29810−4 mM Dissociation constant in buffer

Table 2.21: Parameter values of the ionic balances

93

2.3 Force production in myometrium

The rise of [ Ca2+ ]i in a uterine cell initiates force production and contraction in

the myometrium. Smooth muscles have unique properties that are not observed in

other muscles despite basic function similarities. These similarities include the ex-

istence of actin and myosin in all types of muscles cells and using ATP as a fuel of

energy. Also Ca2+ is vital in supplying the excitation-contraction coupling in me-

chanical production in all types of muscles [52]. The contractile proteins involved

in smooth muscles cells are different from striated and cardiac muscles. Smooth

muscle cells do not contain troponin and contractions are generated through the

calcium-activated phosphorylation and not by calcium binding to troponin like

other muscle types. In vascular smooth muscles, the length of the myosin fila-

ment is significantly longer compared to the ones in striated muscles (1.4 times

longer). Since the lengths of the cross-bridge are the same in both smooth and

striated muscles, the tension generated per filament by vascular smooth muscle

is about 40% higher. The cross-bridges in smooth muscles are parallel arranged

with equal spacing, therefore the force produced by the muscles is proportional to

the length of the myosin fibre [52]. Furthermore, force in smooth muscles can be

retained when [ Ca2+ ]i and the level of phosphorylation are relatively low unlike

skeletal muscles [49]. The two intracellular processes leading to the production of

contractile force in myometrial smooth muscles is shown in figure 2.14. The first

pathway is initiated by the increase of the intracellular content of Ca2+ through

mainly the L-type Ca2+ channel, [ Ca2+ ]i then bind with calmodulin activating

the MLCK process. It further initiates the phosphorylation and cross-bridge cy-

cling causing mechanical contractions [36]. Ca2+ release from the SR through IP3

and ryanodine receptors is the second pathway causing contraction. However, ev-

idence obtained from various species has suggested that little is contributed from

these receptors [135–137].

2.3.1 Kinetic cross-bridge cycling model in smooth mus-

cles

There are various mathematical models developed for different types of smooth

muscles to describe mechanisms and stages leading to force production. Peter-

son developed a simple model of smooth muscle myosin phosphorylation and

dephosphorylation [138]. Kato and Ogasawara further improved the model and

94

Figure 2.14: Two pathways leading to mechanical contraction in my-ometrium. Contraction in myometrium can be initiated by two pathways:1) Ca2+ entry through voltage-gated Ca2+ channels (predominately the L-typeCa2+ channel) increases Ca2+ inside the cell, [ Ca2+ ]i binds with calmodulin acti-vates the myosin light chain kinease (MLCK). It then allows the phosphorylationand cross-bridge cycling causing contraction when myosin binds with actin. 2)Ca2+ is released from the sarcoplasmc reticulum (SR) through IP3 and ryanodinereceptors, triggering mechanical contractions in the uterus. However, experimen-tal evidence obtained from both human and animals has suggested contributionsfrom the receptors is minimal [135–137]. Muscles relaxation in myometrium isachieved through myosin light chain phosphatase (MLCP). The section high-lighted by the dashed line was described mathematically using the kinetic cross-bridge cycling model.

95

construed a kinetic model for isometric contraction in general smooth muscle on

the basis of the myosin phosphorylation hypothesis [139]. Hai et al. proposed

a four-state latch-bridge hypothesis (commonly known as the Hai and Murphy

model) [140] and the formation of the myosin phosphorylation and latch-bridge is

illustrated in figure 2.15. This is the equivalent of the highlighted section shown

in figure 2.14.

Figure 2.15: Four-state kinetic model of cross-bridge cycling in smoothmuscle. The rise of Ca2+ inside the cell binds with calmodulin which activatesMLCK and in turn initiate the cross-bridge cycling process. The four statesare M: free cross-bridges, Mp: phosphorylated cross-bridges, AMp: attachedphosphorylated non-cycling cross-bridges and AM: attached dephosphorylatednon-cycling cross-bridges (as latch bridges), which only exist in smooth muscles[49, 141]. Each of the states are described by their rate constants, K1 whichequals to K6 are [ Ca2+ ]i dependent and describe the MLCK process, while K2

has the same value of K5 and represents the process of MLCP. K7 is the slowdetachment rate constant, this process does not change force generation capacitiesand therefore leads to relaxation.

The four-states describing the formation consists of free cross-bridges (M),

phosphorylated cross-bridges (Mp), attached phosphorylated non-cycling cross-

bridges (AMp) and attached dephosphorylated non-cycling cross-bridges (as latch

bridges (AM)), which only exist in smooth muscles [49]. Actin is bound to Mp

96

state and only this phosphorylated cross-bridges can bind to actin. Dephospho-

rylation is due to the action of MLCP, myosin light chain phosphatase. Both

cross-bridge cycle and phosphorylation/dephosphorylation are powered by the

hydrolysis of ATP. The transitions of each of the states are described by rate

constants; K1 has the same rate as K6 which represents the [ Ca2+ ]i -dependent

MLCK process, and K2 has the same rate as K5 which describes the process

of MLCP [142]. The rate constant K7 is known as slow detachment rate - this

process does not change the capacities of the force generation. The four kinetic

states are subjected to constraint: M + Mp + AMp+ AM = 1. The fraction of

phosphorylated myosin is described by the sum of Mp + AMp while the attached

cross-bridges or fraction stress are described by the sum of AMp + AM.

The Hai and Murphy model has been improved over the years by the same

research group and transformed as a multiple-state model [142–144]. But the

original Hai and Murphy model is still commonly used because of its simplic-

ity and it has also formed the basis of a few mathematical models in various

smooth muscles. This includes the cerebrovascular smooth muscle cell model de-

veloped by Yang et al. [49], the contraction model in the arterial wall by Bennet

et al. [145] and the 12-state MLCK/phosphatase control in the airway smooth

muscle model by Fajmut and Brumen [146]. Bursztyn et al. also constructed a

simple excitation-contraction myometrial cell model describing the intracellular

Ca2+ and ionic flux control leading to tension development.

In the presented myometrial cell model, formulations from Yang et al. were

used and modified to fit the myometrial data from experimental recordings. The

value of the rate constants and the equation describing the rate constant of the

phosphorylation process changing from M to Mp, K1 was obtained from the

Bursztyn et al. model [38] (based on the minimal model of the arterial wall by

Parthmios et al. [147]). K1 has the same value of rate constant changing from

AM to M, K6 as discussed. The unit of the rate constants were provided in s

and was converted to ms in the myometrial model. It was assumed that Ca2+ -

calmudolin-MLCK dependent is the only source of force activation mechanism

in the cell (other mechanisms include protein kinase C (PKC) also contribute

to force activation without changing the intracellular content of Ca2+ and were

therefore not included).

97

Kinetic cross-bridge cycling model

dM

dt= −K1M + K2Mp + K7AM (2.135)

dMp

dt= K4AMp−K1M− (K2 + K3)Mp (2.136)

dAMp

dt= K3Mp + K6AM− (K4 + K5)AMp (2.137)

dAM

dt= K5AMp− (K7 + K6)AM (2.138)

MLCK =MLCKmax

1 +(

[ Ca2+ ]iKMLCK

) pM(2.139)

K1 = CmsMLCK

1 +(

KCa,MLCK

[ Ca2+ ]i

) nM(2.140)

K6 = K1 (2.141)

Fraction phosphorylation = AMp + Mp (2.142)

Fraction stress = AMp + AM (2.143)

Parameters Values DefinitionMLCKmax 0.84 Maximum level of MLCKKMLCK 721.746 nM Half activation of MLCKpM 1 Hill coefficient for MLCKK2 0.0012387 ms−1 Myosin dephosphorylation rate constantK3 0.0001419 ms−1 Cross-bridge formation rate constantK4 0.00035475 ms−1 Cross-bridge detachment rate constantK5 0.001238 ms−1 Myosin dephosphorylation rate constantK7 3.78×10−05 ms−1 Latch state detachment constantCms 0.001 Conversion parameter from s to msnM 8.7613 Hill coefficientKCa,MLCK 256.98 nM Half activation of MLCK set by [ Ca2+ ]i

Table 2.22: Parameter values of the kinetic cross-brodge cycling model

98

2.3.2 Mechanical production in smooth muscles

The number of mathematical model describing mechanical production specifically

for smooth muscles and cells are relatively fewer compared to cardiac and skele-

tal muscles. Gestrelius and Borgstrom constructed a rat portal vein mechanical

model (based on Hill mechanical muscle model [148]) describing the active and

passive force generation, force-velocity and length-force relationships [48]. Carl-

son and Secomb developed a theoretical model to reproduce the response of vessels

to changes in intravascular pressure. It also characterised the active and passive

length-tension relationship in the model [149]. A model investigated the active

tone and passive viscoelastic behaviour undergoing large deformations for general

smooth muscle tissue was proposed by Kroon [150]. The model expanded Hills

three-component model and the mechanical part of the model consisted of sim-

ple active and passive components contributing to total strain energy. The same

research group also studied the mechanochemical response of vascular smooth

muscle. The model focused on the response of the actinmyosin complex and its

force generation. The mechanical section was also based on Hills three-component

model with one internal state variable describing the contraction/relaxation of the

contractile compartment. Stalhand et al. published a review paper detailing the

interaction between mechanical and biochemical components in smooth muscle

cells derived from the constitutive laws of thermodynamics [151]. Numerical ex-

amples were also provided to determined various constants using the response

curve for the stress and fitted to the experimental isometric tension-length rela-

tionship.

In the myometrial cell model, a cerebrovascular smooth muscle model de-

veloped by Yang et al. was used to described the tension/stress development.

Formulations and parameters were modified to match the experimental data in

myometrium [49]. The Yang et al. model was constructed based on a modified

Hill model [52] using experimental recordings from isolated single toad stomach

smooth muscle cells [152–154]. A representation of the mechanical cell model

described by Yang et al. is shown in figure 2.16. The total myometrial force cre-

ated by the cell was made up of different force components and they all coupled

together generating tension and stress. The active force of the cell was produced

by the attached cross-bridge mechanisms described in the previous section and

the cell walls were attached to a viscous-elastic element with a dashpot-like com-

ponent creating the damping effect in the cell.

99

Figure 2.16: The mechanical model of a smooth muscle cell. This me-chanical model was adapted from the Yang et al. cerebrovascular smooth musclecell model [49] which was constructed based on a modified three-component Hill’smodel [52, 155]. The total myometrial force generated by the cell was comprisedof various force components including the passive force, Fp , cross-bridge elasticforce, Fx , active force, Fa and series visco-elastic force, Fs . All the force com-ponents coupled together generating tension and stress of the whole cell. Boththe cross-bridge elastic and the active components were coupled to form a con-tractile component which describe the sliding of the myosin and actin filamentaction. Fp was dependent on the total length of the cell, lc and the length atwhich no passive force was created, ls0 . The elasticity of the passive force com-ponent was described by a spring constant, kp with an exponential force-lengthrelationship. In the cross-bridge elastic component, cross bridges in the cell wereacting together in series and Fx was dependent on the extension length of thecross-bridge, lx and the length of the active contractile component, la whichalso represented the length when myosin and actin filaments overlapped. Themaximum force was achieved when the overlapping of the filaments reached theoptimal length, lopt . Fa was characterised by the friction constants for phos-phorylated cross-bridge and latch bridges, fAMp and fAM and Fs was describedby an elastic Voigt body consisting a spring and a dashpot. The spring and thedashpot were described by spring constant, ks and coefficient of viscoelasticity,µs . The dashpot was acted as a shock absorber to absorb the impact caused bythe spring.

100

2.3.2.1 Passive force

The passive force of the cell, Fp was dependent on the total length of the cell, lc

and the length of the cell when no passive force was created, lc0 . The elasticity

of the cell was described by a spring constant, kp with an exponential force-

length relationship. In an isometric condition, the value of lc was set at 123µm

(provided by Yang et al. model) or 150µm depending on the effect investigated.

Although the length of a late pregnant myometrial cell in rat is about 80 µm to

700 µm [27,72,156,157], other parameter values for example: lopt , ls0 , were not

available experimentally for the uterus cell. In order to use the model by Yang et

al., parameters were kept unchanged when myometrium data were not available.

2.3.2.2 Cross-bridge elastic force

In a single smooth muscle cell, Warshaw et al. determined (using a plot of

cell length against peak isometric force) that the stiffness of the cross-bridges is

about 1.2 to 1.5 times greater than the stiffness of a whole cell [152,154]. It was

also reported that the cross-bridges with an elastic element have an exponential

length-force relationship. Cross bridges were also acting together in series in the

cell. In the mechanical model, the force produced by the cross-bridges, Fx is

described in 2.149. Fx was dependent on both the extension length of the cross-

bridge, lx and the length of the active force component, la which also represented

the length when myosin and actin filaments overlapped. Maximum force can be

achieved when the overlapping of myosin and actin filaments reached the optimal

muscle length, lopt in the active force component. The effect of the overlapping

of the filaments on the active force was estimated by a Gaussian function. Fx

was also described by two types of cross-bridges: cycling (AMp) and latch bridges

(AM) which are characterised by their maximal stiffness factor kx1 and kx2 .

2.3.2.3 Active force

The active force, Fa generated by active force component is described in equation

2.150 and the contractile component is shown in detail in figure 2.17, indicating

the cross-bridge process in the cell. Fa was characterised by the friction constants

for phosphorylated cross-bridge and latch bridges, fAMp and fAM .

101

Contractile component

Cross-bridge

Myosin

l lx a

l

vx

.a

Figure 2.17: The contractile component describing the actin andmyosin filament sliding action. vx is the average velocity of the cross-bridgedisplacement and la is contraction velocity the cell (or the shortening of themuscle) [48].

102

2.3.2.4 Series viscoelastic force

The series viscoelastic force, Fs in the cell model was described by an elastic

Voigt body [52] which contained an elastic element with a spring connected in

parallel to a dashpot. The spring and the dashpot were characterised by spring

constant, ks and coefficient of viscoelasticity, µs . A linear spring in the body

could create deformation proportional to the load while the dashpot acted as a

shock absorber to compensate the effect. Both the spring and the dashpot have

the same displacement in the body and the total force produced by this body was

the sum of both the elastic and viscous force. The spring was expressed with an

exponential length-force relationship obtained from Warshaw et al. [154].

2.3.2.5 Total force produced by the cell

At any instant, the cross-bridge, active, and series viscoelastic force components

were all equal to each other (top section of the mechanical model in figure 2.16),

i.e. Fx = Fa = Fs . Therefore the total force, Ft produced by the whole cell

was Ft = Fa + Fp .

103

Mechanical production model

d ls

dt=

1

µs

( Fs − ks (exp( αs ( ls − ls0 )/ ls0 )− 1)) (2.144)

d la

dt=

Fa exp(β(( la − lopt )/ lopt )2)− ( fAMp AMp vx )

fAM AM + fAMp AMp(2.145)

d lx

dt=

( kx1 AMp + kx2 AM)( lc − la − ls )− ( fAMp AMp vx )

fAM AM + fAMp AMp(2.146)

lx = lc − la − ls (2.147)

Fp = kp (exp( αp ( lc − lc0 )/ lc0 )− 1 (2.148)

Fx = ( kx1 AMp + kx2 AM) lx exp(−β(( la − lopt )/ lopt )2) (2.149)

Fa = ( fAMp AM + fAMp AMp)d la

dt+ ( fAMp AMp vx ))×

exp(−β(( la − lopt )/ lopt )2) (2.150)

Fa = µs

d ls

dt+ ks (exp( αs ( ls − ls0 )/ ls0 )− 1) (2.151)

Fa = Fs = Fx (2.152)

Ft = Fa + Fp (2.153)

2.4 Solutions of the model equations

The model equations are numerically solved in C using Euler’s forward integration

scheme to solve the differential equations. All the simulations in the model were

produced with the time step of 0.01 ms. Voltage-clamp and current-clamp are

applied about 10 ms to 10 s after simulations begin to allow numerical values

of the equations to settle and reach steady-state (the pre-clamped time depends

on the effects investigated). The model also followed experimental conditions

such as ionic concentration and voltage protocol as closely as possible in order to

reproduce the experimental results.

The aim of the model was to provide better understanding of the function

of the uterus in the late pregnant cells. The single cell model reconstructed

the chemical, electrical and mechanical phenomena generated by the cells seen in

experiments. This will also provide an important foundation in further developing

104

Parameters Values Definitionlc 123 or 150* µm Length of the whole celllc0 40 µm Length of the cell at zero passive forcels0 30 µm Length of series viscoelastic component at

zero forcelopt 100 µ Optimal length of active contractile com-

ponentkx1 12.5 µN/µm Phosphorylated cross-bridge stiffness con-

stantkx2 8.8 µN/µm Latch bridge stiffness constantks 0.2 µN Series element stiffness constantkp 0.1 µN Parallel element stiffness constantvx 5 µm/ms Cross-bridge cycling velocityfAMp 1.3 µN ms µm −1 Friction constant for phosohorylated

cross-bridgesfAM 85.5 µN ms µm −1 Friction constant for latch bridgesµs 0.01 µN ms µm −1 Viscosity coefficient of series elementαs 4.5 Length modulation for series viscoelastic

elementαp 0.1 Length modulation for passive elementβ 7.5 Length modulation constant for active

and cross-bridge elements

Table 2.23: Parameter values of the mechanical model

105

a realistic multi-dimensional uterus model for the human.

106

Chapter 3

Results I – Electrophysiological

response

The single cell model was validated by comparing simulated APs and I-V curves

with existing experimental data under control conditions. The model was also

tested by pacing rapidly to investigate the response of both [Ca2+] i and ICa− tot

against the experimentally available data. The model was further validated by

simulating the behaviour of estradiol effect during voltage or current clamp. The

behaviour of all the currents (both membrane and SR) during a 2-s current pulse

was examined. In order to have a deeper understanding on how individual cur-

rents behave and affect a single cell in terms of AP and whole cell membrane

current, application of the well-validated model was used to study the effect of

blocking the individual current during both current and voltage clamp. Fur-

thermore, the model’s ability to reproduce different shapes of AP seen in other

smooth muscle cell was investigated. This shows the model is versatile and can

potentially be adapted for different gestation of myometrial cells or even types of

smooth muscle cells.

3.1 Whole cell membrane current and AP in

control condition

Under control physiological conditions described in Okabe et al. [71], a current-

clamp of 2 s was applied to examine the AP (figure 3.1). The AP showed bursting

107

characteristics with spikes similar to experimental recordings [71, 108]. The cor-

responding [ Ca2+ ]i displayed a step-like feature rather than spikes similar to the

AP. A summation of [ Ca2+ ]i during current-clamp was clearly seen. At equilib-

rium, the resting membrane potential (RMP) of the model was -59 mV (compared

to experimental values of -48 to -68 mV in rat [20, 71, 158] and -70 to -55 mV

in human during late pregnancies [4]). The AP reached a maximum ( Vmax ) of

6.9 mV in 86.1 ms with a maximum rate of rise (dV/dt) of 2.69 V/s (compared

to the experimental value of 5-10 V/s [73]). The duration of AP from dV/dt to

50% repolarisation from Vmax , APD50 was 35.43 ms which agrees well with the

experimental value of 34.4 ± 9.6 ms [156]. [ Ca2+ ]i peaked at 584 nM and took

about 5 s to return to its resting level which was 234 nM.

To investigate the whole cell membrane current in control condition, a voltage-

clamp with a holding membrane potential of -50 mV was applied to the myome-

trial model. I-V of peak outward currents, inward currents and current traces are

showed in figure 3.2. The simulated total current reached its maximum inward

and outward peaks at around 5-10 ms and 125-150 ms respectively. The I-V

curves match qualitatively with the experimental data despite a slight discrep-

ancy on the peak inward current (inward current peaked at -5 mV in simulation

and 0 mV in experiment).

3.1.1 [ Ca2+ ]i and ICa−tot responses on repetitive depolari-

sation

A train of 10 depolarisation pulses, -80 mV to 0 mV for 100 ms at 3 Hz, was ap-

plied to the model to investigate the response of both ICa−tot and [ Ca2+ ]i during

rapid pacing. In experimental studies conducted by Shmigol et al. [69, 123], it is

clear that accumulation of [ Ca2+ ]i was observed when the myometrial cell was

paced with repetitive depolarisations. Each individual Ca2+ transient did not

have time to recover and a spike-like effect was recorded. This is because Ca2+ is

released into the cell at a faster rate, but is slowly being removed. The amplitude

of ICa−tot on the other hand decreases gradually during this process despite the in-

creasing level of [ Ca2+ ]i . This is due to extracellular Ca2+ -induced Ca2+ release

from the SR store via the ryanodine receptors. The model successfully replicated

these effects qualitatively seen in experimental recordings and is shown in figure

3.3.

108

-0.5

0

0 2 4 6 8 10

I st (

pA/p

F)

t (s)

100

300

500

[Ca2+

] i (n

M)

-60

-40

-20

0

20V

(m

V)

Control

1 s 1 sOkabe et al 1999a

Inoue et al 1999

0 mV

-58 mV

Figure 3.1: AP and [ Ca2+ ]i in control condition. A single stimulus cur-rent pulse ( Ist ) from 0 to -0.5 pA/pF was applied for 2 s to the myometrialmodel in control physiological condition. The AP from the simulation (top)showed characteristics which resemble experimental data shown inset (the dura-tion of the current clamp are marked by a horizontal bar). The AP bursted withspikes throughout the duration of the clamp. The corresponding [ Ca2+ ]i (middle)showed an accumulation of [ Ca2+ ]i during the entire current clamp.

109

-8

-4

0

4

8

12

-50 -30 -10 10 30

I tot (

pA/p

F)

V (mV)

Control

Okabe 1999aOkabe 1999b

Simulation

-8

0

8

0 100 200 300 400

I tot (

pA/p

F)

t (ms)

-50 20

mV

Figure 3.2: I-V and current traces of the whole cell in control condition.I-V of maximal inward and outward whole cell membrane current, Itot (top) undercontrol physiological condition. In simulations, the model was voltage-clampedfrom a holding potential of -50 mV and stepped up to 30 mV every 5 mV for 300ms. The I-V curve was compared to the experimental data shown in filled circlesand triangles [26, 71]. The corresponding maximal inward and outward currenttraces (bottom) shows consistency with the experimental recordings.

110

100

300

500

700

[Ca2+

] i (n

M)

A

500 nM

0 nM

10 s

Shmigol 1998a

Shmigol 1998b

-12

-8

-4

0

I Ca-

tot (

pA/p

F)

-80 20

0 2 4 6 8 10 12 14

V (

mV

)

t (s)

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1

0 2 4 6 8 10

Nor

mal

ised

I Ca-

tot

Pulse number

B

Exp ICaSim ICa

Figure 3.3: [ Ca2+ ]i and ICa−tot responses during repetitive depolarisa-tion. (A) Intracellular Ca2+ (top) and ICa−tot (middle) responses during 10 de-polarisation pulses from a holding potential of -80 mV to 0 mV for 100 ms a timebetween the successive pulse of 330 ms (bottom). [ Ca2+ ]i peaked at around 600nM and has shown the same accumulation of [ Ca2+ ]i during rapid pacing as seenin experimental results (insets). Note the difference in the [ Ca2+ ]i magnitude,although both sets of the data were obtained under the same experimental con-dition. The amplitude of ICa−tot gradually declined as [ Ca2+ ]i built up due toCICR from extracellular space. (B) Normalised ICa−tot against number of pulse,both experimental and simulation results show a general trend of gradual declinewith increasing pulse number ( ICa−tot was normalised to the value of the firstpulse).

111

3.2 Whole cell membrane current and AP under

estradiol effect

Estradiol is a natural sex hormone found in both female and male mammalian

cells and organs for example: brains, livers, bones, blood vessels and reproductive

organs. Prominently presented in female and less in male, estradiol together

with estrone and estriol aid uterine blood flow and myometrial growth during

pregnancy. It also stimulates breast growth, producing milk and softens cervical

tissue at term. In human myometrium, the serum level of 17β-estradiol increases

by up to 10 times the normal level and reaches 30 to 100 nM near term [159].

Estradiol is one of the key factors which affects the contractility of the uterus,

regulation of estradiol in the myometrium is therefore absolutely vital during

pregnancy. Studies showed that in the presence of estradiol, spontaneous AP

burst and muscle contractions were suppressed [160]. Voltage-dependent calcium,

calcium- and voltage-dependent potassium were inhibited under estradiol effect

and this attributes to the prevention of spikes in AP [71].

To simulated the estradiol effect, conductances of Voltage-dependent calcium,

calcium- and voltage-dependent potassium were reduced and are listed in table

3.1. Figure 3.4 showed in estradiol cell, spikes were suppressed when a stimulus

current from 0 to -0.5 pA/pF was applied as described in experiments [108,160].

Only the first spike was seen which reached Vmax of -7 mV in 93 ms, followed by a

long plateau, reaching around -23 mV until returning to its a higher RMP of -56

mV. APD50 was 107 ms which was longer compared to control condition. The

corresponding [ Ca2+ ]i peaked at a lower level of 462 nM, and has lost the step-

like feature seen in control, instead, a smooth crescent-like profile with summation

of [ Ca2+ ]i during the clamp was presented.

Similar to in the control condition, in order to investigate the effect of estradiol

on the whole cell membrane current, a voltage-clamp was applied from a holding

potential of -50 mV to the myometrial model with estradiol adjustments. The

peak I-V outward current, inward current and current traces are showed in figure

3.5. Under the effect of estradiol, the amplitudes of both inward and outward

membrane currents were greatly reduced due to the reduction in K+ currents.

112

-0.5

0

0 2 4 6 8 10

I st (

pA/p

F)

t (s)

100

300

500

[Ca2+

] i (n

M)

-60

-40

-20

0

20

V (

mV

)

Estradiol

1 sInoue et al 1999

0 mV

-58 mV

Figure 3.4: AP and [ Ca2+ ]i under effect of estradiol. A single stimuluscurrent pulse ( Ist ) from 0 to -0.5 pA/pF was applied for 2 s to estradiol treatedcell. The parameters needed to simulate the estradiol cell are shown in table 3.1.Simulations of the AP under estradiol effect (top) matched the experimental datashown inset qualitatively. The spikes of the AP were suppressed in the estradiolcell unlike in control condition. Only the first spike was seen followed by a longplateau until returning to its RMP. The corresponding [ Ca2+ ]i (middle) showedan accumulation of [ Ca2+ ]i during the entire current clamp similar to controlcondition but the step-like profile has been smoothed-out under the effect ofestradiol.

113

Voltage-operated potassium currents

gK1 reduced by 30%gK2 reduced by 50%gKA reduced by 60%Calcium-operated potassium currents

gKCa reduced by 60%Voltage-operated calcium currents

gCaL reduced by 60%gCaT reduced by 60%

Table 3.1: Changes made to stimulate cell under estradiol effect.

3.3 Behaviour of all the membrane and SR cur-

rents during current-clamp

Behaviour of of all the membrane and SR currents were examined individually

during a current-clamp using the well validated myometrial model. A stimulus

pulse of 0 to -0.5 pA/pF was applied for 2 s to generate an AP while activ-

ities of all the currents were recorded. This was particularly beneficial when

developing and validating the whole cell model. Comparison of all the currents

side-by-side provided useful information about magnitudes and kinetics of all the

currents. This aided the adjustment and modification of parameters for example:

conductances and activation/inactivation kinetics of a particular current while

constructing the model.

3.3.1 Ca2+ -related currents

In simulations illustrated in figure 3.6, the profile of INSCa and ISOCCa (the cal-

cium component of INSCC and ISOC ) closely matched the shape of the AP during

the current-clamp. These currents shared the same features as the AP, which

showed that the currents have a strong correlation with the membrane potential.

Voltage-dependent calcium channels are activated when the cell membrane poten-

tial is depolarised. The simulated results showed that ICaL had an inverted profile

of the AP with its peaks strongly linked to the peaks of the AP. ICaT however

only matched the initial peak, but the subsequent peaks did not. INaCa and

ICapump displayed a summation of current as time progressed. After reaching

their peaks, both currents decay to their equilibrium level with ICapump decaying

114

-8

-4

0

4

8

12

-50 -30 -10 10 30

I tot (

pA/p

F)

V (mV)

Estradiol

Okabe 1999bSimulation

-8

0

8

0 100 200 300 400

I tot (

pA/p

F)

t (ms)

-50 20

mV

Figure 3.5: I-V and current traces for the whole cell under the effectof estradiol. I-V of maximal inward and outward whole cell membrane current,Itot (top). The simulations fit well with the experimental data obtained by Okabeet al. [26]. The amplitude of the whole cell outward and inward currents (bottom)were reduced compared to results in control condition shown in figure 3.2 asexpected due to the reduction of the K+ currents. The protocol of the whole cellcurrent are shown in the inset.

115

at a faster rate than INaCa . The decay rate of ICapump was dependent on the

magnitude of the pump. The current showed a steeper decay as the magni-

tude of ICapump increased. Since the peak of APs are closely separated, INaCa and

ICapump did not have time to recover and Ca2+ therefore did not have time to

leave the cell. This caused a build-up in these current seen in simulated results.

The magnitude of the Ca2+ -related currents revealed that ICaL is the major con-

tributor to ICa−tot (see definition of ICa−tot in equation 2.125) during the AP and

ISOCCa only accounts for a small proportion. After the AP repolarised to its RMP,

ICa−tot displayed a small but noticeable tail outward current. This is closely linked

to ICapump as Ca2+ was being restored.

3.3.2 Na+ -related currents

As the AP depolarised, Na+ channels activated and there was a rapid influx

of INa seen in figure 3.7, however only the initial peak of INa matched the first

peak of the AP. Ih−Na was not active when the stimulus pulse was applied as

expected, since Ih (responsible for automaticity in the cell) is only activated at

RMP. Ih−Na reached about 0 pA/pF during the current-clamp and slowly re-

turned to stability. ISOCNa , INSNa and INaK were strongly related to the mem-

brane voltage since the morphology of these currents resembled the shape of the

AP. INaKCl−Na did not seem to contribute greatly to INa−tot , the total INa (see

equation 2.127 for definition) and it did not show a strong correlation with the

changing membrane potential. The main contributors during the AP are INa ,

INSCa and INaK . After INa−tot reached its maximum current at the end of the 2

s current-clamp, it slowly returned to its equilibrium value. This is the direct

result of the balance between both INaK and INSNa .

3.3.3 K+ -related currents

As shown in figure 3.8, all the profiles of the K+ current, IK1 , IK2 , IKA , IKleak and

INSK closely resembled the shape of the AP apart from Ih−K and INaKCl−K . Ih−K

behaved the same way as Ih−Na , as described in the previous section with an

increased current magnitude. Ih−K was inactivated during AP since Ih only ac-

tivated at RMP. INaKCl−K has the exact shape and magnitude as INaKCl−Na since

this current is equal to INaKCl−K by definition as explained in equation 2.115 (not

shown). Since IK1 and IK2 are the major contributors to the total K+ current,

116

0.02

0.06

0.1

0.14

0 2 4 6 8 10

I (pA

/pF

)

t (s)

ICapump

-4

-2

0

0 2 4 6 8 10

t (s)

ICa-tot

-2e-4

-1e-4

0

1e-4

0 2 4 6 8 10

I (pA

/pF

)

INSCa

-3.8e-6

-3.4e-6

-3e-6

-2.6e-6

-2.2e-6

0 2 4 6 8 10

ISOCCa

-0.8

-0.6

-0.4

-0.2

0 0 2 4 6 8 10

INaCa

-5-4-3-2-1 0

0 2 4 6 8 10

I (pA

/pF

)

ICaL

-0.03

-0.02

-0.01

0 2 4 6 8 10

ICaT

-60

-40

-20

0

0 2 4 6 8 10

mV

Action potential

-0.5 0

Istim

Figure 3.6: Behaviour of all Ca2+ -related currents during current-clamp. A 2 s current pulse of 0 to -0.5 pA/pF was applied while activitiesof all the Ca2+ -related currents were recorded. The morphology of INSCa andISOCCa closely matched the shape of the AP which shows these currents arestrongly voltage-dependent. The peaks of ICaL greatly correlated to the peaksof the AP and displayed an inverted profile of the AP while only the first peakof ICaT matched the initial peak of the AP. INaCa and ICapump accumulated andreached their peak before decaying to their equilibrium levels. The decay rateof ICapump was faster than INaCa . The accumulation occurred when INaCa andICapump did not have time to recover and as a consequence Ca2+ did not havetime to leave the cell. During the AP, it clearly showed that ICaL is the majorcontributor to ICa−tot and after the cell repolarised, a small tail outward currentwas observed. This tail current is closely linked to ICapump as Ca2+ was beingrestored.

117

0.03

0.05

0.07

0.09

0.11

0 2 4 6 8 10

I (pA

/pF

)

t (s)

INaK

-1.2

-0.8

-0.4

0

0 2 4 6 8 10

t (s)

INa-tot

-7.5e-5

-6.5e-5

-5.5e-5

-4.5e-5

-3.5e-5

0 2 4 6 8 10

I (pA

/pF

)

ISOCNa

-0.25

-0.15

-0.05

0.05

0 2 4 6 8 10

INSNa

-1.19e-3

-1.17e-3

-1.15e-3

-1.13e-3

-1.11e-3

0 2 4 6 8 10

INaKCl-Na

-0.8

-0.6

-0.4

-0.2

0

0 2 4 6 8 10

I (pA

/pF

)

INa

-3.0e-3

-1.5e-3

0

1.5e-3

0 2 4 6 8 10

Ih-Na

-60

-40

-20

0

0 2 4 6 8 10

mV

Action potential

-0.5 0

Istim

Figure 3.7: Behaviour of all Na+ -related currents during current-clamp. Na+ channels are activated when cells depolarise. A rapid influx ofINa was observed when a stimulus pulse was first applied in simulation results.Ih−Na was not active when the stimulus pulse was applied since Ih is only activatedat RMP. ISOCNa , INSNa and INaK are strongly voltage-dependent as the profileof these currents resembled the shape and characteristic of the AP. ISOCNa andINaKCl−Na did not seem to contribute greatly to INa−tot as the magnitude of thesecurrents were relatively low. It did not show a strong correlation with the chang-ing membrane potential. The major contributing currents to INa−tot are INa ,INSCa and INaK . INa−tot returned to its equilibrium value after reaching its peakhas a direct link between the balance of both INaK and INSNa .

118

with the largest magnitudes, the morphology of IK−tot also closely resembled the

shape of the AP.

3.3.4 Other currents

The activities of ICl , ISOC , Ih and INSCC were examined and illustrated in figure

3.9. ICl , ISOC , Ih were all voltage-correlated but the profile of ICl was inverted.

As described in the previous section, Ih was only activated at RMP and was not

active during the current-clamp as expected.

3.4 Investigation of AP and total membrane cur-

rent while inhibiting individual currents

To investigate the AP and whole cell membrane current further, each individ-

ual membrane current was completely blocked in turn under both voltage- and

current-clamp. The results were then compared qualitatively to the simulated

control data under the same clamping condition. This gives a clear indication of

how the ionic current gives rise to certain phenomena observed in the myometrial

cell.

3.4.1 Inhibiting the inward currents

During a 2 s current clamp, elimination of the inward membrane currents, which is

shown in figure 3.10, indicated that the RMP were not affected. Blocking ICaL and

INa have a similar effect on AP, which include significantly reduced Vmax and gen-

eration of a plateau type AP with no spikes. Blocking ICaT reduced the number of

spikes but did not change Vmax , while blocking Ih has no visible effect. Blockage

of the inward currents has an insignificant effect on the total membrane current

apart from ICaL . The maximum inward peak displayed in control has completely

disappeared and only the maximum outward peak was presented.

The results show both ICaL and INa play an important role in cell excitability,

although ICaL is a major contributor to the total inward current. Studies sug-

gested that Na+ is responsible for repetitive firing of spikes with fast upstroke

generation and the propagation of the AP [8, 27, 63, 72, 85]. Inhibition of this

channel not only stopped the spike generation, it also rendered the propagation

119

-6e-4

-4e-4

-2e-4

0

2e-4

0 2 4 6 8 10

I (pA

/pF

)

t (s)

INSK

-0.006

-0.004

-0.002

0

0.002

0 2 4 6 8 10

t (s)

Ih-K

0

1

2

3

4

0 2 4 6 8 10

t (s)

IK-tot

0

0.02

0.04

0 2 4 6 8 10

I (pA

/pF

)

IKCa

0

0.1

0.2

0.3

0.4

0 2 4 6 8 10

IKA

0.1

0.2

0.3

0.4

0.5

0 2 4 6 8 10

IKleak

0

0.4

0.8

0 2 4 6 8 10

I (pA

/pF

)

IK1

0

1

2

3

0 2 4 6 8 10

IK2

-60

-40

-20

0

0 2 4 6 8 10

mV

Action potential

-0.5 0

Istim

Figure 3.8: Behaviour of all K+ -related currents during current-clamp.IK1 , IK2 , IKA , IKleak and INSK were strongly linked to the membrane potential asshown in simulated results. Since Ih−K only activated at RMP, the activity ofIh−K showed this channel is inactive during the current-clamp. The shape ofIK−tot also closely resembled the shape of the AP because IK1 and IK2 accountedfor the majority of IK−tot .

120

-0.01

-0.006

-0.002

0.002

0 2 4 6 8 10

I (pA

/pF

)

t (s)

Ih

-0.3

-0.2

-0.1

0

0.1

0 2 4 6 8 10

t (s)

INSCC

-0.16

-0.08

0

0 2 4 6 8 10

I (pA

/pF

)ICl

-8e-05

-6e-05

-4e-05

0 2 4 6 8 10

ISOC

-60

-40

-20

0

0 2 4 6 8 10

V (

mV

)

Action potential

-0.5 0

Istim

Figure 3.9: Behaviour of other currents during current-clamp. ISOC andIh were both strongly voltage-correlated as the currents showed the same featuresand characteristics as the AP. ICl was also voltage-dependent with an invertedprofile compared to the AP. As expected, Ih was only activated at RMP and wasinactive during the current-clamp.

121

of AP, the cell became less excitable and therefore reduced Vmax . ICaL is vital

in the excitability of a uterus cell and it is responsible for triggering muscle con-

tractions [8, 27, 72]. Blocking ICaL also suppressed production of the spikes and

reduced the cell’s excitability.

3.4.2 Inhibiting the outward currents

Inhibiting IK1 raised the RMP before the stimulus pulse was applied by about 5

mV and also raised Vmax by about 3 mV (figure 3.11). A plateau in the AP was

displayed with only its initial spike seen. After the stimulus pulse was released,

the potential of the cell returned to its equilibrium, which was higher than its

initial value when the stimulus pulse was applied. The AP with IK2 blockage was

very unstable at the start before the stimulus pulse was applied, the potential

dramatically raised and then fell to its equilibrium value of RMP to about -

20 mV. Only a plateau AP was displayed, and Vmax only went up to -15.6 mV

before returning to its RMP when the stimulus pulse was lifted. A 10 s pre-

pulse were also applied to both IK1 and IK2 blockage studies to ensure the model

has reached its equilibrium values and they both displayed the same results (not

shown). Blocking IKA increases the bursting frequency. The AP bursted with

decreasing amplitude followed by a long plateau before returning to its RMP. The

cell potential was not returned to its RMP after the stimulus pulse was released.

There was no visible change in the AP while blocking IKCa . Current traces show

no significant effect was found on the membrane current while blocking both

IKA and IKCa . However, blocking IK1 and IK2 changed the magnitude of Itot .

The maximum outward peak was reduced by about 25% when blocking IK1 and

more than 50% when blocking IK2 .

This study shows the uterus cell became very unstable when IK1 and IK2 were

inhibited, it also shows that K+ currents, especially IK1 and IK2 has an effect on

spike production and setting the RMP of the uterus cell. This finding is consistent

with the experiment described in Khan and Knock et al. [2,29,30] since the role of

K+ current is to dampen the excitibility of the cell and reduce the cell membrane

potential to its RMP. Inhibiting K+ current reduced repolarisation in the cell

since damping was reduced, this in turn raised RMP, as seen in the results.

122

-60

-40

-20

0

0 5 10

V (

mM

)

t(s)

ICaL

block

-60

-40

-20

0

0 5 10

t(s)

ICaT

block

-60

-40

-20

0

0 5 10

t(s)

INa

block

-60

-40

-20

0

0 5 10

t(s)

Ih

block

-8

0

8

0 200 400 600

I tot (

pA/p

F)

t (ms)

-8

0

8

0 200 400 600

t (ms)

-8

0

8

0 200 400 600

t (ms)

-8

0

8

0 200 400 600

t (ms)

Figure 3.10: Effects of inhibiting the inward currents. ICaL , ICaT , INa andIh (left to right) were blocked individually to investigate the overall effect on thewhole cell AP (top) and the membrane current (bottom). While blocking certaincurrent as indicated on the figure, a 2 s current stimulus pulse from 0 to -0.5pA/pF was applied to examine the AP and a 300 ms voltage pulse from -50to 20 mV was applied to examine the membrane current. The results (red)were then compared to AP produced in control condition (black). It is clearlyshown that blocking inward currents did not alter the RMP and blocking ICaL andINa changed the shape of the AP and Vmax . Inhibiting ICaL eliminated the inwardpeak on the total membrane current while inhibiting other inward currents didnot show any significant effect.

123

-60

-40

-20

0

0 5 10

V (

mM

)

t(s)

IK1

block

-60

-40

-20

0

0 5 10

t(s)

IK2

block

-60

-40

-20

0

0 5 10

t(s)

IKA

block

-60

-40

-20

0

0 5 10

t(s)

IKCa

block

-8

0

8

0 200 400 600

I tot (

pA/p

F)

t (ms)

-8

0

8

0 200 400 600

t (ms)

-8

0

8

0 200 400 600

t (ms)

-8

0

8

0 200 400 600

t (ms)

Figure 3.11: Effects of inhibiting the outward currents. Inhibition ofIK1 , IK2 , IKA and IKCa (left to right) were investigated to see the effect on theAP (top) and the membrane current (bottom) with the results (red) comparedto recordings obtained in control condition (black). The cell became unstablewhen IK1 and IK2 were blocked, only plateau type APs were shown and RMP hadrisen. Blocking IKA resulted in the AP starting bursting and gradually increasedthe spiking frequency. Insignificant change was seen when IKCa was blocked. Asignificant reduction in Itot was observed while inhibiting IK1 and IK2 but therewere no visible effect when IKA and IKCa were inhibited.

124

3.4.3 Inhibiting the other currents

Results illustrated in figure 3.12 showed that a complete blockage of ICl still

produced bursting type APs with reduced spiking frequency despite the second

spike being suppressed. The RMP remained the same, but the magnitude at the

foot of the AP was reduced. Inhibiting INSCC drastically reduced the RMP to

around -75 mV, while Vmax stayed unchanged. The shape of the AP was also

changed, with only the first spike seen. The cell depolarised and repolarised and

the cell potential was slowly building up with time, creating a slow-rising curve

rather than a flat plateau. The AP with INaK block displayed a slight increase

with the spiking frequency and a slight rise in the magnitude at the foot of the

AP. Blocking INSCC slightly reduced the RMP and Vmax . The cell was heavily

damped and spikes gradually died down to form a plateau around -40 mV. In

terms of total membrane current, only blockage of ICl has a little effect on the

total current (increase slightly after the stimulus pulse was applied), inhibiting

other currents, INSCC , INaK and INaCa has no visible change.

Experimental studies conducted by Jones et al. [32] showed that when ICl

channel blocker drugs was applied to the myometrial cell, the frequency of spon-

taneous contractions significantly reduced. Simulation results showed that only

a slight reduction of spike frequency was observed. Miyoshi et al. [33] suggested

that INSCC has an important function in reconstructing the pace-making activity

and regulating the contractility in myometrium. It is clearly shown in simula-

tions, that inhibiting this current completely changed the shape and contractility

of the cell.

3.5 Different types of AP

Depending on the gestation and physiological conditions, different shapes of AP

are also seen experimentally in mammalian myometrial cells. These different

shapes of AP can be classified into six groups: 1) Bursting type where each spike

is fully repolarised and returned to its RMP, 2) Plateau type with orderly bursting

spikes, 3) Disorderly bursting-plateau type, 4) Disorderly plateau-bursting type,

5) Plateau type and 6) Long ADP plateau type.

The model was used to imitate these different shapes of AP by changing

selected parameters in the model (e.g. Increasing or decreasing the size of the

certain currents). The results are shown in figure 3.13 and the precise change of

125

-60

-40

-20

0

0 5 10

V (

mM

)

t(s)

ICl

block

-80

-60

-40

-20

0

0 5 10

t(s)

INSCC

block

-60

-40

-20

0

0 5 10

t(s)

INaK

block

-60

-40

-20

0

0 5 10

t(s)

INaCa

block

-8

0

8

0 200 400 600

I tot (

pA/p

F)

t (ms)

-8

0

8

0 200 400 600

t (ms)

-8

0

8

0 200 400 600

t (ms)

-8

0

8

0 200 400 600

t (ms)

Figure 3.12: Effects of inhibiting other current. ICl , INSCC , INaK andINaCa (left to right) were blocked individually to examine their effect on the AP(top) and the membrane current (bottom) with the results (red) compared torecordings obtained in control condition (black). Inhibiting ICl slowed downthe spiking frequency while RMP remained unchanged. Blocking INSCC andINaCa completely changed the shape of AP and RMP was drastically decreasedthrough blocking INSCC alone. INaK has little effect on AP while inhibited. Allthe membrane currents showed negligible change when currents were blocked.Only inhibition of ICl showed a slight rise after the voltage-pulse was returnedback to its original potential.

126

the parameters are listed in table 3.2. The cell was under current-clamp (from

0 to 0.5 pA/pF) for 2 s, and the results were validated using experimental data

obtained from Inoue et al. [108] and Okabe et al. [26] under the effects of estradiol

and control conditions. This shows the model’s capability to adapt to reproduce

a variety of AP seen in other smooth muscle cells. This also shows that the model

can potentially be used to reproduce results from different gestations of the cells

or even similar type of smooth muscle cells (e.g. circular myometrial cell).

Figure 3.13 Type of AP Changes in parameterA Bursting (spikes return to RMP) gNa =0.42, gK1 =0.526,

gK−leak =0.006B Bursting (spikes not return to RMP) gNa =0.204, gK1 =0.282,

gK−leak =0.006C Mixture of bursting and plateau gNa =0.18, gK1 =0.182

D Plateau gK1 =0.205, gK2 =0.088,gK−leak =0.006

E Irregular gK1 =0.683, gK2 =0.008,gK−leak =0.006

F Irregular gNa =0.18, gK1 =0.341,gK−leak =0.006

Table 3.2: Parameters changed in producing various types of AP seenin smooth muscle cells.

127

-60

-40

-20

0

0 2 4 6

V (

mV

)

A

-60

-40

-20

0

0 2 4 6

B

-60

-40

-20

0

0 2 4 6

C

-60

-40

-20

0

0 2 4 6

V (

mV

)

t (s)

D

-60

-40

-20

0

0 2 4 6

t (s)

E

-60

-40

-20

0

0 2 4 6

t (s)

F

Figure 3.13: Other possible types of APs produced by the model.Various types of APs produced under current clamp with changes of parameterlisted in table 3.2. (A) Bursting AP with each spike returned to its RMP beforethe next spike is fired. (B) Bursting AP without spike repolarised to RMP. (C) Amixture of bursting-plateau type AP, with AP starting bursting with small spikesand gradually becoming a plateau. (D) A plateau type AP with a plateau levelaround -40 mV. (E-F) APs with spikes of irregular frequencies and amplitudes.

128

Chapter 4

Results II – Mechanical

production

The myometrial model was tested to investigate the excitation-contraction cou-

pling and the mechanical behaviour of a single uterine smooth muscle cell. Pre-

dictions made by the model were compared to the experimental data from my-

ometrium when available. Different testing voltage or current protocols applied

to the model followed the information provided in literature as closely as possible.

However, the magnitude of force provided by the model was considerably smaller

compared to experimental recordings as expected. This is due to incorporation of

the cerebrovascular mechanical model by Yang et al., with the majority of the pa-

rameters provided by the mechanical model left unmodified. As explained in the

previous chapter, most of the parameters were not available for the myometrium,

and using parameters from another type of smooth muscle gave a good indication

of how a uterine cell produced mechanical force. Even though the magnitude of

the force was significantly less, this did not compromise on the outcome of the

results and the major characteristics qualitatively, and similarities to the exper-

iments were still observed. All the simulations were carried out in an isometric

condition (constant cell length) with lc = 150µm unless otherwise stated.

4.1 Fractional stress production in myometrium

Fractional stress produced by a single myometrial cell was investigated using a

modified Hai and Murphy, four-state cross-bridge cycling model [56, 57] (figure

4.1). Two different protocols (a single pulse or 10 repetitive pulses) were applied

129

to the myometrial model. The peak of stress produced by the model with a single

pulse stimulation was 58.63% and it took 4.98 s to reached this peak. Repetitive

voltage pulses generated a maximal stress of 77.50% and the time taken to reached

the peak was 5.67 s. The rise of the stress level was due to [ Ca2+ ]i accumulation

when the model was repetitively stimulated. The model also showed stress pro-

duced by the repetitive pulses was more prolonged and generated a more efficient

production of stress than a single pulse firing as described in experiments [73].

It took about the same time (around 45 to 50 s) for both cases to relax back to

their equilibrium stress level.

In both cases, the distribution of the four states of the cross-bridges were also

investigated. The levels of the states increased when the external voltage pulses

were applied in all the states apart from the free myosin state (M). The profile

of the M state is inversely related to the fractional stress because less myosin

was available when stress was generated in the cell. The level of state M slowly

recovered back to its equilibrium level when the original stress of the cell was

restored. The sudden increased level of the phosphorylated cross-bridge state

(Mp) when voltage stimuli was applied caused the state of the latch bridge (AM)

to decline before reaching its peak.

4.2 Single-, twin- and multiple-pulse simulation

in myometrium

A twin-pulse stimulation was applied to the myometrium and the results from

simulations were compared to experimental data. Burdyga et al. [161] examined

the simultaneous activity of an isolated 21-day pregnant myometrial cell in rat.

The cells were placed under a current-clamp and spontaneous activity of Ca2+ -

spikes was recorded (figure 4.2(A)). It was found [ Ca2+ ]i rose with fast upstroke

and decayed exponentially and slowly. It took 155.8± 4.5 to reach its peak and

the duration at 50% was 998±60 ms. The actual stimulation used in experiments

were unknown. In simulations, the first stimulus was applied from 0 to -3 pA/pF

for 800 ms and the same stimulus was applied again 1000 ms later. The fast

upstroke and slow decay characteristics were also observed. The time taken to

reach the peak of [ Ca2+ ]i was 828.2 ms and the duration at 50% was 2.40 s. Both

of these values were greater compared to the recordings obtained from Burdyga

et al. [161].

130

-80 0

V (

mV

)A

-80 0

V (

mV

)B

100

150

200

[Ca2+

] i

200

400

600

800

[Ca2+

] i

20

40

60

80

Str

ess

(%)

20

40

60

80

Str

ess

(%)

0

0.2

0.4

0.6

0.8

1

0 10 20 30 40 50

Fra

ctio

n

t (s)

MAM

AMpMp

0

0.2

0.4

0.6

0.8

1

0 10 20 30 40 50

Fra

ctio

n

t (s)

Figure 4.1: Stress development in a single myometrial cell. Two dif-ferent protocols were applied to the myometrial model to investigate [ Ca2+ ]i ,fractional stress and temporal distribution of the cross-bridge cycling states. Asingle pulse from -80 to 0 mV for 200 ms stimulation in (A) showed the fractionstress produced by the cell reached about 60 % and the 10 repetitive pulses from-80 to 100 ms at 3 Hz stimulation (B) reached a higher level of about 80%. Thisis due to the increased content of [ Ca2+ ]i available producing more tension inthe cell. The time taken for both cases to decay back to their equilibrium levelwas about 45 to 50 s.

131

In figure 4.2(B), the associated AP in the experiment was evoked by a depo-

larising voltage step and the AP was shown in an expanded time scale illustrated

in figure 4.2(C). The resting membrane potential reported from this experiment

was −57.5± 3.2 mV with an overshoot of 4.82± 1.52 mV. In simulation, low fre-

quency oscillations followed by a plateau type of AP was observed. The resting

membrane potential recorded, and the overshoot of the AP were -67.01 mV and

5.40 mV respectively.

Single- and multiple-pulse stimulations were also applied to the model to

investigate the Ca2+ -spikes and total force, Ft generated by the model. Results

illustrated in figure 4.3 showed data from simulation compared to experimental

recordings from Burdyga et al. [161]. In a single-pulse stimulation, a brief current

pulse from 0 to -5 pA/pF with a pulse duration of 20 ms was applied and the

corresponding AP is shown in figure 4.3(A,left). The associated Ca2+ -spike and

total force produced by the model are illustrated in figure 4.3(B and C, left).

The time taken for the Ca2+ -spike go reach the peak was 60 ms and it took

more than 15 s to decay back to the equilibrium value. Experimental data shown

inset reported different Ca2+ -spike profiles from two different regions of the same

myometrial strip. The total force produced by the model generated a peak force

of 2.7 µN in 2.74 s and the time taken for the model to decay back to its steady-

state value was more than 20s, which was longer compared to the experimental

recordings.

In the multiple-pulse stimulation, the same pulse was applied four times with

6 s intervals in between the successive stimulations. It is shown in figure 4.3 (B

and C, right) that the level of the Ca2+ -spikes accumulated and resulted in a

larger but incomplete force production. Each spike produced by the current pulse

corresponded to the contraction, and the magnitude of this contraction depended

on the number of spikes and the burst frequency in the [ Ca2+ ]i transient. This

was confirmed by the experimental data shown inset.

4.3 Other mechanical responses in a single uter-

ine cell

Active force, Fa and the individual length components in the myometrial cell

were investigated and represented in figure 4.4. The model was voltage-clamped

from a holding potential of -60 to 0 mV for 1 s, with the length of the cell, lc

132

1

2.6

F/F

0

A

Experiment Simulation

5 s

-70

20

V (

mV

)

B

5 s

-73

20

V (

mV

)

C

0.5 s

Figure 4.2: Twin-pulse simulation in myometrium compared to exper-imental recordings. In simulations, two current pulses from 0 to -3 pA/pFwere applied, 1000 ms apart (the actual stimulation used in experiments wereunknown). (A) Ca2+ -spikes have fast upstrokes with slow decays similar to ex-periment recordings from Burdyga et al. [161], however the time to peak andduration at 50% were both longer compare to the experimental data. (B) Thecorresponding AP in simulations has a resting membrane potential of -67.01 mVwith an overshoot of 5.40 mV. (C) The AP in an expanded time scale shows theAP oscillated in low frequency before settling into a plateau. Both simulationsand experimental recordings were shown in the same time scale.

133

-80

-40

0

40

V (

mV

)

A Action potential

1

1.1

1.2

1.3

F/F

0

B Ca2+

transient

2

2.5

3

3.5

Ft (

µN)

C Force

5 s 10 s

1

1.4

5 s

0

1.4

mN

5 s

1

1.45

5 s

0

2.5

mN

5 s

Figure 4.3: Ca2+ -spikes and force production in a single myometrialcell with single- and multiple-pulse stimulations. (Left) A single currentstimulus from 0 to -5 pA/pF was applied and the corresponding Ca2+ spikes (B)and total force (C) was recorded. The calcium spike displayed a fast upstroke witha slow decay similar to the experimental data shown inset. The two recordingswere obtained from two different regions of the same myometrial strip. The totalforce produced peaked at 2.7 µN in 2.74 s and the time taken to relax back toits steady-state value was more than 20s. (Right) The same stimulus was appliedfour times with a 6-s interval before the next pulse. The force generated bya multiple-stimulation was greater than a single-stimulation, although the forceproduced was incomplete. The incomplete force accumulated to generate greaterforce similar to the data reported in the experiment. Each spike corresponded toeach contraction and the amplitude of the contraction depended on the numberof spikes and burst frequency in the [ Ca2+ ]i transient.

134

= 123 µm. The time taken to reach the peak [ Ca2+ ]i transient, and the peak

active force generated, were 6.57 and 7.71 s respectively which showed a clear

delay in muscle contractions. During the contraction, the length of the active

component, la decreased or shortened and consequently the lengths of the series

and cross-bridge components, ls and lx increased, compensating the augmented

level of force.

The myometrial model also examined the [ Ca2+ ]i transient, total force, Ft

and the peak force rate, dF/dt with increasing value of membrane voltages. In

an isometric condition, with lc = 123 µm, the model was clamped with a single

voltage holding from -40 to 60 mV every 5 mV for 1 s. The simulation illustrated

in figure 4.5 reported that the peak of [ Ca2+ ]i corresponded directly to the total

force and the peak force rate. The peaks of [ Ca2+ ]i , total force and force rate

reached their maximum values at -20 mV.

135

140

160

180

200

220

240

260

0 10 20 30 40 50 60

[Ca2+

] i (n

M)

A

1

1.5

2

2.5

0 10 20 30 40 50 60

Fa

(µN

)

t (s)

B

43

45

47

0 10 20 30 40 50 60

l s (

µm)

C

75.5

76.5

77.5 78.5

79.5

0 10 20 30 40 50 60

l a (

µm)

D

0.3

0.4

0.5

0 10 20 30 40 50 60

l x (

µm)

E

-60

0

mV

Figure 4.4: [ Ca2+ ]i transient, active force, Fa and the individual lengthcomponents in the myometrial cell. A 1-s voltage stimulus pulse from -60to 0 mV was applied (A, inset) and the corresponding [ Ca2+ ]i transient (A)and active force (B) showed a delay in muscle contraction. The length of theactive component, la was shortened during contraction and the lengths of theseries viscoelastic component, ls and cross-bridge component, lx were thereforeincreased to compensate the rise of the force (C-E).

136

150

350

550

-40 -30 -20 -10 0 10 20 30 40 50 60

peak

[Ca2+

] i(n

M)

A

1.4

1.8

2.2

2.6

-40 -30 -20 -10 0 10 20 30 40 50 60

peak

Ft

(µN

)

B

0

0.5

1

1.5

2

-40 -30 -20 -10 0 10 20 30 40 50 60

peak

rat

e(µ

N/s

)

V (mV)

C

Figure 4.5: Comparison of [ Ca2+ ]i transient, total force and peak forcerate. The myometrial model was put under voltage-clamp from -40 to 60 mVevery 5 mV for 1 s. The maximal peak of the [ Ca2+ ]i transient directly corre-sponded to the maximal peaks of total force and peak force rate at -20 mV.

137

Chapter 5

Drug actions – Investigation of

the properties of [ Ca2+ ]i transient

and contractile force

To understand the functionality of a cell, and give an insight into certain roles

of a specific current or transporter in a sub-cellular level, drugs were applied

experimentally to inhibit the current and see how the cell behaved without it.

In simulations, drug actions seen in experiments can be mimicked by inhibiting

certain currents to see if the same effects could be replicated. In particular, how

[ Ca2+ ]i transient and contractile force are operated in a single myometrial cell

were focused on. Since [ Ca2+ ]i is directly linked to the generation of arrhythmic

contractility in the uterus towards the end of the pregnancy, regulation of this

contractile force is absolutely vital. Simulations of drug actions focussing on these

two categories will not only help in the understanding of mechanisms underlying

[ Ca2+ ]i transient and contractility in myometrium, it also puts the model to the

test, and provides useful information for future model improvement.

5.1 Properties of [ Ca2+ ]i transient in myometrial

cells

To investigate the properties of the [ Ca2+ ]i transient in myometrium, voltage-

dependence of the Ca2+ transient were looked at by applying depolarising pulses

with various magnitudes to the model and compared with the experimental data.

138

Carboxeyosin was used to inhibit the sarcolemmal Ca2+ pump to study the con-

tribution of ICapump as a calcium extraction mechanism, and examine how the

decay rate changed when the drug was used. Nifedipine, an L-type Ca2+ blocker

was used to investigate the contribution of Ca2+ and ICaL to the inward current.

Finally, the role of CICR and its contribution to the [ Ca2+ ]i transient was also

testing with a CICR inhibitor, ryanodine.

5.1.1 Depolarising pulse with varying magnitude

The effects of varying depolarising pulses on [ Ca2+ ]i transient and ICa−tot in

late pregnant rat myometrium were investigated by Shmigol et al. [69]. Cells

were held at a holding potential of -50 mV briefly (10 ms) before stepping from

-40 to 20 mV every 10 mV, then 20 to 40 mV every 20 mV for 200 ms. Each cell

was allowed to relax to its steady state and was not held at a particular voltage

before the next voltage-clamp was applied (figure 5.1 (A, top)). If this separation

of the pulses was not introduced, Ca2+ might not have finished decaying and

therefore have an affect on the amplitude of the subsequent peak. The same

investigation was repeated in simulations. In experiment, it was reported that in

cells under normal physiological condition, the magnitudes of both [ Ca2+ ]i and

ICa−tot increased as the magnitudes of the depolarising pulses increased. After

both [ Ca2+ ]i and ICa−tot reached their peaks around -10 to 0 mV, the amplitudes

of [ Ca2+ ]i and ICa−tot began to decline. In simulations, the strongest peaks with

similar amplitudes of [ Ca2+ ]i were found when depolarising pulses were -20 and

-10 mV, while the maximum peak of ICa−tot agreed well with the experimental

data. Both ∆ [ Ca2+ ]i and ICa−tot showed a bell-shape dependency on the voltage

in experiments and simulations as illustrated in 5.1(C). It also showed results in

experiments with 6 cells revealed that [ Ca2+ ]i actually peaked when depolarising

pulses were -10 and 0 mV.

5.1.2 Carboxyeosin, CE

Carboxyeosin (CE) is a sarcolemmal Ca2+ pump inhibitor used extensively in

cardiac smooth muscle to study the rate at which, and how the Ca2+ extrusion

process occurs [162–164]. It was reported that carboxyeosin raised the resting

[ Ca2+ ]i level and the magnitude of the [ Ca2+ ]i transient in rat ventricular my-

ocyctes [162]. It also showed that the rate of its relaxation was increased by

139

-50

60

mV

A

-40-30-20 -10 0 10 20 40 60

100

200

300

400

[Ca2+

] i (n

M)

-8

-4

0

I Ca-

tot (

pA/p

F)

5 s

-50

60

B

-40-30-20-10 0 10 20 40 60

250

300

350

400

-8

-4

0

4 5 s

0

100

200

300

∆[C

a2+] i

(nM

)

C

SimExp

-12

-8

-4

0

4

-60-40-20 0 20 40 60pe

ak I C

a-to

t (pA

/pF

)V (mV)

Figure 5.1: The effect of depolarisation with different magnitudes on[ Ca2+ ]i transient and ICa−tot . Voltage clamps with a holding potential of -50mV before stepping from -40 to 20 mV every 10 mV, then 20 to 40 mV every20 mV for 200 ms in experiment (A, top) and simulations (B, top). In experi-ments by Shmigol et al. [69], each cell was allowed to relax to its steady statebefore the next voltage-clamp was applied. The same condition was achieved insimulations by separating each depolarising pulse so the model was not held ata particular potential. Both experiment and simulations showed the amplitudesof [ Ca2+ ]i (middle) and ICa−tot (bottom) increased as the magnitudes of the de-polarising pulses increased before reaching a certain voltage and the amplitudesgradually decline. A plot of ∆ [ Ca2+ ]i and ICa−tot against membrane potentialdisplayed a bell-shape dependency on the voltage in experimental and simulationresults.

140

5%. Experiments conducted by Shmigol et al. [123] in isolated uterine cells from

pregnant rats showed that cells treated with carboxyeosin displayed a significant

decrease in the rate of [ Ca2+ ]i transient decay. The rate of decay dropped from

0.66±0.24 s−1 in control to 0.435 ± 0.670.015 s−1 in cells pre-treated with CE.

It was also reported the resting level of [ Ca2+ ]i increased from 109 ± 25 nM

to 206 ± 11 nM when treated with CE. [ Ca2+ ]i was still able to restore in car-

boxyeosin treated cells and sarcolemmal calcium pump accounted for 30% of the

Ca2+ extrusion process from cell.

The same experiment was repeated in simulation under the same physiological

condition as illustrated in figure 5.2. A train of 10 depolarising pulses from -80 to

0 mV at 3 Hz was applied to the model in control and with ICapump blockage. The

resting [ Ca2+ ]i elevated to 180.48 nM in the model with ICapump blockage compare

to 167.27 nM in the control case (12.61% increase). The peak of [ Ca2+ ]i was also

raised in ICapump blocked results with peak value of 662.56 nM compare to 585.94

nM in control (13.08% increase). Blocking ICapump showed the decay rate of the

[ Ca2+ ]i has slightly increased compared to the control case. The decay rates were

0.672 s−1 and 0.669 s−1 in control and ICapump condition respectively. This shows

an inconsistency to the experimental recordings. However, as shown in figure 5.2

(C), although the model in both conditions decayed at similar rates, both decays

merged to a same point at around 15 s after reaching their peak values which

resembles the experimental data.

5.1.3 Nifedipine, NI

Nifedipine is an L-type calcium channel blocker which is widely used in differ-

ent forms to treat patients with pre-term labour, tetanus, cardiac diseases and

even cancer-related muscle spasms. Nifedipine relaxes and widens blood ves-

sels, veins and arteries to lower hypertension in heart diseases. In the uterus,

nifedipine used as a form of tocolyticis is used to treat pre-term labour by slow-

ing down the uterine contractions. Nifedpine is a preferable choice of drug as

studies showed that nifedipine gives fewer side-effects and implications to new-

borns [165]. It is reported in human pregnant myometrium, application of this

drug vastly reduced or completely eliminated the level of [ Ca2+ ]i and sponta-

neous contractions [25, 28, 69, 166]. Simulations were carried out by blocking

ICaL to see whether the same results can be re-produced. As shown in figure

5.3, a voltage-clamp of 10 s from -80 to 0 mV was applied to both control and

141

0

250

500

750

1000

[Ca2+

] i (n

M)

ExperimentA

5 s

Control CE

150

350

550

[Ca2+

] i (n

M)

SimulationB

5 s

Control CE

0

0.2

0.4

0.6

0.8

1

5 10 15

Nor

mal

ised

[Ca2+

] i

t (s)

C

CE (exp)Control (exp)

CE (sim)Control (sim)

Figure 5.2: The effect of carboxyeosin on [ Ca2+ ]i transient. A train of 10depolarising pulses from -80 to 0 mV at 3 Hz was applied experimentally (A) andto the model (B) in control (labelled with Control) and with ICapump blockage(labelled with CE). Results obtained in experiments obtained by Shmigol etal. [123] recorded an increase in both resting and peak [ Ca2+ ]i levels, this wasshown in simulation results with a 12.61% and 13.08% increase in resting andpeak [ Ca2+ ]i respectively. In (C), the decays of both control (black) and car-boxyeosin effect (red) in simulation (lines) were compared to experimental data(points). The traces were normalized to the peak [ Ca2+ ]i after subtracting theirequilibrium level of [ Ca2+ ]i . In simulations, the decay rate of the model withICapump block has increased which was inconsistent with the experimental data.However, the decay rates in both conditions merged at around 15 s and reachedzero after their peaks, similar to experiment recordings.

142

ICaL blocked model and levels of [ Ca2+ ]i and ICa−tot were recorded. The results

were then compared to the experimental recordings obtained by Wray et al. [13].

It is clearly seen that the amplitude of [ Ca2+ ]i and ICa−tot were greatly dimin-

ished but not completely abolished when ICaL was inhibited. This confirms that

Ca2+ entry is mostly through L-type calcium channels and the current responsible

for the remaining of the [ Ca2+ ]i entry is ICaT .

5.1.4 Ryanodine, Ry

Ryanodine (Ry) is an alkaloid derived from a plant called Ryania. There are no

known medications created from ryanodine to date. It is produced synthetically

and used commonly in experiments to examine how calcium channels are operated

and how the influx/efflux of Ca2+ are controlled in cardiac, skeleton and uterine

smooth muscle cells [69,167–169]. Ryanodine inhibits CICR and blocks the phasic

release of Ca2+ . In myometrium, it is observed that ryanodine-sensitive stores

are responsible for the high frequency calcium oscillation which lead muscles to

contract massively in pregnant cultured [170] and non-cultured human uterine

cell [136, 171]. It is also reported that ryanodine enhanced contractility in tissue

by 60% from pregnant rat myometrium at term but not non-pregnant [135]. This

may be explained by the increased number of calcium channels and changing

sensitivity in ryanodine receptors (RyR) towards the end of the pregnancy.

To investigate the role of CICR in contributing to the [ Ca2+ ]i transient,

Shmigol et al. [69] conducted a study on single isolated pregnant rat myome-

trial cells using a low dose (5 µM) of ryanodine to inhibit the CICR process. A

twin voltage-pulse from a holding potential of -50 to 0 mV was applied with a

500 ms interval between the two pulses, to both control and ryanodine treated

cells. It was reported in control cells, the second increment in [ Ca2+ ]i (elicited

by the second pulse) was the same or larger than the first increment (elicited by

the first pulse). The peak of ICa−tot however showed the opposite, displaying a

negative staircase effect. In simulations, the experiment was replicated and illus-

trated in figure 5.4. It showed a small but significant rise in [ Ca2+ ]i elicited by

the second voltage-pulse (6.07% increase was recorded). The peaks of ICa−tot on

the other hand decreased from -8.64 pA/pF evoked by the first pulse to -7.94

pA/pF by the second pulse with 8.1% decreased recorded. When ryanodine was

added to the cell experimentally, the second [ Ca2+ ]i increment was smaller than

the first one, while the amplitude of ICa−tot was also reduced. The same results

143

-80

0

mV

A

50

250

450

[Ca2+

] i (n

M)

B Control Nifedipine

3 s

ExpSim

-10

-5

0

I Ca-

tot (

pA/p

F)

C

-4

0

pA/p

F

-4

0

pA/p

F

Figure 5.3: The effect of nifedipine on [ Ca2+ ]i transient. A voltage-clamp from a holding potential of -80 to 0 mV for 10 s was applied to bothcontrol and ICaL cases (A). The activities of [ Ca2+ ]i (B) and ICa−tot (C), in thesimulated results (dotted line) were compared to the experimental recordings(dotted line) [13]. With the presence of NI, the amplitudes of [ Ca2+ ]i were sig-nificantly reduced in both experimental and simulated recordings, and no riseof [ Ca2+ ]i was detected, indicating the majority of the Ca2+ entry occurred viaL-type calcium channels. It is shown inset that nifedipine prevented the inwardcurrent in experiment while a small amount of ICaT inward current could still beseen in simulations.

144

were also seen in simulations with Irel inhibited, a 5.49% and 3.4% decrease in

the second [ Ca2+ ]i increment and peak ICa−tot respectively was recorded. It was

also found that the decays of [ Ca2+ ]i elicited by both pulses were slower in ryan-

odine condition. The activities of the calcium uptake and release concentrations

[ Ca2+ ]u , [ Ca2+ ]r , uptake, transfer and release currents Iup , Itr and Irel were

recorded in both ryanodine and control condition during the twin-pulse (figure

5.4(C) and (D)). It has revealed that [ Ca2+ ]u showed a gradual decline in control

and stayed constant when CICR was inhibited. Itr and Irel were also decreasing

while Iup stayed at zero with the presence of CICR block.

5.2 Properties of contractile force in myome-

trial cells

The properties of contractile force in uterine cells were studied using carbachol,

cyclopiazonic acid, nifedipine, wortmannin and 0-Ca solution. Cells treated with

carbachol release Ca2+ from the SR store while cyclopiazonic acid inhibits Ca2+ -

ATPase. In simulations, both drugs were used to examine the SR contribution

to contractile force. L-type calcium and MLCK process were also examined with

nifedipine and wortmannin to highlight the importance of these mechanisms in

force generation. Finally 0-Ca solution was used to demonstrate the activities of

[ Ca2+ ]i and contraction with an absence of any external Ca2+ .

It is important to note that all the results obtained from simulations were

carried out in a shorter time scale than the experimental data. The duration of

the experiments on contractile force were always done in a great length of time

(around 10-40 mins). This exceeds the stability of the model and caused the model

to behave unexpectedly. The duration of the simulations was therefore shortened

but carefully chosen so the major effects, features and morphology could still be

seen and compared to experiments. Also, the uterine cells used in experiments

were all freshly isolated, and contractions were occurring spontaneously without

any external stimulus due to the pacemaking activity in myometrium. This effect

was not seen in simulations and stimulus must be applied to induce force pro-

duction. This stimulus was also cautiously designed depending on the drug used

and the effect investigated so the contractile force in simulation matched with the

force production seen in experiments. The magnitude of the force produced by

the model was smaller than the recordings seen in experiments due to the reasons

145

100

200

300

400

[Ca2+

] i (n

M)

A

1 s

Exp Control

1 s

Exp RyR

200

250

300

350

400

[Ca2+

] i (n

M)

B

1 s

Sim Control

1 s

SimRyR

-6

-4

-2

0

2

I Ca-

tot (

pA/p

F)

-8

-6

-4

-2

0

2

I Ca-

tot (

pA/p

F)

0.07

0.09

0.11

0.13

0 1 2 3 4 5

[Ca2+

] i (n

M)

t (s)

C

[Ca]u

[Ca]r

0

0.04

0.08

0.12

0.16

0 1 2 3 4 5

I (pA

/pF

)

t (s)

D

Irel

IupItr

ControlRyR

-50

0

mV

-50

0

mV

Figure 5.4: The effect of ryanodine on [ Ca2+ ]i and ICa−tot in my-ometrium. A twin voltage-pulse from a holding potential of -50 to 0 mV wasapplied with a 500 ms interval between the two pulses to both control and ryan-odine treated cells in experiment by Shmigol et al. [69] (A) and simulations (B). Incontrol, both experiment recordings and simulations showed that the second incre-ment in [ Ca2+ ]i was the same, or larger than the first increment (6.07% increasewas recorded in simulations) while the second peak in ICa−tot was smaller thanthe first (8.1% decreased recorded in simulations) displaying a negative staircaseeffect. When ryanodine was introduced to the cell, the second [ Ca2+ ]i incrementwas seen smaller than the first one, while the amplitude of ICa−tot was also re-duced. A 5.49% and 3.4% decrease in the second [ Ca2+ ]i increment and peakICa−tot respectively was recorded in simulations. The behaviour of the calciumuptake and release concentrations [ Ca2+ ]u , [ Ca2+ ]r (C), uptake, transfer andrelease currents Iup , Itr and Irel (D) were recorded in both ryanodine (dottedline) and control (solid line) condition. [ Ca2+ ]u declines gradually in controland stayed at a constant level when CICR was inhibited. Itr and Irel were alsodecreasing gradually while Iup stayed at zero with the presence of CICR block.

146

descried in the previous chapter. Again, this did not affect the model’s ability

to produce the mechanical characteristics seen in a single uterine cell and results

from the model was compared qualitatively to the experimental data. In terms of

the [ Ca2+ ]i transient, only the general shapes and features were compared qual-

itatively to the experimental data. This is because [ Ca2+ ]i is always measured

in the units of fura-2, indo-1 and fluo-3 ratios in experiments, using a fluorescent

indicator excited by UV or visible light. The absolute value of [ Ca2+ ]i cannot

be calibrated and determined without extra experimental procedures [172] as the

function of most indicators modified the intracellular environment.

5.2.1 Carbachol, CCh

Carbachol (CCh) is a drug that mimics the action of acetylcholine. It stimulates

the muscarinic receptors, and enhances contractions by releasing Ca2+ from the

SR store in many organs. It is commonly used as a medication to treat different

ophthalmic problems, and also to increase the strength of muscle contractions in

bladders. In research, carbachol is used extensively in various types of smooth

muscles from different organs including intestine [173], uterus [161,174–177] and

cervix [178]. This drug is not well absorbed in the gastro-intestinal tract and

although the use of carbachol for dilatation of the cervix have been demonstrated

in cows [178], there is no known medication existed for the human pregnant

uterus.

Experiments demonstrated by Burdyga et al. [161] showed that 21-day preg-

nant rat myometrial cells treated with carbachol have a 30-40% increase in max-

imal force. When carbachol was removed, the frequency and amplitude of the

Ca2+ spikes decreased until it reached equilibrium. The effect of carbachol was

replicated in simulations by increasing the probability of Ca2+ release from the

SR store to one. A train of stimulus current pulse was applied to the model (from

0 to -5 pA/pF) at t = 12 s to t = 58 s, the effect of carbachol was introduced at

t = 12 s and removed at t = 50 s. The cell was held at 0 pA/pF for a further

55 s before the end of the simulation. It is observed that carbachol increased

the magnitude of the force as shown in figure 5.5, and the amplitude dropped

when the effect of carbachol was withdrawn, as expected. The amplitude was

decreased by 2.4% in Ca2+ spikes and 0.5% in contractile force when carbachol

was removed.

147

0

1

2

3

4

5

For

ce (

mN

)

A

CCh

Experiment

1.4

1.8

2.2

2.6

3

For

ce (

µN)

CCh

Simulation

1

1.2

1.4

1.6

F/F

0

B

20 s

CCh

0.8

1.2

1.6

2

2.4

Nor

mal

ised

[Ca2+

] i

20 s

CCh

0.1µN

10 s

0.3

10 s

Figure 5.5: The effects of carbachol on contractile force in my-ometrium. (A) Carbachol increased the maximal force by 30-40% comparedto control cells in experiments obtained by Burdyga et al. (the recordings of cellsin control condition was not published) [161]. The experiment was repeated insimulations to show the effect of introducing and removing carbachol on force inthe model (right) compared to control (left). A train of stimulus current pulsefrom 0 to -5 pA/pF was applied to the model at t = 12 s to t = 58 s. The effect ofcarbachol was introduced at t = 12 s and removed at t = 50 s. Upon the removalof carbachol, a decrease of 0.5% in amplitude was observed. (B) The frequencyof Ca2+ spikes was reduced when carbachol was removed in experiments and re-sults from simulations show similar effects. A decrease of 0.5% in amplitude inCa2+ spikes was observed. The [ Ca2+ ]i recorded was normalised to the resting[ Ca2+ ]i level (160 nM). The effects of carbachol on both force and [ Ca2+ ]i areenlarged and shown in the insets.

148

5.2.2 Cyclopiazonic acid, CPA

Cyclopiazonic acid (CPA) is a highly potent neurotoxic. It is fatal if the drug

is ingested, inhaled or enters into contact with the skin. It inhibits the Ca2+ -

ATPase in the SR, and blocks the uptake current, Iup . CPA is used commonly to

investigate the contractile activities of pregnant uterine smooth muscles in both

rat [45, 135, 179] and human [4, 13, 136]. It is found in both species that when

CPA is introduced to the cells, a passive leakage from the SR store is observed

as Ca2+ is unbalanced. The internal Ca2+ can no longer be stored in the SR

since the influx of Ca2+ to the SR is inhibited. As reported by Kupittayanant et

al. [136] and Wray et al. [4], this has a small but significant effect on [ Ca2+ ]i ,

raising the amplitude and amplitude duration by 3% and 11% respectively. The

amplitude and duration of force were each elevated by 4%. Although the basal

level of [ Ca2+ ]i was increased by 3% with CPA treatment, it was not observed

in contraction (figure 5.6 (A and B)). Only slight increments were observed when

Ca2+ -ATPase was inhibited, which shows SR only plays a small part in myome-

trial contraction. It is not known why the basal level of contraction was not

increased with augmented basal [ Ca2+ ]i level. It has been suggested that ca-

pacitative or store-operated mechanisms exist in non-excitable cells, which are

responsible [180,181].

Simulated results described in figure 5.6 (C and D) shows the effect of blocking

Iup on contraction force and [ Ca2+ ]i respectively. The morphologies and features

from simulations were compared to experimental recordings shown in figure 5.6 (A

and B). The basal level of [ Ca2+ ]i was raised 6.33% when Iup blockage was first

applied at t = 150 s and then the level of [ Ca2+ ]i gradually declined. The ampli-

tude of [ Ca2+ ]i has elevated 3.7% but no change in amplitude was observed when

Iup was inhibited. In contractile force, the basal level has increased significantly

and declined slowly when Iup was applied, which was not seen in experiments.

The amplitude of force was raised 0.47% and the duration of the amplitude has

remained the same. Figure 5.6 (right panel) shows the superimposed contractile

force and [ Ca2+ ]i of experimental recordings and simulations in both control and

Iup inhibited condition. The recordings are expanded, and only a single peak was

displayed which clearly showed the effect of cyclopiazonic acid raising the level

of the amplitude and basal levels when applied.

149

0

10

20

30

For

ce (

mN

)

A CPA

0.4

0.6

0.8

1

1.2

Indo

-1R

atio

B

5 mins

CPA

CPA

1 min

2

2.4

2.8

3.2

For

ce (

µN)

C CPA

155

175

195

215

[Ca2+

] i (n

M)

D

1 min

CPA

10 s

CPA

Figure 5.6: The activity of contraction and [ Ca2+ ]i with presenceof CPA. The effect of cyclopiazonic acid (CPA) on both contraction and[ Ca2+ ]i were examined. Experimental recordings (A and B) showed raised[ Ca2+ ]i basal level but not in contractile force when CPA was applied. Theamplitude and its duration were both increased with the presence of CPA. Insimulations, the model was stimulated by a train of current-pulses from 0 to -5pA/pF with pulse duration of 20 ms and basic cycle length of 30 s. Iup was in-hibited at t = 150 s until the end of the simulation at 300 s. The basal level andamplitude of both [ Ca2+ ]i transient and contractile force were increased, whereasthe amplitude durations were unchanged when Iup was blocked. Superimposedforce and cai (right panel) in control (black), and in the presence of cyclopiazonicacid (red), demonstrated the effect of inhibiting Iup when CPA was applied.

150

5.2.3 Nifedipine, NI

As discussed in the previous section, nifedipine (NI) is an L-type Ca2+ channel

blocker. When nifedipine was applied to myometrial cells, no rise in [ Ca2+ ]i was

observed and the peak of ICa−tot was abolished [13]. Contractile force was also

abolished with cells pre-treated with nifedipine [45, 161]. Results in human

myometrium at term obtained from Parkington et al. [25] showed that both

[ Ca2+ ]i and tension development were abolished even with the presence of prostaglandins

E2 (PGE, a naturally occurring drug which causes contraction and labour in my-

ometrium [182, 183]). Simulations were carried out to investigate the effect of

inhibiting ICaL on contractile force (figure 5.7). Both contractile force and nor-

malised force were greatly diminished (by 72.24% and 99.36% respectively) com-

pare to control cases, although a small rise of force was still seen, while the peaks

in experimental recordings were completely abolished. This small rise is thought

to be the ICaT contribution to the contractile force in the model. The results in

simulations confirm Ca2+ entry through L-type Ca2+ channel plays an important

role in generating contractile force.

5.2.4 Wortmannin, W

Wortmannin is a toxin which inhibits MLCK. It is standalone or combined with

therapy to treat various type cancerous cells as wortmannin inhibits DNA repair-

ing and cell proliferating. However there is no known medication which exists for

treatments in pregnant myometrium. Experimental recordings showed that wort-

mannin gradually abolished spontaneous contractions in oxytocin-induced and

non-induced rat myometrial cells [13, 184]. It was reported that while current

tracings of ICa−tot revealed that ICa−tot showed no signs of differentiation when

cells were treated with wortmannin, contractile force in wortmannin treated cells

was decreased by 12 ± 1% in rat and 37 ± 8% in human [184]. Results simulated

by the model with a shorter simulation time, illustrated in figure 5.8, showed

that contractile force was completely abolished almost instantaneously and no

further contractions were seen when the Ca2+ -calmodulin-MLCK process was

inhibited. However, the level of [ Ca2+ ]i was not affected. There were no signs

of change observed either in ICa−tot indicating MLCK is an essential process in

force generation, consistent with the experiment data.

151

0

1

2

For

ce (

mN

)

A Experiment

Nifedipine

1.4

1.8

2.2

2.6

3

For

ce (

µN)

Nifedipine

Simulation

1

2

3

4

F/F

0

B

1 min20 mins

Nifedipine

0.8

1.2

1.6

2

[Ca2+

] i

20 s

Nifedipine

Figure 5.7: The effects of nifedipine on [ Ca2+ ]i and contractile forcein myometrium. Effects of nefedipine on contractile force (A) and normalisedforce (B) in experiments by Burdyga et al. [161] and simulations. After experi-mental tracings were recorded in control condition, a 20 mins break was appliedto the cell before nifedipine was added to the cell to ensure equilibrium wasachieved. In simulations, the model was simulated separately for both controland ICaL inhibition. A train of stimulus current-pulse from 0 to -5 pA/pF withpulse duration of 20 ms was applied for 38 seconds before letting the model set-tled back to its steady-state. Both contraction force and Ca2+ showed a drasticdecrease in activity when ICaL was inhibited, similar to experimental recordings.

152

0

0.4

0.8

1.2

1.6

For

ce (

mN

)

A

Wortmannin

Experiment

0

1

2

3

Ft (

µN)

Wortmannin

Simulation

0.18

0.22

0.26

0.3

Indo

-1

400

/500

B

10 mins 155

170

185

200

[Ca2+

] i (n

M)

1 min

-6

-4

-2

0

2

4

40 60 80 100 120 140

I Ca-

tot (

pA/p

F)

t (ms)

C

ControlWortmannin

-12

-8

-4

0

40 60 80 100 120 140

I Ca-

tot (

pA/p

F)

t (ms)

ControlWortmannin

Figure 5.8: The effects of wortmannin on [ Ca2+ ]i and ICa−tot in my-ometrium. An MLCK inhibitor, wortmannin was applied to human myometrialcell in experiments conducted by Longbottom et al. [184] to see the effect it has onspontaneous contraction force (A), [ Ca2+ ]i (B) and ICa−tot (C). In simulations,a current-clamp from 0 to -5 pA/pF (with pulses of duration of 2 ms, and basiccycle length of 15 s) was continuously stimulated for 5 mins. The process ofMLCK was inhibited at t = 1.5 mins until the end of the simulation. The resultsproduced by the model agreed with the experimental recordings that the activ-ity of contractile force had markedly reduced when wortmannin was introduced,while the amplitudes of the [ Ca2+ ]i and ICa−tot on the other hand remained un-changed. Although the slow gradual abolishment of contractility in simulationswas not see, the model showed that the MLCK process is the key mechanism ingenerating force production in the myometrium.

153

5.2.5 Calcium-free solution

The effects of 0-Ca solution on contractile force and intracellular calcium in

myometrium have been demonstrated extensively in experiments with or with-

out other application of drugs [4, 13, 45, 135, 136, 179]. In simulations, this was

achieved by changing the level of extracellular calcium, [ Ca2+ ]o to a very small

value (1 × 10−10 nM) instead of zero to avoid mathematical errors in equations.

For example when calculating the reversal potentials of ionic currents, setting

[ Ca2+ ]o equals to zero is mathematically impossible. Simulated results described

in figure 5.9, show that the total contractile force declined to zero when 0-Ca

condition was applied and raised back to its original level when the 0-Ca con-

dition was lifted. In experiments, contractile force was instantaneously abol-

ished when 0-Ca solution was applied. The level of [ Ca2+ ]i slowly decreased in

both experiments and simulations as the balanced Ca2+ of the cell was restored

naturally. This confirms the importance of external Ca2+ in contractions. No

[ Ca2+ ]i transient and therefore no force was produced as there was no Ca2+ influx

from extracellular space via voltage-dependent calcium channels.

154

0

2

4

6

8

10

For

ce (

mN

)

A

0-Ca

Experiment

0

1

2

3

For

ce (

µN)

0-Ca

Simulation

1.2

1.4

1.6

1.8

2

Cal

cium

(

Indo

-1)

B

10 mins 40

80

120

160

200[C

a2+] i

(nM

)

1 min

Figure 5.9: The effects of zero external calcium on contractile forceand [ Ca2+ ]i in myometrium. The cell behaviour of force and [ Ca2+ ]i with anabsence of external Ca2+ was investigated experimentally (A) and in simulations(B). A very small value of [ Ca2+ ]o was used (1× 10−10 nM) to imitate the effectof 0-Ca solution in simulations. A train of stimulus current pulses with pulseduration of 20 ms, and basic cycle length of 22 s, was applied to the model from 0to -5 pA/pF. External Ca2+ was withdrawn at t = 136 s and introduced again at t= 200 s, until the simulation terminated at the 4.5 mins (268 s). The contractionforce has abolished when the external Ca2+ was withdrawn in both experimentand simulations. [ Ca2+ ]i expressed in Indo-1 ratio in experiment showed a declinelevel of activity while 0-Ca solution was applied, which is consistent with resultsobtained in simulations.

155

Chapter 6

Sensitivity analysis

During pregnancy, the length, weight, depth, volume and the capacity of the

uterus change to accommodate the growth of the foetus. This growth also in-

volves cell hypotrophy and hyperplasia. Throughout the pregnancy, remodelling

of some ionic channels occurs to increase the contractility in the uterus as pre-

viously discussed in chapter 1. It is found that the number of L-type calcium

channels and sodium channels increase during mid- and late-pregnancy respec-

tively [27], while the number of calcium-activated potassium channels, IKCa loses

sensitivity with labour [17]. The number of non-specific cation channels also seem

to have decreased towards the end of the pregnancy. Levels of progesterone de-

clined markedly and levels of estradiol start increasing towards the late pregnancy

and markedly rise on the day of parturition [185, 186]. The number of oxytocin

receptors also increase [17, 187] towards parturition. The resting membrane po-

tential of the cell increased from -52 to -46 mV during delivery [188] and there

are also a slight different in the ionic content of the cell due to the changes in sex

hormone levels during pregnancy [189].

In a tissue level, the number of gap junctions increases [190], while the to-

tal tissue and the gap junction resistances decreases towards delivery. Histologic

analysis of myometrial tissue strips showed an elevated level of the cellular frac-

tion, but areas of the nuclei decreases in pregnant myometrium [191]. This im-

proves the transmission of the electrical signal through out the whole organ. The

physiological balance of the uterus is particularly vital towards the late pregnancy

to ensure a synchronised contraction during delivery.

Since the uterus undergoes enormous physiological and structural changes

before, during, and even after pregnancies, a sensitivity analysis was carried out

156

to test the model’s ability to adapt to all these changes. Therefore, the model

can eventually be used to simulate cells not just in late pregnancy but also in

cells from non-pregnant to different stages of pregnancy. The parameters tested

using the myometrial model were: cell area to volume ratio (Ac /Vc ), length

of the myometrial cell ( lc ), current density of Ca2+ ( ICa−tot ); Na+ ( INa−tot );

K+ ( IK−tot ) and finally MLCK. The original values of these parameters are given

in the appendix.

Each of the parameters were altered within physiological conditions, but in

some cases they were not. A voltage pulse of from -80 to 0 mV was applied for

200 ms to investigate the characteristics and behaviour of the [ Ca2+ ]i transient

and total force, Ft , to see how the changing of the parameters affected the model.

Furthermore, changes in time to peak force and [ Ca2+ ]i , as well as changes of

force and [ Ca2+ ]i after the first 3 s were also examined. The values obtained from

the model in control condition are presented in table 6.1. Since the model could

become unstable when parameters were changed during the sensitivity analysis,

the model was clamped to a holding potential for a long period of time to settle

numerically until equilibrium or steady-state was reached before applying the

voltage stimulus pulse.

Peak[Ca2+]i(nM)

Time topeak

[Ca2+]i (s)

[Ca2+]i in3 s (nM)

Peaktotalforce(µN)

Time topeaktotal

force (s)

Total forcein 3 s (µN)

214.823 0.216 178.425 1.413 3.53 1.397

Table 6.1: Values of peak [ Ca2+ ]i and total force, time to reach thepeak of [ Ca2+ ]i and total force and [ Ca2+ ]i and force in 3 s producedby the model in control condition. Control condition implies the parametersused to produced the set of results were not modified from the parameters givenfrom the appendix.

6.1 Cell area to volume ratio

Cell area and cell volume both vary enormously during and after pregnancy. As

previously discussed in chapter 1, the capacity of the human uterus can increase

500-times during pregnancy [6]. This cell hypertrophy is caused by stretching

of the myometrial tissue while accommodating a growing foetus. Shynlova et al.

157

conducted a stereological analysis on cell volume changes throughout different

gestations in rats [192]. From non-pregnant to early days of pregnancy (day 8),

it was reported that the size and the volume of a uterine cell showed very little

differences and the cell volumes were 264±112µm3 and 245±90µm3 respectively.

The cell volume began to increase towards the middle of the pregnancy and on

day 14, the cell volume was 448 ± 225µm3. The volume of the cell continued

to grow in late pregnancy and reached 771 ± 402µm3 and 716 ± 364µm3 in day

19 and day 22. However, there was no change in size from the last state of the

pregnancy till term to stabilise the uterus and the foetus. Four days after the

delivery, it was found that the myometrial cell volume was 296± 101µm3. It was

reported by Yoshino et al. that the surface area of a rat myometrial cell increased

from 928.4±235.1 and 5193±188.4 to 7599.4±639.7 µm2 in non-pregnant, mid-

pregnant and late pregnant cells.

In a modelling environment, a sensitivity analysis was carried out to provide

a prediction on physiological responses with various values cell area to volume

ratio. This gives an insight into whether the model potentially be adapted to

simulate cells from different gestation of the pregnancy or even post-parturition.

The model’s response to the changing value of Ac /Vc is shown in figure 6.1.

It is clearly seen that when Ac /Vc was increased 150%, the peak and the resting

level of [ Ca2+ ]i transient increased significantly (about 125%) compared to values

from control condition. It took a longer time for [ Ca2+ ]i to reach its peak with de-

creasing Ac / Vc , although the results were not apparent. The [ Ca2+ ]i transient

also decayed at a slower rate when Ac /Vc decreased.

The time taken for the contraction to reach its peak was noticeably faster

(about 40%) and stronger force was generated (about 90%) when Ac /Vc was aug-

mented by 50% its original values. However, when the magnitude of Ac /Vc was

dropped to 75% of its original value, the strength of the contraction fell dras-

tically and only a slight rise in force was observed when the voltage pulse was

applied. The actual changes recorded are presented in table 6.2.

6.2 Length of the myometrial cell

The changes of the length of a myometrial cell results from hypertrophy and

stretch during pregnancy. Mollard et al. found that the length of a freshly

dissociated late-pregnant longitudinal myometrial cell in rat was 80 to 300 µm,

158

50

150

250

350

450

5 10 15 20

[Ca2+

] iA

1.5*(Ac/Vc)Ac/Vc

0.25*(Ac/Vc)

0

1

2

3

20 40 60

Ft (

µN)

t (s)

B

-100

-50

0

50

100

150

% c

hang

ein

pea

k

25%

150%

C

CalciumForce

-40

-20

0

20

40

60

% c

hang

e in

time

to p

eak

25%

150%

-100

-50

0

50

100

% c

hang

e of

[Ca2+

] i or

Ft i

n 3

s

25%

150%

Figure 6.1: The effects of changing values of cell area to volume ra-tio, Ac / Vc on the myometrial model. The [ Ca2+ ]i transient (A) and to-tal force (B) were investigated with different values of Ac /Vc . The dottedline indicated the peak values of [ Ca2+ ]i and Ft generated by the control case(black). The peak and the resting level of [ Ca2+ ]i significantly increased whenAc /Vc increased. However, the time taken to reach the peak of [ Ca2+ ]i also de-creased with an elevated level of Ac /Vc . In force production, the time taken toreach its peak was 40.76% faster and 90.2% stronger when Ac /Vc was increasedby 50% of its original value. However, when the magnitude of Ac /Vc was dropped75%, the contraction produced fell 90.8% compared to values obtained from con-trol condition. Only a slight increased in force was seen when the stimulus wasapplied. The actual numerical changes recorded in the sensitivity analysis forAc /Vc are listed in table 6.2.

159

Parameter Changes inparameter

value

Changes inpeak [Ca2+]i

Changes intime to peak

Changes in[Ca2+]i in 3 s

Ac /Vc-75% -48.09% +18.69% -39.19%

+150% +124.54% -32.04% +23.73%

Parameter Changes inparameter

value

Changes inpeak force

Changes intime to peak

Changes inFt in 3 s

Ac /Vc-75% -90.8% +67.29% -91.69%

+150% +90.2% -40.76% +87.87%

Table 6.2: Numerical changes of [ Ca2+ ]i transient and total force withchanging cell area to volume ratio in the sensitivity analysis.

with a diameter of 6 to 12 µm [156]. Ohya et al. reported the length of a late

pregnant cell in rat was 200-700 µm, with a diameter of 8-30 µm [72]. Inoue et

al. measured the longitudinal length of cell from different gestation of pregnant

rats (5, 9, 14, 18 and 21 days) immediately after cells isolation, it was reported

the length of cells was 200 to 700 µm, with a diameter of 3 to 30 µm [27].

The changing of the cell length caused by stretch has been long known to have

various effects on myometrium during pregnancy [2, 28, 193–195]. It is known to

stimulate hyperplasia and hypertrophy in early pregnancy [196] and initiate con-

tractile activity through stretch-induced currents at term. This effect, although

significant, has not been described in the myometrial model (for more details see

future plan in chapter 10).

The sensitivity analysis was performed to see how the model reacted to chang-

ing lengths of the cell with a brief 200 ms voltage stimulus. The results il-

lustrated in figure 6.2 showed the model’s response to force generation (since

[ Ca2+ ]i transient is not length dependent). The change of the length of the cell

alone and the strength of the contractions are not directly proportional. This

implied the longer cell length did not generate greater force. The largest length

of the cell tested was 200 µm in the sensitivity analysis, but this length was still

short compared to experimentally recorded values (200-700 µm). Preliminary re-

sults (not illustrated) showed that longer cell lengths produced less contractions,

even with a longer and stronger voltage stimulus. The cell length which generated

the maximum force was 150 µm. The cell (especially the individual force com-

ponent) might also undergo deformation if the cell was being stretched too much

160

and therefore not generating the force properly. This is due to the fact that the

mechanical model adapted was originally constructed for vascular smooth muscle

cells and not myometrium. Parameters were mostly left unchanged as data for

myometrium were not available. This affected the model’s ability to correctly

predict the force generation in the correct magnitude.

Parameter Changes inparameter

value

Changes inpeak force

Changes intime to peak

Changes inFt in 3 s

lc

-18.7% -56.4% -6.71% +56.3%+50% +38.1% +12.4% +38.16%

+62.6% -92.2% -7.08% -92.2%

Table 6.3: Numerical changes of total force with changing lengths of amyometrial cell.

6.3 Current density of ICa−tot

Ca2+ currents undergo remodelling before and during pregnancy. In non-pregnant

adult rat myometrium, progesterone can increase the Ca2+ current density in

cells [197], while in pregnant rat myometrium, withdrawal of progesterone causes

the mRNA levels of voltage-gated Ca2+ channel subunits to increase before term

and preterm labour [198]. It was demonstrated by Tezuka et al. that the in-

crease of the mRNA levels indicated the changes in the expression to provide

the necessary excitability ready for contraction. Similar results were also found

in pregnant guinea pig cells [199]. Ohkubo et al. reported the expression of L-

type channels changed markedly during pregnancy, but the T-subtype channel

(α1G) only increased near term. This change was different between longitudinal

and circular cells. Although the number of the L-type Ca2+ channels increase in

pregnancy, it was shown by Mershon et al. that this channel did not change prior

to, or with the onset of labour and the mRNA expression declined drastically at

parturition, when the voltage-dated Ca2+ channels were at their peak [200].

Figure 6.3 shows the effects of changing the current density of the total of

Ca2+ current ( ICa−tot = ICaL + ICaT + INSCa + INaCa + ISOCCa + ICapump ). The

peaks of [ Ca2+ ]i transient and total force were proportional to the changes in

the ICa−tot , i.e. when the total current density of Ca2+ increased, the peaks

of [ Ca2+ ]i and Ft increased and vice versa. The peak of [ Ca2+ ]i has increased

161

0

0.5

1

1.5

2

20 40 60

Ft (

µN)

t (s)

A

lc = 200lc = 150lc = 123lc = 100

-40

0

40

80

% c

hang

ein

pea

k

81.3%

50% 62.6%

B

Force

-8

-4

0

4

8

12

% c

hang

e in

time

to p

eak

81.3%50% 62.6%

-100

-75

-50

-25

0

25

% c

hang

e of

[Ca2+

] i or

Ft i

n 3

s

81.3%

50%

62.6%

Figure 6.2: The effects of changing values of cell length on the my-ometrial model. (A) The total force generated by the model and the length ofthe cell were not directly proportional (the length of the cell lc was measured inµm). The length that produced the maximum total force was 150 µm and thetime taken for all the contractions to reach their peaks by different lengths wereapproximately the same (3.3 to 4 s). It took a longer time for the contractionto relax back to its steady-state when the magnitude of the total force was high.(B) The percentage changes of the peak total force, time to peak and force in3 s compared to the control condition were presented in the bar charts. Thenumerical results are shown in table 6.3.

162

335% and the peak of Ft has increased 54% of its original value when ICa−tot was

raised four times. However, the basal levels of both [Ca2+]i transient and Ft did

not respond significantly to various values of ICa−tot . It took a longer time for

the peaks of [ Ca2+ ]i transient and Ft to decay back to their steady-state values

as ICa−tot declined. In [ Ca2+ ]i transient simulations, it took approximately the

same time (0.20 to 0.22 s) to reach the peak, whereas the time taken for Ft to

reach the peak reduced as ICa−tot increased. This showed the elevated values of

ICa−tot prompted a faster activation in the force production without altering the

activation kinetics in the [ Ca2+ ]i transient.

Parameter Changes inparameter

value

Changes inpeak [Ca2+]i

Changes intime to peak

Changes in[Ca2+]i in 3 s

ICa−tot

-50% -19.08% +1.89% -7.31%+200% +60.23% -2.16% +2.15%+400% +334.65% -6.89% -14.15%

Parameter Changes inparameter

value

Changes inpeak force

Changes intime to peak

Changes inFt in 3 s

ICa−tot

-50% -26.17% +36.38% -28.94%+200% +34.57% -32.97% +33.20%+400% +53.73% -48.12% +41.43%

Table 6.4: Numerical changes of both [ Ca2+ ]i transient and total forcewith changing values of IK−tot .

6.4 Current density of INa−tot

Similar to the Ca2+ channel current, the Na+ channel current and the Na+ -

Ca2+ activity also increases near term [109, 122]. In pregnant rat longitudinal

cells, although the I-V relationship of the Na+ current does not change during

gestation, the average current density of the Na+ current drastically increases

towards labour [27, 85]. Inoue and Sperelakis measured the current density of

the Na+ current throughout the gestation in rat longitudinal single cells, it was

reported that the current density increased from 0 and 0.19± 0.16 pA/pF in day

5 and day 9 respectively, to 0.86 ± 0.14 in day 21 of the pregnancy [27]. This

increase was almost linear due to an increase in the fraction of cells that contain

163

100

300

500

700

5 10 15 20

[Ca2+

] iA 4*ICa-tot

2*ICa-totICa-tot

0.5*ICa-tot

0.6

1

1.4

1.8

2.2

20 40 60

Ft (

µN)

t (s)

B

0

100

200

300

% c

hang

ein

pea

k

50%

200% 400%

C CalciumForce

-40

-20

0

20

40

% c

hang

e in

time

to p

eak

50%

200% 400%

-20

0

20

40

% c

hang

e of

[Ca2+

] i or

Ft i

n 3

s

50%200%

400%

Figure 6.3: The effects of changing the current density of ICa−tot in[ Ca2+ ]i transient and the total force. The current density of ICa−tot washalved (blue), doubled (green) and quadrupled (red) and the changes of the peak[ Ca2+ ]i and Ft , time taken to reach the peaks and [ Ca2+ ]i and Ft in 3 s wereinvestigated and compared with the results obtained in simulated control con-dition. The dotted lines in the figures indicated the time when the peaks wasreached in [ Ca2+ ]i transient (A) and total force, Ft (B) by the model in controlcondition (black) when a voltage stimulus pulse from 0 to -80 mV was appliedfor 200 ms. The peaks of [ Ca2+ ]i and Ft were proportional to the changes ofICa−tot but the basal level of both [ Ca2+ ]i and Ft were not significantly affectedby this change. The time taken for contractions to reach maximum was reducedwith increasing values of ICa−tot , but this did not significantly influence [ Ca2+ ]i .(C) The model’s response to the various values of ICa−tot compared to the controlcase and the actual numerical are presented in table 6.4.

164

Na+ channels. Since Na+ currents are responsible for cell excitability, this shows

the Na+ channel activity becomes more important near term in preparation for

labour.

The effects on changing the current density of INa−tot ( INa−tot = INa + INaCa +

INaK + ISOCNa + INSK + INaKCl−Na + Ih−Na ) are shown in figure 6.4 and the

actual changes in percentage compared to the control case are listed in table

6.6. When the total Na+ current, INa−tot was increased more than three times,

the model became very unstable, even though the model was held over 2 mins

at a holding potential of -80 mV before applying the voltage stimulus pulse.

Both [ Ca2+ ]i transient and total force decayed at a constant rate and equilibrium

was not reached, simulated results were therefore not included in the sensitivity

analysis. Varying INa−tot did not affect the time to peak in [ Ca2+ ]i , the maximum

contraction and the relaxation significantly. However, the kinetics of the decay

changed noticeably in the [ Ca2+ ]i transient when various INa−tot values was used.

[ Ca2+ ]i decayed at the slowest rate when INa−tot was halved and faster when

INa−tot was doubled compare to the control values.

Parameter Changes inparameter

value

Changes inpeak [Ca2+]i

Changes intime to peak

Changes in[Ca2+]i in 3 s

INa−tot-50% +1.89% -4.53% +6.64%

+200% -0.79% -4.53% -5.5%

Parameter Changes inparameter

value

Changes inpeak force

Changes intime to peak

Changes inFt in 3 s

INa−tot-50% -3.38% +2.38% -3.25%

+200% -1.81% +6.55% -1.46%

Table 6.5: Numerical changes of both [ Ca2+ ]i transient and total forcewith changing values of INa−tot .

6.5 Current density of IK−tot

The decrease in K+ current near term increases the excitability and contractions

in preparation for labour. This decrease is due to a decline in K+ channel ex-

pression and a reduction in the channel conductance [201]. The reduction in the

K+ channel conductance is the result of an up-regulation in β-subunit (KCNE2)

165

100

120

140

160

180

200

220

5 10 15 20

[Ca2+

] i

A

2*INa-totINa-tot

0.5*INa-tot

0.4

0.6

0.8

1

1.2

1.4

20 40 60

Ft (

µN)

t (s)

B

-4

-2

0

2

4

% c

hang

ein

pea

k

50%

200%

C

CalciumForce

-6-4-2 0 2 4 6 8

% c

hang

e in

time

to p

eak

50%

200%

-8

-4

0

4

8

% c

hang

e of

[Ca2+

] i or

Ft i

n 3

s 50%

200%

Figure 6.4: The effects of changing the current density of INa−tot in[ Ca2+ ]i transient and the total force. (A) The maximum [ Ca2+ ]i and timeto peak in [ Ca2+ ]i only varied within approximately 4% or less when differentvalues of INa−tot were used. The [ Ca2+ ]i transient decayed at the slowest ratewhen INa−tot was halved, but when INa−tot was doubled, [ Ca2+ ]i decayed fastercompare results in the control condition (black) and the basal values deceased asINa−tot increased. (B) Varying values of INa−tot did not have a significant effectson the total force. (C) The response to the various values of INa−tot compared tothe control case and the actual numerical are presented in table 6.4.

166

as demonstrated by Greenwood et al. [202]. Smith et al. investigated the role

of voltage-gated K+ channels in mouse uterine contractility [203]. It was re-

ported that contractions were not inhibited in pregnant mouse myometrium when

a voltage-gated K+ blocker, 4AP was applied, but contractions were caused in

non-pregnant tissues. A Kv4.2 and Kv4.3 blocker, phrixotoxin-2 was also used

and the same effects was observed. It was found that channel expression Kv4.3

disappeared in term pregnant mouse tissues. Song et al. demonstrated that the

gene expression Kv4.3 found in K+ channel diminished greatly towards the end of

the pregnancy. This is a result of an increasing level of estrogen near term before

parturition. Estrogen also indirectly reduced the plasma membrane of the Kv4.3

protein and controlled the cell trafficking in late-pregnant myometrium [204].

Figure 6.5 depicts the physiological responses of the model with various values

of IK−tot ( IK−tot = IK1 + IK2 + IKA + IKCa + IKleak + INSK + INaK + INaKCl−K +

Ih−K ). The model became very unstable when the value of IK−tot was modified

and a long pre-pulse of over 2 min was applied before stimulating the model

with a 200 ms voltage stimulus. Changing values of IK−tot had little effects on

both the [ Ca2+ ]i transient and total force. The peak and the basal value of

[ Ca2+ ]i decreased but only within 1.53 to 4.38 % when IK−tot was increased 200

and 400% of its original value. Increasing and decreasing IK−tot only diminish

the force generated by the model compared to the control case. There were also

no noticeable differences for [ Ca2+ ]i and Ft to reach their peaks. The actual

numerical changes compare to the control condition are presented in table 6.5.

Parameter Changes inparameter

value

Changes inpeak [Ca2+]i

Changes intime to peak

Changes in[Ca2+]i in 3 s

INa−tot

-50% +17.7% -4.53% +6.64%+200% +1.89% -4.53% -5.50%+400% -0.79% -4.53% -5.70%

Parameter Changes inparameter

value

Changes inpeak force

Changes intime to peak

Changes inFt in 3 s

INa−tot

-50% -16.00% +6.55% +16.46%+200% -3.38% +2.38% -1.46%+400% -3.54% -4.63% -3.25%

Table 6.6: Numerical changes of [ Ca2+ ]i transient and total force withvarious values of INa−tot .

167

110

140

170

200

5 10 15 20

[Ca2+

] i

A

4*IK-tot2*IK-tot

IK-tot0.5*IK-tot

0.5

0.75

1

1.25

1.5

20 40 60

Ft (

µN)

t (s)

B

-3

-2

-1

0

% c

hang

ein

pea

k

50% 200%

400%

C

CalciumForce

-2

-1.6

-1.2

-0.8

-0.4

0

% c

hang

e in

time

to p

eak

50% 20% 400%

-1.6

-1.2

-0.8

-0.4

0

0.4

% c

hang

e of

[Ca2+

] i or

Ft i

n 3

s

50%200% 400%

Figure 6.5: The myometrial model’s physiological response to differentvalues of IK−tot . Changing values of IK−tot did not affect the model significantlyin terms of [ Ca2+ ]i transient (A) and total force (B). The basal level and the peakvalue of [ Ca2+ ]i transient decreased slightly when IK−tot was increased. The totalforce declined when the value of IK−tot was both increased and decreased compareto the original value. Both time to peak in [ Ca2+ ]i and force were unchangedwhen IK−tot was varied. (B) The changes recorded in the sensitivity analysis ofIK−tot are presented in the bar charts.

168

6.6 Myosin light chain kinase

The Ca2+ -calmodulin-MLCK pathway is directly linked to contractions in my-

ometrium [13,205,206]. The increased level of [ Ca2+ ]i binds with calmodulin to

initiate the MLCK process and this Ca2+ sensitivity has been discussed in my-

ometrium [207] and other smooth muscles such as blood vessels [208,209]. Force

can also be modulated by agonists without changing the level of [ Ca2+ ]i , and

MLCK is one of the mechanisms which can modify contractions (while MLCP

is responsible for relaxation in the cells). This has been confirmed experimen-

tally and in simulation with a MLCK inhibitor, Wortmannin, as demonstrated

by Wray and previously shown in simulations [13].

Figure 6.6(A) depicts the model’s reaction to various levels of MLCK. Since

changing the level of MLCK does not affect the [ Ca2+ ]i in the model, only effects

on total force were examined. The contraction generated by the model changed

proportionally with the level of MLCK, i.e. the magnitude of the force together

with its resting level increased as the level of MLCK increased and vice versa. The

profile of the force remained approximately the same, although the contractions

generated with an augmented level of MLCK tend to reach their peak at a faster

rate. The time taken for the contractions to relax to their steady-state also

increased as the level of MLCK increased. Figure 6.6 (B) illustrates the numerical

changes in peak force, time to peak and force in 3 s as the level of MLCk was

altered to half, double and quadruple its value. The results are summarised in

table 6.7.

Parameter Changes inparameter

value

Changes inpeak force

Changes intime to peak

Changes inFt in 3 s

MLCK-50% -36.51% +3.82% -3.13%

+200% +36.82% -10.40% +38.2%+400% +68.60% -23.34% +71.78%

Table 6.7: Numerical changes of total force with changing level ofMLCK in the sensitivity analysis.

169

0

0.5

1

1.5

2

2.5

20 40 60

Ft (

µN)

t (s)

A

4*MLCK2*MLCK

MLCK0.5*MLCK

-40

0

40

80

% c

hang

ein

pea

k

50%

200% 400%

B

Force

-25

-20

-15

-10

-5

0

5

% c

hang

e in

time

to p

eak 50%

200% 400%

-40

0

40

80

% c

hang

e of

[Ca2+

] i or

Ft i

n 3

s

50%200% 400%

Figure 6.6: The effects of various levels of MLCK on the total force. (A)Line in black represents results from control condition and the dotted line repre-sents the peak value produced under control condition. Changing the strength ofMLCK did not change the shape or profile of the total force, although the peaksof the total force slightly left shifted with increasing levels of MLCK, i.e. con-traction reached the peak faster when MLCK increased. It took the contractionsa longer time to recover to their equilibrium values as levels of MLCK increased.(B) Bar charts of the percentage changes in peak total force (top), time taken toreach the peak (middle) and force in 3 s (bottom). The actual numerical changesare listed in table 6.7.

170

Chapter 7

1-D strand model in myometrium

7.1 Introduction

A single myometrial cell model was integrated into a 1-D fibre model to investigate

the conduction velocity and how the action potential propagates along a strand

of a late pregnant rat longitudinal tissue. Action potentials and electrical signals

change their shapes and activities when they travel along a strand of fibre. This

is because neighbouring cells dampen the electrical signals after they have been

initiated. Towards the end of the pregnancies, uterine contractions are found to be

more synchronised with increased amplitudes [4]. It is reported that this increase

in coordinated synchronisation and strength in contraction is directly related to

an increased number of gap junctions towards parturition [210–212]. Sims et

al. observed that the internal and junctional resistances dropped significantly at

delivery in longitudinal rat myometrial tissues, but both resistances increased 1.5

to 2 days after delivery [188]. All these changes occurring towards parturition

improve the electrical coupling and cell-to-cell communications in the uterine

smooth muscle. It also allows an extensive spread of the action potential between

cells and fibres to facilitate co-ordinated contractions in the uterus to ensure a

successful delivery [212–214].

To date, the vast majority of one- and multi-dimensional mathematical mod-

els were developed for cardiac tissues and there are very little models which

describe other types of tissues, including uterine muscles. Benson et al. devel-

oped a 2-D uterine tissue model using a modified FitzHugh-Nagumo excitable

neuron model [215–217]. Weakly and strongly coupled cell-to-cell coupling dur-

ing early and late pregnancies can be modelled by varying mean value of current

171

〈I〉 and diffusion coefficient, D or mean value of diffusion coefficient 〈D〉 stochas-

tically with space. It was observed that excitation activity was localised and

non-synchronised when values of 〈I〉 and D or 〈D〉 were low. As the value of I

increased, frequency and amplitude of excitation increased and when values of D

or 〈D〉 raised, the spatial coherence improved and the temporal and spatial ac-

tivity became more synchronised. Rihana and Marque constructed a 2-D tissue

model by incorporating the single cell ion channel model and reaction diffusion

equations at a tissue level [218, 219]. It was assumed that electrical activities

were uniformly and isotropically propagated, and electrical membrane properties

remained constant throughout. Synchronous and non-synchronous wave propa-

gation were computed by changing axial resistance values. Conduction velocity

in a 2-D tissue was also measured with different values of axial resistance.

In order to fully understand uterine contractility, multi-dimensional models

must be constructed with an aim to eventually reach organ level to enable the

possibility that we can control excitability and contraction in myometrium, giving

insights into areas such as pre-term labour. At the present time however, 1-D is

a good start on the path to making a clinical impact.

7.2 1-D strand model development

Myometrial smooth muscles show cable like properties [124, 220], and the cable

equation for a continuous cable is widely used in nerve and cardiac tissue mod-

elling to describe propagation through a single strand of tissue [221–226]. The

following reaction-diffusion-type non-linear cable equation was used in developing

the 1-D myometrial strand model.

∂V

∂t= ∇·D∇V − Itot

C(7.1)

where Itot is the total ionic current in a single cell level, C is the cell membrane

capacitance, x is the separation of the cells and D is a diffusion coefficient. Figure

7.1 shows the definition of x in a 1-D strand of cells. Experimentally, x was mea-

sured to be 0.015 µm [157]. The diffusion coefficient controls how currents and

electrical signals travel through intracellular gap junction in excitable media [215].

The experimental value of D measured from 20-day pregnant longitudinal rat tis-

sue strips was 0.24± 0.09× 10−2 µm2/s [211]. However, different combination of

172

these parameters were used in trial simulations (results not shown) and parame-

ters were adjusted to ensure wave propagated along the strand while maintaining

a normal cellular potential.

x

lc

D

Figure 7.1: A chain of myometrial smooth muscles demonstrates pa-rameters in the 1-D strand model. Adjacent cells in a strand are linkedby low resistance gap junction which is equivalent to a cylinder with a seriesof porous membranes. Cells with cell length, lc are separated by a cell-to-cellchannel or length x. The longitudinal diffusion movement (from left to right) in asingle strand depends on the diffusion coefficient, D, which controls how electricalsignals propagate along the strand [211].

The diffusion term of equation 7.1, i.e. ∇·D∇V defines the dimension of

the model and the reaction term, i.e. Itot /C describes the myometrial model

in a single cell level. In the current 1-D strand model, only one direction was

considered, i.e. action potentials could only propagate along direction x, and

therefore equation 7.1 becomes:

∂V

∂t=

∂D

∂ x

∂V

∂ x− Itot

C(7.2)

It was assumed that resistance was uniform and the ionic concentration did not

change over the time of interest. Myometrial cells were assumed to be uniformly

coupled and D was spatially homogeneous. Defined pacemaking sites in the uterus

are still not located anatomically [15,215]. It is still not known which uterine cells

are pacemaking cells, or whether all uterine cells have pacemaker activity [13].

In the 1-D strand model, the signal was initiated at a defined point for simplicity

173

and a current stimulus from 0 to -1 pA/pF for 2 s was applied to the first 10

cells. Conduction velocity and the resting membrane potential were measured in

the strand containing 50 cells.

1-D myometrial strand

∂V

∂t=

∂D

∂ x

∂V

∂ x− Itot

C(7.2)

Parameters Values Definitionx 0.028 µm Separation of cellsD 0.003 µm/ms Diffusion coefficientdt 0.01 ms Time step

Table 7.1: Parameter values of 1-D strand.

7.3 Results

A current stimulus pulse (from 0 to -1 pA/pF) was applied from t = 2 s to t

= 4 s, to the first 10 cells in the strand to investigate how the action potential

propagated through a chain of cells. As shown in figure 7.2, the AP was slowly

dampened as it propagated along the strand. Vmax was dropped to around -

48 mV at the end of the strand compare to about 0 mV at the beginning of

the strand. Although the AP was able to travel along around 60 cells, further

propagation was not observed, even with different parameter values of D and x

in trial simulations (not shown). When x was set too high (about 0.01 µm),

while D was set to the experiential observed value (0.24 µm/ms), the AP did not

propagate properly and the maximum potential Vmax was too low. In some cases,

the AP was only able to propagate to only the next 10 cells and the maximum

potential, Vmax was about -40 mV (compared to 0 to 10 mV in a single cell under

control condition).

It is also clearly shown that the AP was damped as it progressed through the

strand, i.e. it did not cause a full excitation. The amplitude of the AP was about

89% smaller at the end of the strand (50th cell) compare to the AP observed

at the beginning of the strand (1st cell). The resting membrane potential was

constant (about -59 mV) from the beginning to the end of the strand. A 1-D

strand changed the shape of the AP from a bursting type, previously seen in a

174

single cell, to a plateau type AP. This was due to the electrical loading of the

neighbouring cells. The electrical load affected the potential of the strand and

dampened any bursting seen in a single cell.

A comparison of conduction velocities from different gestation of the uterus

in human and rat are listed in table 7.2. In general, studies have found that the

conduction velocity increased towards delivery in pregnant rat tissues. However

no difference in conduction velocity was observed between delivery and before

term tissues from research conducted by Sims et al. [188]. Human uterine tissues

extracted during labour has a lower propagation velocity than tissues extracted

from rats. The conduction velocity obtained from simulations was 1.32 cm/s,

which is slightly slow for rat myometrium but still agrees with the experimental

values.

175

1 2

3 4

5t (s) 0

10 20

30 40

50

No. of cell

-60

-50

-40

-30

-20

-10

0

V (mV)

-60

-50

-40

-30

-20

-10

0

1 1.5 2 2.5 3 3.5 4 4.5 5

t (s)

0

10

20

30

40

50

no. o

f cel

l

-60

-50

-40

-30

-20

-10

0

Figure 7.2: AP conduction in the 1-D myometrial strand model. Resultsare shown in 3-D (top) and 2-D (bottom) where the membrane potential of thestrand is represented by the colour key on the right. The first 10 cells of thestrand were stimulated with a brief current pulse. The shape of the AP becamea plateau, rather than bursting type as previously seen in a single cell due tothe electrical loading from neighbouring cells. The AP was also dampened as itprogressed.

176

Model

orA

nim

alR

efer

ence

Con

duct

ion

velo

city

Ges

tati

onT

ype

ofti

ssue

exper

imen

t(c

m/s

)an

dre

gion

Exper

imen

tR

atK

urj

aket

al.[2

27]

9.2

-10

.5D

eliv

ery

Lon

gitu

din

al,m

yom

etri

um

Exper

imen

tR

atM

ille

ret

al.[2

13]

9.2

Lat

eLon

gitu

din

al,m

yom

etri

um

10.5

Del

iver

yLon

gitu

din

al,m

yom

etri

um

Exper

imen

tR

atO

hka

wa

[158

]1.

3to

512

-15

dpre

gnan

tLon

gitu

din

al,m

yom

etri

um

Exper

imen

tR

atSim

set

al.[1

88]

3.4±

0.5

Bef

ore

term

Myo

met

rium

3.4±

0.5

Del

iver

yM

yom

etri

um

Exper

imen

tR

atSpec

ht

[228

]8.

46±

0.96

Pre

gnan

tU

teru

sE

xper

imen

tR

atW

olfs

etal

.[2

29]

4.2

to9.

6P

regn

ant

Ute

rus

Exper

imen

tR

atSm

ith

[230

]13

.5±4

.2Par

turi

ent

Myo

met

rium

Exper

imen

tH

um

anW

olfs

and

Rot

tingh

uis

et[2

31]

2.5

to5

Lab

our

Ute

rus

Model

Rat

Rih

ana

etal

.[2

32]

abou

t10

Pre

gnan

tU

teru

sM

odel

Rat

Sim

ula

tion

1.32

Lat

eLon

gitu

din

al,m

yom

etri

um

Table

7.2

:C

om

pari

son

of

conduct

ion

velo

citi

es

inra

tand

hum

an

from

vari

ous

gest

ati

on.

The

last

entr

yw

assi

mula

ted

usi

ng

the

curr

ent

1-D

myo

met

rial

stra

nd

model

.

177

Chapter 8

Discussion

A biophysically detailed mathematical model of a single late pregnant rat myome-

trial cell was constructed. The model successfully reproduced electrical, chemical

and mechanical behaviour of a single cell. The simulated results were validated

using available experimental recordings. Drug actions and sensitivity analysis

were also utilised for the model validation, as this highlighted some limitations

of the model and provided guidance for future improvements. The cell model

was further integrated into a 1-D strand to investigate how an electrical signal

propagate along a fibre.

8.1 Whole cell membrane, SR currents model

In the whole cell myometrial model, all the major currents identified experimen-

tally so far are included and the descriptions in the model of all the currents agreed

well with the experimental voltage- and current-clamp recordings. The model

was divided into electrochemical and chemo-mechanical subsystems. The elec-

trochemical subsystem consisted of five inward currents ( Ih , ICaL , ICaT , INa and

ISOC ), five outward currents ( IK1 , IK2 , IKA , IKCa and IKleak ) and two other cur-

rents that flows in both directions ( ICl and INSCC ). Ionic pumps, exchanger and

co-transporter currents ( ICapump , INaK , INaCa and INaKCl ) were included to reg-

ulate the cellular concentration across the membrane. A simple calcium-induced

calcium release mechanism in the SR was also described. Calcium buffering and

the balance of intracellular Na+ and K+ were also taken into account in the

myometrial model.

The reversal potential for ICaL and ICaT in the myometrial model was fixed

178

at 45 mV. Although this approach might be less realistic, it was proven to be

a good estimation since the simulations of ICaL and ICaT fitted the I-V relation-

ships from the experimental data well. However, an improved description of

the Ca2+ reversal potential is needed for a more realistic depiction of the cell

behaviours and activities.

Unlike cardiac and other types of muscles, there are only limited experimental

data specifically for ionic channels in myometrium available to date. Most of the

time, extractions of data from raw experimental recordings in published literature

were often needed and this increased potential errors in the model. When data

in myometrium was not available, for example: IKCa , ISOC and INaKCl , record-

ings from different types of smooth muscle data in similar excitable organs were

adapted. However, experimental data often required modifying and rescaling,

which also increased the uncertainties in the model.

Furthermore, the model was developed based on body temperature experimen-

tal data recordings, but some experiments were carried out in room temperature.

Although a temperature coefficient, Q10, accounted for this temperature differ-

ence in Ih and INaK , when Q10 was not available, it was assumed the dynamics of

the channel do not change significantly. This limited the model’s ability to predict

cell behaviour, since some macroscopic gating properties, for example: steady-

state inactivation, inactivation time and I-V relationship in ICaL , and also the

levels of [ Ca2+ ]i , all have different temperature dependencies and some changes

are highly dependent on the temperature [233].

Different types of potassium channels in myometrium are poorly differenti-

ated, meaning only limited experimental data can be used to construct the model.

The lack of experimental data, especially in activation and inactivation time con-

stants, highly affected the shape of I-V relationship as well as the rise and decay

kinetics in current traces. This in turn imposed an effect on the whole cell action

potential.

There are also some disagreements over the role and the significance of the

Ca2+ -activated Cl− current in myometrial cells. A research group reported the

functional importance of ICl in the plateau phase of the AP [15,32], while another

independent research group claimed that Cl− channels are not important in uter-

ine electrophsyiology at all [12]. Both groups however, agreed that only 30%

of the pregnant myometrial cells consist of Cl− channels and all of the samples

are from rat uterus. Also an inhibition of ICl significantly reduced the frequency

179

of the spontaneous contractions (this effect was seen in simulations). More de-

tailed experimental data is needed to clarify this dispute in order to improve the

myometrial model.

In the myometrial SR mode, a two-pool uptake and release compartment sys-

tem, which is used widely in cardiac modelling was adapted. In myometrium,

there is evidence that subcompartments exist within the SR and they are func-

tionally separated [234]. Although the majority of the cardiac models have

included a IP3-sensitive store, releasing Ca2+ from SR in the uptake comport-

ment [112,134,235–237], in the myometrial model, the uptake compartment con-

sists of only the SERCA pump to replenish the Ca2+ store by cytosolic Ca2+ and

no release mechanism was considered. Despite the fact that IP3 receptor has also

been identified in the uterus, studies from various species have suggested release

from IP3 receptor does not play an important role in activation of force [15].

However, omitting this mechanism may lead to inaccurate prediction of the cell

behaviour, as IP3 receptors still contribute and regulate the content of Ca2+ in

a cell. More detailed and qualitative analysis on how the receptor operates is

needed to fully describe the effects on a myometrial cell.

8.2 Electrophysiological responses

8.2.1 Cell dynamics in control condition

The membrane current model described and predicted the uterine activity rela-

tively well in terms of the shape of the AP and the magnitude of the spikes in

the normal physiological condition. The profile of the APs matched closely to the

experimental recordings observed in Okabe et al. and Inoue et al. [71,108]. This

spiking type of AP can be found in less than 50% of the non-labouring late preg-

nant myometrial cells in human [15]. The I-V relationship of the peak outward

current agreed relatively well with the experimental data (figure 3.2), however

the inward current was slightly exaggerated as a consequence of a larger than

normal contribution of the Ca2+ -related currents. The inward current peaked at

around -8 mV in simulations while the reported experimental value was around

0 mV [26,71].

180

8.2.1.1 Repetitive depolarisation

When the myometrial model undergos repetitive depolarisation, it was shown

that the maximum peak of ICa−tot did not decay as quickly as it did in the ex-

periment. The incomplete recovery of [ Ca2+ ]i was more severe in experiments

than results produced in simulations. This was due to the Ca2+ extrusion mecha-

nisms, with for example, the Ca2+ pump and Na+ /Ca2+ exchanger set too strong

in the model. However, trial simulations showed that if the strength or the mag-

nitude of the extrusion mechanisms were not as strong (results not shown), the

[ Ca2+ ]i transient would not decay after reaching its initial peak when the model

was held at a voltage-clamp for a long period of time (see control condition in

nifedipine drug action). If the magnitude of the extrusion mechanisms were set

higher, the peaks would be negatively shifted when the model was depolarised

with various magnitude pulses, and deviate from the experimental recordings (see

figure 5.1). On balance, the model’s parameters were chosen so that the results

would best match the most features from the experimental data.

8.2.2 Cell dynamics under estradiol condition

The myometrial model mimicked an estradiol effect by reducing the conductances

of the voltage-gated, Ca2+ -activated K+ currents and voltage-gated Ca2+ currents

by 30-60%. The model was able to produce a plateau type AP with an elevated

level of RMP compared to control (-56 mV), similar to the recordings seen in

experiments [108]. Spikes were suppressed as described in experiments and only

the first spike was observed. The corresponding [ Ca2+ ]i peaked at 462 nM, and

a smooth crescent-like profile with a summation of [ Ca2+ ]i during the clamp was

presented. The I-V relationship showed the magnitude of the total cell current

was reduced in both inward and outward directions in estradiol condition (since

the conductances of the K+ and Ca2+ currents were reduced). Both inward and

outward currents fitted well with the experimental data, although the peak of the

inward current was left shifted toward the negative membrane potential.

The description of the estradiol effect in the myometrial model is still incom-

plete. Yamamoto reported the voltage-dependent inactivation curve of Ba2+ was

left-shifted in cells treated with estradiol [68]. Estradiol also influences a my-

ometrial cell in a dose- and time-dependent manner. In studies carried out by

Okabe et al., the concentration of estradiol used in the experiment was about

181

3 µM higher than the normal level during pregnancy (the 17β-estradiol in the

serum level is about 30 to 100 nM in human [159]). Such high concentration

was needed to observe the inhibition of uterine contractions. Furthermore, it was

reported that the longer estradiol was present in the cell, the greater the inhi-

bition of Ca2+ and K+ currents. In order to improve the myometrial model, all

this information will have to be considered and more detailed studies have to be

carried out.

8.2.3 Inhibiting individual current

In experiments carried out in estrogen-treated non-pregnant rat tissues, it was re-

ported that myometrial excitation was not sustained by only Ca2+ or Na+ alone

[238]. It is shown in simulations that inhibiting ICaL and INa have similar effects

on the AP. The profile of the AP dramatically changed, spikes were suppressed

and Vmax was noticeably lowered, but the resting membrane potential remained

approximately the same. Blocking ICaL completely diminished the inward por-

tion of the total membrane current, but had little effect when INa was inhibited.

This proved that ICaL is the major contributor of the total inward current in the

myometrium [13].

Inhibiting INaCa slightly lowered the resting membrane potential, but sup-

pressed the generation of spikes without noticeably altering the magnitude of the

total membrane current. In experiments, it was suggested that the electrogenic

Na+ pump plays an important role in the normal resting membrane potential in

smooth muscles [239] and myometrium [240]. Experiments on guinea pig taenia

muscles revealed that blocking the Na+ pump with ouabain lowered the resting

membrane potential [241] and this decrease was due to the loss of K+ in the

cell [242].

Inhibiting K+ currents, IK1 and IK2 , has a dramatic effects on the AP in simu-

lations. The resting membrane potentials in both cases were raised significantly,

no spikes were formed (apart from the initial spike which was slightly raised)

and the APs were not returned to their original values. Blocking both currents

individually reduced the outward part of the total membrane current. The ef-

fect of blocking the fast K+ current was examined in experiment using TEA, a

K+ blocker, in non-pregnant guinea-pig myometrial tissues [243]. It was recorded

that spike amplitude was increased in the AP with a slow repolarisation phase,

undershoot was abolished and the membrane resistance was increased when a

182

current pulse was applied. A noticeable increase was also recorded in the in-

ward current. The differences in the results might be due to the remodelling the

K+ channels undergo during pregnancy, which modify the physiological responses

in the myometrial cell.

Jones et al. reported that inhibiting ICl significantly decreased the frequency

of the uterine contractions [32]. This effect was observed in simulations by block-

ing ICl and reduced the frequency of the spikes in the AP without significantly

changing the spike amplitude, resting membrane potential and the total current.

The non-specific cation channel, NSCC is responsible in maintaining the

pacemaking activity and regulates the frequency of the contraction in the my-

ometrium. Miyoshi et al. investigated the effects of inhibiting one of the NSCC

mechanisms, ATP receptors using Mg2+ [244]. It was reported that the ATP-

induced depolarisation was suppressed in 18-22 days pregnant isolated rat my-

ometrium. In simulations illustrated in figure 3.12, the resting membrane po-

tential was significantly reduced and the spikes were suppressed when INSCC was

blocked.

8.3 Cell mechanics and force generation

In the chemo-mechanical subsystem, the myometrial model adapted the Hai and

Murphy 4-state model to describe the myosin phosophrylation process, which

was developed originally for smooth muscles. This model was then coupled to the

Yang cerebrovascular model to generate mechanical contractions. The mechanical

model was described by separate force components (passive, active, visocelastic,

cross-bridge) that coupled together to create tension. The equations and param-

eters were mostly unmodified since the data for uterus was not available.

The myometrial model successfully predicted contractile activities in single,

twin and multiple stimulations with characteristics similar to experimental record-

ings observed in myometrium. When a train of repetitive depolarisation pulses

were applied to the model, it was shown that the maximum stress level generated

by this protocol was higher than a single pulse stimulation, although the durations

of the contraction was approximately the same. Ca2+ -spikes from the twin-pulse

simulations illustrated in figure 4.2 shows that the Ca2+ decayed slower compared

to the experimental recordings. The [ Ca2+ ]i transient elicited from the first pulse

was not able to fully recover before the next pulse was applied in simulations,

183

this indicates the rate of the Ca2+ extrusion mechanism in the myometrial model

might be too slow.

The incorporation of the cerebrovascular mechanical model from Yang et al.

allowed the myometrial model to predict cellular events that are not easily mea-

sured. This includes the temporal distribution of the cross-bridge cycling states

and the changes in the internal length components. It is shown in figure 4.4 that

la was shortened when a stimulus voltage pulse was applied. Both ls and lx

expanded to compensate for this shortening, but the expansion of lx was much

shorter compared to ls .

The magnitude of the total force from simulations was considerably lower

compared to experiential data. This is because the majority of the parameter sets

obtained from the Yang et al. model were unchanged due to a lack of experimental

data for myometrium. Since the myometrial model assumed Ca2+ -Calmodulin-

MLCK is the only excitation pathway, the low magnitude of the total force may

also be due to the model’s exclusion of the extra pathways which lead to force

generation, for example: agonist- and stretch-induced contractions.

8.4 Drug actions

Imitating different drug actions in simulations can serve as a validation of the

model, and also address the limitations of the model. Drug action simulations

were carried out to focus on the effects drugs have on [ Ca2+ ]i transient and

contractile force. As discussed previously, simulation times were carried out in a

shorter time scale than the experiments due to the instability of the model in a

long duration of time.

The mechanisms responsible for the lowering of the intracellular Ca2+ when a

cell is relaxed are the plasmalemmal Ca2+ pump and the Na+ /Ca2+ exchanger.

Although the sarcolemmal mechanisms do play a part in reducing the level of

[ Ca2+ ]i , the contribution from them is relatively small since the SR Ca2+ stores

have a finite capacity [123]. When the plasmalemmal Ca2+ pump was inhibited

using carboxyeosin in experiments demonstrated by Shmigol et al. [123], the

decay rate of [ Ca2+ ]i markedly reduced after a train of 10 voltage pulses were

applied compared to the control case. No detectable change in the current was

observed in carboxyeosin treated cells. In simulations, although a slight rise

in the peak [ Ca2+ ]i and the resting level of [ Ca2+ ]i was recorded similar to the

184

experiments when ICapump was blocked, the decay of the [ Ca2+ ]i transient was not

changed significantly compared to the control case. The inconsistency may due

to the tissue and species differences of the adopted equations describing INaCa and

ICapump . However, there are some conflicting results in the literature. Shmigol et

al. found that the Ca2+ pump contributes to about 30% of the Ca2+ decay and

the remaining 70% is attributed to the Na+ /Ca2+ exchanger and the SR-related

mechanisms. But Kosterin et al. claimed that Ca2+ pump is the most dominant

Ca2+ extrusion mechanism in myometrium [245]. More studies have to be done to

clarify the confusion in order to provide better descriptions of the Ca2+ extrusion

mechanisms, INaCa and ICapump .

The L-type Ca2+ current proved to be the major contributor of [ Ca2+ ]i ,

ICa−tot and mechanical contractions since inhibiting the current nearly completely

abolished both the [ Ca2+ ]i transient, ICa−tot and force. However, the inconsis-

tency in the [ Ca2+ ]i decay caused by the inaccurate descriptions of INaCa and

ICapump , also affected the simulated results when a long voltage pulse (10 s) was

applied. As illustrated in figure 5.3, although the upstroke of the [ Ca2+ ]i fit well

with the experimental data in control, the decay rate of the [ Ca2+ ]i transient

was noticeably slower than the experimental recordings.

The effect of ryanodine was also examined in simulations and it was found

that when a twin-pulse protocol was applied, the second increment in [ Ca2+ ]i was

slightly smaller than the first increment. Whereas in the experiments, the dif-

ference between the first and the second peaks with the presence of ryanodine

was not notable. A negative staircase effect was observed in both simulations

and experiments, i.e. the second peak of the [ Ca2+ ]i transient increased while

the corresponding peak of the total Ca2+ current decreased. It is important to

note that a relatively low dose of ryanodine (5 µM) was used in the experiment,

and such a low dose is not sufficient to totally inhibit CICR, but rather slowly

dissipate and eventually exhaust the releasing of Ca2+ from the SR [69]. Since

Irel was blocked completely in simulations, this may contribute to the discrepancy

obtained in simulations.

Carbachol increased the maximal force compared to control cells in experi-

ments, by releasing Ca2+ from the internal SR store. This effect was replicated

in simulations and the results agreed well with experimental data. The myome-

trial model also successfully replicated the CPA response of the [Ca2+]i transient.

Simulations showed the amplitude of the [ Ca2+ ]i transient increased when CPA

185

was applied. The basal level of [ Ca2+ ]i was raised and gradually declined as time

progressed. It also showed the prolonged duration of the [ Ca2+ ]i transient and

force when Iup was inhibited, similar to experimentally observed data [136]. How-

ever, unlike experimental recordings, the basal level of force was also increased

in response to the elevated level of [ Ca2+ ]i in simulations. It is unclear why the

contractile proteins were not affected in CPA treated cells [13], more research has

to be done experimentally in order to improve the myometrial model.

Application of Wortmannin to rat myometrial cells showed the spontaneous

contractions were gradually abolished while the [ Ca2+ ]i level and ICa−tot were

unchanged. The model was able to demonstrate this effect when MLCK was

inhibited, but the gradual decline in force was not seen. This is due to the as-

sumption that no additional force generating pathways, apart from Ca2+ -CaCM-

MLCK exisit in the myometrial model. In reality, various different pathways can

also contribute to the generation of force without increasing the level of Ca2+ .

Luckas et al. demonstrated that agonist-induced (e.g. oxytocin) contractions can

fire a single contraction in human pregnant myometrium at term in the absence of

extracellular Ca2+ [28, 246]. This explains the rapid declination in force seen in

simulations when the effect of Wortmannin was applied. The level of [ Ca2+ ]i was

unchanged when MLCK was inhibited. Including the Ca2+ -independent force

producing pathways will provide a more realistic approach in predicting the con-

tractile activities in myometrium.

When the effect of Ca2+ -free solution was demonstrated on myometrium, it

was found that the total contractile force declined to zero and raised back to

its original level when the Ca2+ was re-introduced. The level of [ Ca2+ ]i slowly

declined in both experiments and simulations as the balance of Ca2+ in the cell

was restored, thus confirming the importance of external Ca2+ in contractions.

8.5 Sensitivity Analysis

Sensitivity analysis was performed to predict the physiological effects it has on

changing parameters. This also tested the model’s ability to see whether the

model can be used in different gestation of the pregnancy.

The cellular surface area to volume ratio has a direct effect on the intracel-

lular concentration of Na+ , K+ , Cl− and Ca2+ . It is also inversely related to

ICapump and INaCa . The total force reached a maximum faster when the surface

186

area to volume ratio was increased, although the duration of the contraction

remained approximately the same when the ratio was varied.

Using the current mechanical descriptions of the force and length of the cell

from Yang et al. cerebrovascular smooth muscle model, the analysis showed

that contraction increased until lc reached 150 µm, and further extension of

the length restricted force generation drastically. This is most likely due to the

constraint imposed by the existing force model equations which were optimised

to the length of the cerebrovascular cell (123 to 140 µm). Although the length

of a late pregnant myometrial cell in rat is about 200 to 700 µm, parameters of

all the force components for examples: the series and parallel stiffness constants,

optimal length of the active force component and cell length at zero passive force

were intended for cells with cell length similar to a vascular smooth muscle cell.

Therefore the parameter set belonging to the mechanical model will have to be

adjusted before applying to the myometrium.

Changing the current density of ICa−tot provided the most effective way of

raising the level of [ Ca2+ ]i , since the L-type Ca2+ channel is the major contrib-

utor to the Ca2+ influx in myometrium. When increasing the current density of

ICa−tot to four times the original value, the [ Ca2+ ]i level was increased drastically

to 334.65%, however the increase in contractions was only 53.73% compared to

the control case. This indicates other factors and mechanisms, not just the rising

level of [ Ca2+ ]i , also played a role in force generation. These factors include the

cell surface area to volume ratio and the length of the cell discussed previously.

Furthermore, varying values of ICa−tot changed the activation and inactivation

kinetics of the total force. When the influx of Ca2+ increased, the corresponding

contractions were more pronounced and lasted for a shorter time despite being

increased in magnitude. In contrast, the contractions were more sustained when

less Ca2+ was introduced to the cell.

Varying the current densities of INa−tot and IK−tot did not significantly affect

the maximum, time to peak and the relaxation of the contractions. This is due

to the fact that both Na+ and K+ currents do not play an important role in

force generation in myometrium. However, the decay of the [ Ca2+ ]i transient

was influenced strongly by the intracellular content of Na+ through the Na+ -

Ca2+ exchanger. Although the activation kinetics were not affected, the increase

of the influx of Na+ caused the [ Ca2+ ]i transient to decay faster in the model.

187

This contradicted the results obtained reported from Shmigol et al. [69]; the de-

cay of the [ Ca2+ ]i should be steeper when less Na+ was presented in the cell. As

discussed previously in carboxyeosin drug actions, the discrepancy might due to

the model’s description of INaCa . The equations describing INaCa were adapted

from the Hund-Rudy canine ventricular model, and not from pregnant rat my-

ometrium. Such tissue and species specific differences may account for the dis-

agreement.

Changes of the level of MLCK directly affected the magnitude of the contrac-

tions, because the Ca2+ -calmodulin-MLCK process was assumed to be the only

pathway for force generation in the myometrial model. Increasing the MLCK

level not only almost linearly elevated the basal and maximal contraction, it also

accelerated the activation kinetics so contractions reached their peak faster.

8.6 1-D strand

The myometrial cell model was integrated into a more complex description. A

1-D strand model was developed using a reaction-diffusion-type cable equation

to investigate how an action potential propagated along a fibre containing 50

cells. It was assumed that the myometrial cells were uniformly coupled and the

diffusion coefficient was spatially homogeneous. A current stimulus pulse was

applied to the first 10 cells in the strand for 2 s to investigate how the action

potential propagated.

Simulations from a 1-D strand model show the resting membrane potential

agreed well with the experimental values and stayed constant, while the AP

dampened along the strand. The conduction velocity measured from the model

was 1.32 cm/s which was slower compared to the experimentally observed values

in rat. Simulations also showed the AP was able to propagate along the strand

when the first 10 cells were stimulated. However, test simulations showed that

propagation over 60 cells were not observed even when different values of param-

eters were used. This may indicate that myometrial cells do not initiate electrical

signals from a defined area which consists of pacemaking cells unlike cardiac tis-

sues [227]. This also suggest that myometrial cells may have pacemaker activity

as proposed by various research groups [15, 247]. It was demonstrated in exper-

iments that multiple pacemaker sites were found in progesterone-dominated rat

and rabbit myometrium [248, 249]. More simulations have to be conducted with

188

multiple stimulating sites randomly distributed to clarify this finding.

8.7 Model limitations

The model successfully replicated the electrophysiological and mechanical activi-

ties seen in experimental recordings, however, the model was limited by the lack

of experimental data available for some of the membrane currents and the ex-

changer. Qualitative data on the SR was also not available in the myometrium

and the descriptions from other tissue types and different animals were therefore

substituted. Since the myometrium has unique characteristic which are not seen

in other smooth muscles, this might give rise to the discrepancies seen in some

simulated results. Furthermore, spatial heterogeneity of the cell, stretch- and

agonist-induced contractions are all important factors in regulating the myome-

trial cell function during pregnancy. These factors have to be considered in future

generations of the model in order to enhance the prediction of the myometrial

cell activity.

8.7.1 Mixing species and tissues data

Due to the shortage of late-pregnant rat myometrial data, descriptions of the

equations and parameters from human and other animals or different similar tis-

sues were substituted in the model. These equations were then adjusted to fit

the experimental recordings from myometrium. This limits the model’s ability

to accurately reproduce results seen in experiments, both in ionic currents and

contraction related data. Shmygol et al. addressed that there are clear discrep-

ancies between human and rat myometrial pacemaking mechanisms, which leads

to the concerns whether mechanisms found in rodent models can be eventually

applicable to human myometrium, and how relevant rodent studies are to hu-

man [15,201]. Different types of tissues also posed discrepancies and ideally, the

model will be significantly improved with additional myometrial data in both

species. Extra data is needed especially on INaCa and ICapump which greatly af-

fected the model’s output according to simulations of drug actions and sensitivity

analysis.

189

8.7.2 Stretch-induced effects on myometrium

Stretch-induced channel currents have been identified in various smooth muscles,

pregnant and non-pregnant myometrial cells [193, 250, 251], and is also studied

extensively in myocardium [252]. Kasai et al. reported that stretching tissues or

cells enhances uterine contractions in rats [253]. It induces a myogenic transient

contraction due to a sudden Ca2+ influx, the contraction generated depends on

the excursion and velocity of the stretch, i.e. rapidly stretching a tissue 30 to

35% its original length for 0.2 to 0.5 s create maximum contractions, whereas

stretching slowly rarely induces a contraction. In human cultured myometrial

cells, Dalrymple et al. found that stretch changes the [ Ca2+ ]i signalling and the

transient receptor potential canonical (TRPC) isoform expression [196]. This

effect has been described in various smooth muscle cell models [49], but it has

never been quantified in myometrium due to lack of experimental data. Including

this effect may increase the total force to a magnitude similar to that observed

experimentally.

8.7.3 Spatial heterogeneity

Spatial heterogeneity has been widely discussed in cardiac modelling [254–257],

but not in smooth muscles. The majority of existing smooth muscle models

assume spatial homogeneity and ignore some of the internal cellular compart-

ments. It has been reported that spatial heterogeneity in the cell result in dy-

namic changes and complex regulatory function which can affect physiological

responses in gastrointestinal and myometrial cells [4,36,135,179,258–262]. How-

ever more detailed studies are needed to fully describe the complicated structure

and dynamics in the SR.

8.7.4 Addition force generation pathways

As demonstrated in Wortmannin and force simulation, the description of con-

tractile force in the myometrial model is incomplete. Additional contractile force

generation pathways, aside from Ca2+ -CaCm-MLCK, do exist and the exclusion

of the extra pathways has therefore introduced greater limitations to the model.

Ultratonic agonists such as oxytocin, endothelin-1, carbachol, prostaglandins

and acetylcholine are known to increase [ Ca2+ ]i and contraction in myometrium

190

[246, 263, 264]. Various agonist-induced uterine contractions through protein ki-

nase C (PKC) were reported but not quantified. Endothelin-1 was found to be

involved in contraction through PKC in both non-pregnant [265] and late preg-

nant human tissues [266, 267]. In isometric force studies on human longitudinal

tissues, it was shown that the activation of PKC was vital for oxytocin-induced

contraction in labour [268]. However, this contradicted studies performed in

pregnant rat myometrial tissues in which it was reported that PKC activation

suppressed phasic contractions [269]. Furthermore, it was demonstrated that

phorbol 12,13-dibutylate also induced sustained contractions without increasing

[ Ca2+ ]i in both non-pregnant and pregnant human myometrium [263]. Exclu-

sion of all these mechanisms may result in the low magnitude of the total force

recorded and a rapid drop in contraction when MLCK was inhibited in simulation.

Therefore, to enhance the model’s ability in predicting the excitation-contraction

coupling in a myometrial cell, extra pathways must be considered.

8.7.5 1-D strand

The incorporation of the 1-D strand model was a simple one. The stimulus sites

initiated propagation at the beginning of the strand and occupied 20% (10 cells)

of the length of the total strand (50 cells). Although the number of stimulus sites

was relatively high, it was necessary for sufficient propagation along the strand.

However, there is no evidence as yet to support a defined anatomical pacemaking

site in the uterus, and if the pacemaking sites were distributed in several different

areas, less stimulus sites may be needed.

191

Chapter 9

Future plan

The understanding of the regulation of uterine function and contractility is in-

complete. As mentioned in the introduction, pre-term birth is by far the most

common cause of mortality and morbidity around the world and 40-45% of all pre-

term births are the result of pre-term labour [270]. The precise cause of pre-term

labour still remains elusive and little has changed in the last 40 years [271]. Stud-

ies have identified four different pathways that can cause pre-term labour, if one

or more of the pathways are activated. They are: 1) precocious fetal endocrine

activation, 2) uterine over-distension, 3) decidual bleeding and 4) intrauterine in-

flammation or infection [272]. However, prevention of pre-term labour has proven

difficult. Most of the drugs available to date lack uterine selectivity and may

present serious side effects to both mother and foetus [273]. Further development

of the myometrial model may help in the testing or validation of theories and

hypotheses raised. This may help in the identification of the underlying causes

of pre-term labour and allow better drugs to be developed for its treatment.

9.1 Single cell level

Simulated results highlighted some limitations of the myometrial model which

need to be addressed in order to improve the model. As discussed previously, there

are small but significant functional differences between human and rat myometrial

cells and tissues. Focus on developing a purely human myometrial model will

provide more accurate predictions when integrated into a tissue model. It has

been shown in experiments that the effects of oxytocin are important during

pregnancies in both human and rats. Oxytocin is a naturally occurring drug

192

produced in the third stage of labour [171]. The release of the drug provides

extra force, necessary to expel the foetus during delivery (the drug can also be

given externally). The effect of oxytocin has never been quantified in a modelling

environment. Stretch-induced contraction also proved to be significant, especially

during late gestation of the pregnancies [195] - adding these mechanisms may be

beneficial. The calcium handling of the model is a simple process. A more

complex system, for example including descriptions of IP3 receptors in the cell, is

needed in order to accurately predict the cellular calcium activities. Other force

generation pathways, aside from Ca2+ -CaCm-MLCK, should also be added in

the future generations of the model as it provides a more realistic approach.

Modelling G protein signalling and second messenger formation is also impor-

tant, since experimental evidence has shown that the increased levels of G α s,

promotes the effect of agonists that increase cAMP formation which assist re-

laxation in the uterus. Spontaneous labour may occur when the levels of G α s

drop, triggering contractions before term [273]. Therefore the management of this

G protein is vital during pregnancy and modelling this mechanism will provide

deeper insights into the cause of pre-term labour.

9.2 Tissue level

More detailed 1-D studies are needed to develop the model further. It is also

necessary to further integrate the 1-D strand model into a more complex 2-D

or 3-D model. Stimulations can be initiated from multiple sites (randomly or

fixed) to investigate electrical and mechanical activities during contraction and

observe how these signals travel across a tissue. This may help clarify and explain

whether all uterine cells have the capacity to act as pacemaker cells to initiate

spontaneous contractions, or if the uterus has defined pacemaking cells where

contractions are generated from.

193

Bibliography

[1] Wray S. Uterine contraction and physiological mechanisms of modulation.

Am J Physiol 1993;264(1 Pt 1):C1–18.

[2] Khan RN, Matharoo-Ball B, Arulkumaran S, Ashford ML. Potassium chan-

nels in the human myometrium. Exp Physiol 2001;86(2):255–64.

[3] Hall. The importance of preterm birth. Churchill Livingstone, London,

1997.

[4] Wray S, Jones K, Kupittayanant S, Li Y, Matthew A, Monir-Bishty E,

Noble K, Pierce SJ, Quenby S, Shmygol AV. Calcium signaling and uterine

contractility. J Soc Gynecol Investig Jul 2003;10(5):252–264.

[5] Adelstein RS, Hathaway DR. Role of calcium and cyclic adenosine 3’:5’

monophosphate in regulating smooth muscle contraction. Mechanisms of

excitation-contraction coupling in smooth muscle. Am J Cardiol 1979;

44(5):783–7.

[6] Blackburn S, Loper D. Maternal, fetal, and neonatal physiology: A clinical

perspective. Saunders Philadelphia, 1992.

[7] Hurd WW, Gibbs SG, Ventolini G, Horowitz GM, Guy SR. Shortening

increases spontaneous contractility in myometrium from pregnant women

at term. Am J Obstet Gynecol Apr 2005;192(4):1295–301; discussion 1301–

3.

[8] Yoshino M, Wang SY, Kao CY. Sodium and calcium inward currents in

freshly dissociated smooth myocytes of rat uterus. J Gen Physiol 1997;

110(5):565–77.

194

[9] Young RC. Tissue-level signaling and control of uterine contractility: the

action potential-calcium wave hypothesis. J Soc Gynecol Investig 2000;

7:146–52.

[10] Lammers WJ, Stephen B, Hamid R, Harron DW. The effects of oxytocin

on the pattern of electrical propagation in the isolated pregnant uterus of

the rat. Pflugers Arch Feb 1999;437(3):363–370.

[11] Kawarbayashi T. The Uterus-Electrophysiology of the human myometrium.

Cambridge University Press, 1994.

[12] Young RC. Myocytes, myometrium, and uterine contractions. Ann N Y

Acad Sci 2007;1101:72–84.

[13] Wray S, Kupittayanant S, Shmygol A, Smith RD, Burdyga T. The physio-

logical basis of uterine contractility: a short review. Exp Physiol Mar 2001;

86(2):239–246.

[14] Horowitz B, Ward SM, Sanders KM. Cellular and molecular basis for electri-

cal rhythmicity in gastrointestinal muscles. Annu Rev Physiol 1999;61:19–

43.

[15] Shmygol A, Blanks AM, Bru-Mercier G, Gullam JE, Thornton S. Control of

uterine Ca2+ by membrane voltage: toward understanding the excitation-

contraction coupling in human myometrium. Ann N Y Acad Sci 2007;

1101:97–109.

[16] Duquette RA, Shmygol A, Vaillant C, Mobasheri A, Pope M, Burdyga T,

Wray S. Vimentin-positive, c-kit-negative interstitial cells in human and

rat uterus: a role in pacemaking? Biol Reprod Feb 2005;72(2):276–283.

[17] Sanborn BM. Ion channels and the control of myometrial electrical activity.

Semin Perinatol 1995;19(1):31–40.

[18] Wilde DW, Marshall JM. Effects of tetraethylammonium and 4-

aminopyridine on the plateau potential of circular myometrium from the

pregnant rat. Biol Reprod May 1988;38(4):836–845.

[19] Bengtsson B, Chow EM, Marshall JM. Activity of circular muscle of rat

uterus at different times in pregnancy. Am J Physiol 1984;246(3 Pt 1):C216–

23.

195

[20] Chamley WA, Parkington HC. Relaxin inhibits the plateau component of

the action potential in the circular myometrium of the rat. J Physiol 1984;

353:51–65.

[21] RUDZIK AD, MILLER JW. The effect of altering the catecholamine con-

tent of the uterus on the rate of contractions and the sensitivity of the

myometrium to relaxin. J Pharmacol Exp Ther Oct 1962;138:88–95.

[22] RUDZIK AD, MILLER JW. The mechanism of uterine inhibitory action

of relaxin-containing ovarian extracts. J Pharmacol Exp Ther Oct 1962;

138:82–87.

[23] Sanborn BM. Relationship of ion channel activity to control of myometrial

calcium. J Soc Gynecol Investig 2000;7(1):4–11.

[24] Gilloteaux J, Szczepanski M. The fibre dimensions of uterine smooth muscle

of the rabbit following treatment by female sex steroids. Tissue Cell Jun

2000;32(3):243–248.

[25] Parkington HC, Tonta MA, Brennecke SP, Coleman HA. Contractile activ-

ity, membrane potential, and cytoplasmic calcium in human uterine smooth

muscle in the third trimester of pregnancy and during labor. Am J Obstet

Gynecol Dec 1999;181(6):1445–1451.

[26] Okabe K, Inoue Y, Kawarabayashi T, Kajiya H, Okamoto F, Soeda H. Phys-

iological significance of hyperpolarization-activated inward currents (Ih) in

smooth muscle cells from the circular layers of pregnant rat myometrium.

Pflugers Arch 1999;439(1-2):76–85.

[27] Inoue Y, Sperelakis N. Gestational change in Na+ and Ca2+ channel

current densities in rat myometrial smooth muscle cells. Am J Physiol

1991;260(3 Pt 1):C658–63.

[28] Tribe RM. Regulation of human myometrial contractility during pregnancy

and labour: are calcium homeostatic pathways important? Exp Physiol

Mar 2001;86(2):247–254.

[29] Knock GA, Smirnov SV, Aaronson PI. Voltage-gated K+ currents in freshly

isolated myocytes of the pregnant human myometrium. J Physiol 1999;518

(Pt 3):769–81.

196

[30] Knock GA, Tribe RM, Hassoni AA, Aaronson PI. Modulation of potas-

sium current characteristics in human myometrial smooth muscle by 17beta-

estradiol and progesterone. Biol Reprod 2001;64(5):1526–34.

[31] Wang SY, Yoshino M, Sui JL, Wakui M, Kao PN, Kao CY. Potassium

currents in freshly dissociated uterine myocytes from nonpregnant and late-

pregnant rats. J Gen Physiol 1998;112(6):737–756.

[32] Jones K, Shmygol A, Kupittayanant S, Wray S. Electrophysiological charac-

terization and functional importance of calcium-activated chloride channel

in rat uterine myocytes. Pflugers Arch 2004;448(1):36–43.

[33] Miyoshi H, Yamaoka K, Garfield RE, Ohama K. Identification of a non-

selective cation channel current in myometrial cells isolated from pregnant

rats. Pflugers Arch 2004;447(4):457–64.

[34] Molleman A. Patch Clamping : An Introductory Guide to Patch Clamp

Electrophysiology. New York John Wiley & Sons, Ltd., 2003.

[35] Wray S, Ravens U, Verkhratsky A, Eisner D. Two centuries of excitation-

contraction coupling. Cell Calcium Jun 2004;35(6):485–489.

[36] Wray S. Insights into the uterus. Exp Physiol Jul 2007;92(4):621–631.

[37] Wray S, Burdyga T, Noble K. Calcium signalling in smooth muscle. Cell

Calcium 2005;38(3-4):397–407.

[38] Bursztyn L, Eytan O, Jaffa AJ, Elad D. Mathematical model of excitation-

contraction in a uterine smooth muscle cell. Am J Physiol Cell Physiol

2007;292(5):C1816–29.

[39] Shmigol AV, Eisner DA, Wray S. The role of the sarcoplasmic reticulum

as a ca2+ sink in rat uterine smooth muscle cells. J Physiol 1999;520 Pt

1:153–163.

[40] Word RA. Myosin phosphorylation and the control of myometrial contrac-

tion/relaxation. Semin Perinatol Feb 1995;19(1):3–14.

[41] Wray S, Noble K. Sex hormones and excitation-contraction coupling in the

uterus: the effects of oestrous and hormones. J Neuroendocrinol Apr 2008;

20(4):451–461.

197

[42] Matthew A, Kupittayanant S, Burdyga T, Wray S. Characterization of

contractile activity and intracellular ca2+ signalling in mouse myometrium.

J Soc Gynecol Investig May 2004;11(4):207–212.

[43] Sanborn BM. Hormones and calcium: mechanisms controlling uterine

smooth muscle contractile activity. the litchfield lecture. Exp Physiol Mar

2001;86(2):223–237.

[44] Sanborn BM. Hormonal signaling and signal pathway crosstalk in the

control of myometrial calcium dynamics. Semin Cell Dev Biol Jun 2007;

18(3):305–314.

[45] Noble K, Matthew A, Burdyga T, Wray S. A review of recent insights

into the role of the sarcoplasmic reticulum and ca entry in uterine smooth

muscle. Eur J Obstet Gynecol Reprod Biol May 2009;144 Suppl 1:S11–S19.

[46] Lodish H, Berk A, Zipursky SL, Matsudaira P, Baltimore D, Darnell JE.

Molecular Cell Biology. 4th edition. W. H. Freeman & Co., 1999.

[47] Martini F. Fundamentals of Anatomy and Physiology. 7th edition. Pearson

Education, 2006.

[48] Gestrelius S, Borgstrm P. A dynamic model of smooth muscle contraction.

Biophys J Jul 1986;50(1):157–169.

[49] Yang J, Clark JW, Bryan RM, Robertson C. The myogenic response in

isolated rat cerebrovascular arteries: smooth muscle cell model. Med Eng

Phys Oct 2003;25(8):691–709.

[50] Hai CM, Murphy RA. Cross-bridge phosphorylation and regulation of latch

state in smooth muscle. Am J Physiol Jan 1988;254(1 Pt 1):C99–106.

[51] Hai CM, Murphy RA. Regulation of shortening velocity by cross-bridge

phosphorylation in smooth muscle. Am J Physiol Jul 1988;255(1 Pt 1):C86–

C94.

[52] Fung Y. Biomechanics: mechanical properties of living tissues. 2nd edition.

Springer, 1993.

198

[53] Youm JB, Kim N, Han J, Kim E, Joo H, Leem CH, Goto G, Noma A,

Earm YE. A mathematical model of pacemaker activity recorded from

mouse small intestine. Philos Transact A Math Phys Eng Sci May 2006;

364(1842):1135–1154.

[54] Rihana S, Santos J, Mondie S, Marque C. Dynamical analysis of uterine

cell electrical activity model. Conf Proc IEEE Eng Med Biol Soc 2006;

1:4179–4182.

[55] Kapela A, Bezerianos A, Tsoukias NM. A mathematical model of ca2+

dynamics in rat mesenteric smooth muscle cell: agonist and no stimulation.

J Theor Biol Jul 2008;253(2):238–260.

[56] Hai CM, Murphy RA. Ca2+, crossbridge phosphorylation, and contraction.

Annu Rev Physiol 1989;51:285–298.

[57] Hai CM, Murphy RA. Crossbridge phosphorylation and regulation of vascu-

lar smooth muscle contraction. Am J Hypertens Aug 1990;3(8 Pt 2):235S–

237S.

[58] P. FC, Marland ES, Wagner JM, Tyson JJ. Computational cell biology.

Springer Science, 2002.

[59] Hodgkin AL, Huxley AF. A quantitative description of membrane current

and its application, to conduction and excitation in nerve. J Physiol 1952;

117:500–544.

[60] Satoh H. Identification of a hyperpolarization-activated inward current

in uterine smooth muscle cells during pregnancy. Gen Pharmacol 1995;

26(6):1335–8.

[61] Chang DC. Dependence of cellular potential on ionic concentrations. data

supporting a modification of the constant field equation. Biophys J Aug

1983;43(2):149–156.

[62] Miyoshi H, Urabe T, Fujiwara A. Electrophysiological properties of mem-

brane currents in single myometrial, cells isolated from pregnant rats.

Pflugers Arch 1991;419(3-4):386–393.

199

[63] Sperelakis N, Inoue Y, Ohya Y. Fast Na+ channels and slow Ca2+ current

in smooth muscle from pregnant rat uterus. Mol Cell Biochem 1992;114(1-

2):79–89.

[64] Young RC, Smith LH, McLaren MD. T-type and l-type calcium currents in

freshly dispersed human uterine smooth muscle cells. Am J Obstet Gynecol

1993;169:785–792.

[65] Erulkar SD, Ludmir J, Ger B, Nori RD. Expression of different potassium

channels in cells isolated from human myometrium and leiomyomas. Am J

Obstet Gynecol 1993;168(5):1628–39.

[66] Jmari K, Mironneau C, Mironneau J. Inactivation of calcium channel

current in rat uterine smooth muscle: evidence for calcium- and voltage-

mediated mechanisms. J Physiol 1986;380:111–126.

[67] Amedee T, Mironneau C, Mironneau J. The calcium channel current of

pregnant rat single myometrial cells, in short-term primary culture. J Phys-

iol 1987;392:253–272.

[68] Yamamoto T. Effects of estrogens on Ca channels in myometrial cells iso-

lated from pregnant rats. Am J Physiol 1995;268(1 Pt 1):C64–9.

[69] Shmigol AV, Eisner DA, Wray S. Properties of voltage-activated [Ca2+]i

transients in single smooth muscle cells isolated from pregnant rat uterus.

J Physiol 1998;511 ( Pt 3):803–11.

[70] McDonald TF, Pelzer S, Trautwein W, Pelzer DJ. Regulation and mod-

ulation of calcium channels in cardiac, skeletal, and smooth muscle cells.

Physiol Rev Apr 1994;74(2):365–507.

[71] Okabe K, Inoue Y, Soeda H. Estradiol inhibits Ca2+ and K+ channels

in smooth muscle cells from pregnant rat myometrium. Eur J Pharmacol

1999;376(1-2):101–8.

[72] Ohya Y, Sperelakis N. Fast Na+ and slow Ca2+ channels in single uterine

muscle cells from pregnant rats. Am J Physiol 1989;257(2 Pt 1):C408–12.

[73] Parkington HC, Coleman HA. Excitability in uterine smooth muscle. Front

Horm Res 2001;27:179–200.

200

[74] Ohkubo T, Inoue Y, Kawarabayashi T, Kitamura K. Identification and

electrophysiological characteristics of isoforms of T-type calcium channel

Ca(v)3.2 expressed in pregnant human uterus. Cell Physiol Biochem 2005;

16(4-6):245–54.

[75] Asokan KT, Sarkar SN, Mishra SK, Raviprakash V. Effects of mibefradil

on uterine contractility. Eur J Pharmacol Nov 2002;455(1):65–71.

[76] Mishra SK, Hermsmeyer K. Selective inhibition of t-type ca2+ channels by

ro 40-5967. Circ Res Jul 1994;75(1):144–148.

[77] de Paoli MM, Manongi R, Klepp KI. Counsellors’ perspectives on antenatal

hiv testing and infant feeding dilemmas facing women with hiv in northern

tanzania. Reprod Health Matters Nov 2002;10(20):144–156.

[78] Inoue Y, Nakao K, Okabe K, Izumi H, Kanda S, Kitamura K, Kuriyama H.

Some electrical properties of human pregnant myometrium. Am J Obstet

Gynecol 1990;162(4):1090–8.

[79] Serrano JR, Perez-Reyes E, Jones SW. State-dependent inactivation of the

alpha1g t-type calcium channel. J Gen Physiol 1999;114:185–201.

[80] Hering J, Feltz A, Lambert RC. Slow inactivation of the Cav3.1 isotype of

T-type calcium channels. J Physiol 2003;555:331–344.

[81] Knock GA, Aaronson PI. Calcium antagonistic properties of the

cyclooxygenase-2 inhibitor nimesulide in human myometrial myocytes. Br

J Pharmacol 1999;127:1470–1478.

[82] Blanks AM, Zhao ZH, Shmygol A, Bru-Mercier G, Astle S, Thornton S.

Characterization of the molecular and electrophysiological properties of the

t-type calcium channel in human myometrium. J Physiol 2007;581:915–926.

[83] Amde T, Renaud JF, Jmari K, Lombet A, Mironneau J, Lazdunski M. The

presence of na+ channels in myometrial smooth muscle cells is revealed by

specific neurotoxins. Biochem Biophys Res Commun Jun 1986;137(2):675–

681.

[84] Young RC, Herndon-Smith L. Characterization of sodium channels in cul-

tured human uterine smooth muscle cells. Am J Obstet Gynecol Jan 1991;

164(1 Pt 1):175–181.

201

[85] Sperelakis N, Inoue Y, Ohya Y. Fast Na+ channels in smooth muscle from

pregnant rat uterus. Can J Physiol Pharmacol 1992;70(4):491–500.

[86] Greenwood IA, Leblanc N. Overlapping pharmacology of Ca2+-activated

Cl- and K+ channels. Trends Pharmacol Sci 2007;28(1):1–5.

[87] Putney JW. A model for receptor-regulated calcium entry. Cell Calcium

Feb 1986;7(1):1–12.

[88] Dalrymple A, Slater DM, Beech D, Poston L, Tribe RM. Molecular iden-

tification and localization of trp homologues, putative calcium channels, in

pregnant human uterus. Mol Hum Reprod Oct 2002;8(10):946–951.

[89] Yang M, Gupta A, Shlykov SG, Corrigan R, Tsujimoto S, Sanborn BM.

Multiple trp isoforms implicated in capacitative calcium entry are expressed

in human pregnant myometrium and myometrial cells. Biol Reprod Sep

2002;67(3):988–994.

[90] Beech DJ, Muraki K, Flemming R. Non-selective cationic channels of

smooth muscle and the mammalian homologues of drosophila trp. J Physiol

Sep 2004;559(Pt 3):685–706.

[91] Beech DJ. Trpc1: store-operated channel and more. Pflugers Arch Oct

2005;451(1):53–60.

[92] Babich LG, Ku CY, Young HWJ, Huang H, Blackburn MR, Sanborn BM.

Expression of capacitative calcium trpc proteins in rat myometrium during

pregnancy. Biol Reprod Apr 2004;70(4):919–924.

[93] Shimamura K, Kusaka M, Sperelakis N. Oxytocin induces an inward cur-

rent in pregnant rat myometrial cells. Can J Physiol Pharmacol Jul 1994;

72(7):759–763.

[94] Albert AP, Large WA. Store-operated ca2+-permeable non-selective cation

channels in smooth muscle cells. Cell Calcium 2003;33(5-6):345–356.

[95] Arnon A, Hamlyn JM, Blaustein MP. Na(+) entry via store-operated chan-

nels modulates ca(2+) signaling in arterial myocytes. Am J Physiol Cell

Physiol Jan 2000;278(1):C163–C173.

202

[96] Piedras-Renteria E, Stefani E, Toro L. Potassium currents in freshly dis-

persed myometrial cells. Am J Physiol 1991;261:C278–84.

[97] Khan RN, Smith SK, Ashford ML. Contribution of calcium-sensitive

potassium channels to NS1619-induced relaxation in human pregnant my-

ometrium. Hum Reprod 1998;13(1):208–213.

[98] Amberg GC, Baker SA, Koh SD, Hatton WJ, Murray KJ, Horowitz B,

Sanders KM. Characterization of the A-type potassium current in murine

gastric antrum. J Physiol 2002;544(Pt 2):417–28.

[99] Amberg GC, Koh SD, Imaizumi Y, Ohya S, Sanders KM. A-type potassium

currents in smooth muscle. Am J Physiol Cell Physiol 2003;284(3):C583–95.

[100] Anwer K, Toro L, Oberti C, Stefani E, Sanborn BM. Ca(2+)-activated K+

channels in pregnant rat myometrium: modulation by a beta-adrenergic

agent. Am J Physiol 1992;263(5 Pt 1):C1049–56.

[101] Zhou XB, Wang GX, Ruth P, Hneke B, Korth M. Bk(ca) channel activation

by membrane-associated cgmp kinase may contribute to uterine quiescence

in pregnancy. Am J Physiol Cell Physiol Dec 2000;279(6):C1751–C1759.

[102] Prez GJ, Toro L, Erulkar SD, Stefani E. Characterization of large-

conductance, calcium-activated potassium channels from human my-

ometrium. Am J Obstet Gynecol Feb 1993;168(2):652–660.

[103] Brainard AM, Miller AJ, Martens JR, England SK. Maxi-K channels local-

ize to caveolae in human myometrium: a role for an actin-channel-caveolin

complex in the regulation of myometrial smooth muscle K+ current. Am J

Physiol Cell Physiol 2005;289(1):C49–57.

[104] Shmygol A, Noble K, Wray S. Depletion of membrane cholesterol eliminates

the ca2+-activated component of outward potassium current and decreases

membrane capacitance in rat uterine myocytes. J Physiol Jun 2007;581(Pt

2):445–456.

[105] Flandroy L, Galand P. Oestrogen-induced increase in uterine cgmp con-

tent in vitro: effects of inhibitors of protein and rna synthesis. Mol Cell

Endocrinol Jan 1982;25(1):49–54.

203

[106] Izumi H, Garfield RE. Relaxant effects of nitric oxide and cyclic gmp on

pregnant rat uterine longitudinal smooth muscle. Eur J Obstet Gynecol

Reprod Biol Jun 1995;60(2):171–180.

[107] Mistry DK, Garland CJ. Nitric oxide (no)-induced activation of large con-

ductance ca2+-dependent k+ channels (bk(ca)) in smooth muscle cells iso-

lated from the rat mesenteric artery. Br J Pharmacol Jul 1998;124(6):1131–

1140.

[108] Inoue Y, Okabe K, Soeda H. Augmentation and suppression of action

potentials by estradiol in the myometrium of pregnant rat. Can J Physiol

Pharmacol 1999;77(6):447–53.

[109] Mironneau J. Ion channels and the control of uterine contractility. 1994.

[110] Manabe K, Takano M, Noma A. Non-selective cation current of guinea-pig

endocardial endothelial cells. J Physiol Sep 1995;487 ( Pt 2):407–419.

[111] Coleman HA, Parkington HC. Single channel cl- and k+ currents from cells

of uterus not treated with enzymes. Pflugers Arch Nov 1987;410(4-5):560–

562.

[112] Luo CH, Rudy Y. A dynamic model of the cardiac ventricular action po-

tential. II. Afterdepolarizations, triggered activity, and potentiation. Circ

Res 1994;74(6):1097–113.

[113] Hartzell C, Putzier I, Arreola J. Calcium-activated chloride channels. Annu

Rev Physiol 2005;67:719–58.

[114] Grover AK, Kwan CY, Daniel EE. Na–ca exchange in rat myometrium

membrane vesicles highly enriched in plasma membranes. Am J Physiol

1981;240:C175–82.

[115] Grover AK, Kwan CY, Rangachari PK, Daniel EE. Na-ca exchange in a

smooth muscle plasma membrane-enriched fraction. Am J Physiol 1983;

244:C158–65.

[116] Grover AK, Kwan CY. High affinity ca-binding to smooth muscle plasma

membrane: Inhibition by cations. Cell Calcium 1986;7:187–192.

204

[117] Taylor GS, Paton DM, Daniel EE. Evidence for an electrogenic sodium

pump in smooth muscle. Life Sci 1969;8:769–773.

[118] Taylor GS, Paton DM, Daniel EE. Characteristics of electrogenic sodium

pumping in rat myometrium. J Gen Physiol 1970;56:360–375.

[119] Turi A, Marcsek Z, Mullner N, Kucsera M, Bori Z. The activity

of na+/k(+)-atpase and abundance of its mrna are regulated in rat

myometrium during pregnancy. Biochem Biophys Res Commun 1992;

188:1191–1198.

[120] Nakao M, Gadsby DC. [na] and [k] dependence of the na/k pump current-

voltage relationship in guinea pig ventricular myocytes. J Gen Physiol 1989;

94:539–565.

[121] Nakamura Y, Ohya Y, Abe I, Fujishima M. Sodium-potassium pump cur-

rent in smooth muscle cells from mesenteric resistance arteries of the guinea-

pig. J Physiol 1999;519:203–212.

[122] Taggart MJ, Wray S. Agonist mobilization of sarcoplasmic reticular calcium

in smooth muscle: functional coupling to the plasmalemmal na+/ca2+

exchanger? Cell Calcium Nov 1997;22(5):333–341.

[123] Shmigol A, Eisner DA, Wray S. Carboxyeosin decreases the rate of decay of

the [Ca2+]i transient in uterine smooth muscle cells isolated from pregnant

rats. Pflugers Arch 1998;437(1):158–60.

[124] Abe Y. Effects of changing the ionic environment on passive and ac-

tive membrane properties of pregnant rat uterus. J Physiol Apr 1971;

214(1):173–190.

[125] Weber CR, Ginsburg KS, Philipson KD, Shannon TR, Bers DM. Allosteric

regulation of Na/Ca exchange current by cytosolic ca in intact cardiac my-

ocytes. J Gen Physiol 2001;117:119–131.

[126] Hund TJ, Rudy Y. Rate dependence and regulation of action potential and

calcium transient in a canine cardiac ventricular cell model. Circulation

2004;110(20):3168–3174.

205

[127] Johns A, Cutshaw SV. Furosemide-sensitive thallium fluxes in smooth mus-

cle of rabbit uterus. Am J Physiol Dec 1983;245(6):F778–F783.

[128] Pace AJ, Lee E, Athirakul K, Coffman TM, O’Brien DA, Koller BH. Fail-

ure of spermatogenesis in mouse lines deficient in the na(+)-k(+)-2cl(-)

cotransporter. J Clin Invest Feb 2000;105(4):441–450.

[129] Russell DW. Oxysterol biosynthetic enzymes. Biochim Biophys Acta Dec

2000;1529(1-3):126–135.

[130] Parkington HC, Tonta MA, Davies NK, Brennecke SP, Coleman HA. Hy-

perpolarization and slowing of the rate of contraction in human uterus in

pregnancy by prostaglandins e2 and f2alpha: involvement of the na+ pump.

J Physiol Jan 1999;514 ( Pt 1):229–243.

[131] Strieter J, Stephenson JL, Palmer LG, Weinstein AM. Volume-activated

chloride permeability can mediate cell volume regulation in a mathematical

model of a tight epithelium. J Gen Physiol Aug 1990;96(2):319–344.

[132] Volpe P, Martini A, Furlan S, Meldolesi J. Calsequestrin is a component of

smooth muscles: the skeletal- and cardiac-muscle isoforms are both present,

although in highly variable amounts and ratios. Biochem J Jul 1994;301 (

Pt 2):465–469.

[133] Milner RE, Baksh S, Shemanko C, Carpenter MR, Smillie L, Vance JE,

Opas M, Michalak M. Calreticulin, and not calsequestrin, is the major

calcium binding protein of smooth muscle sarcoplasmic reticulum and liver

endoplasmic reticulum. J Biol Chem Apr 1991;266(11):7155–7165.

[134] Jafri MS, Rice JJ, Winslow RL. Cardiac ca2+ dynamics: the roles of

ryanodine receptor adaptation and sarcoplasmic reticulum load. Biophys J

Mar 1998;74(3):1149–1168.

[135] Taggart MJ, Wray S. Contribution of sarcoplasmic reticular calcium to

smooth muscle contractile activation: gestational dependence in isolated

rat uterus. J Physiol Aug 1998;511 ( Pt 1):133–144.

[136] Kupittayanant S, Luckas MJM, Wray S. Effect of inhibiting the sarcoplas-

mic reticulum on spontaneous and oxytocin-induced contractions of human

myometrium. BJOG Mar 2002;109(3):289–296.

206

[137] Noble K, Wray S. The role of the sarcoplasmic reticulum in neonatal uterine

smooth muscle: enhanced role compared to adult rat. J Physiol Dec 2002;

545(Pt 2):557–566.

[138] Peterson JW. Simple model of smooth muscle myosin phosphorylation

and dephosphorylation as rate-limiting mechanism. Biophys J Feb 1982;

37(2):453–459.

[139] Kato S, Osa T, Ogasawara T. Kinetic model for isometric contraction in

smooth muscle on the basis of myosin phosphorylation hypothesis. Biophys

J Jul 1984;46(1):35–44.

[140] Murphy RA, Rembold CM, Hai CM. Contraction in smooth muscle: what

is latch? Prog Clin Biol Res 1990;327:39–50.

[141] Murphy RA, Rembold CM. The latch-bridge hypothesis of smooth muscle

contraction. Can J Physiol Pharmacol Oct 2005;83(10):857–864.

[142] Hai CM, Kim HR. An expanded latch-bridge model of protein kinase c-

mediated smooth muscle contraction. J Appl Physiol Apr 2005;98(4):1356–

1365.

[143] Hai CM, Rembold CM, Murphy RA. Can different four-state crossbridge

models explain latch and the energetics of vascular smooth muscle? Adv

Exp Med Biol 1991;304:159–170.

[144] Rembold CM, Murphy RA. Models of the mechanism for crossbridge at-

tachment in smooth muscle. J Muscle Res Cell Motil Jun 1993;14(3):325–

334.

[145] Bennett MR, Farnell L, Gibson WG. A quantitative description of the con-

traction of blood vessels following the release of noradrenaline from sympa-

thetic varicosities. J Theor Biol May 2005;234(1):107–122.

[146] Fajmut A, Brumen M. Mlc-kinase/phosphatase control of ca2+ signal

transduction in airway smooth muscles. J Theor Biol Jun 2008;252(3):474–

481.

[147] Parthimos D, Edwards DH, Griffith TM. Minimal model of arterial chaos

generated by coupled intracellular and membrane ca2+ oscillators. Am J

Physiol Sep 1999;277(3 Pt 2):H1119–H1144.

207

[148] Gasser H, Hill A. The dynamics of muscular contraction. Proceedings of

the Royal Society of London Series B Containing Papers of a Biological

Character 1924;96(678):398–437.

[149] Carlson BE, Secomb TW. A theoretical model for the myogenic response

based on the length-tension characteristics of vascular smooth muscle. Mi-

crocirculation Jun 2005;12(4):327–338.

[150] Kroon M. A constitutive model for smooth muscle including active tone

and passive viscoelastic behaviour. Math Med Biol Jul 2009;.

[151] Stlhand J, Klarbring A, Holzapfel GA. Smooth muscle contraction:

mechanochemical formulation for homogeneous finite strains. Prog Biophys

Mol Biol 2008;96(1-3):465–481.

[152] Warshaw DM, Fay FS. Cross-bridge elasticity in single smooth muscle cells.

J Gen Physiol Aug 1983;82(2):157–199.

[153] Warshaw DM. Force: velocity relationship in single isolated toad stomach

smooth muscle cells. J Gen Physiol May 1987;89(5):771–789.

[154] Warshaw DM, Rees DD, Fay FS. Characterization of cross-bridge elastic-

ity and kinetics of cross-bridge cycling during force development in single

smooth muscle cells. J Gen Physiol Jun 1988;91(6):761–779.

[155] Holmes JW. Teaching from classic papers: Hill’s model of muscle contrac-

tion. Adv Physiol Educ Jun 2006;30(2):67–72.

[156] Mollard P, Mironneau J, Amedee T, Mironneau C. Electrophysiological

characterization of single pregnant rat myometrial cells in short-term pri-

mary culture. Am J Physiol 1986;250(1 Pt 1):C47–54.

[157] Loewenstein WR. Junctional intercellular communication: the cell-to-cell

membrane channel. Physiol Rev Oct 1981;61(4):829–913.

[158] Okawa H. Electrical and mechanical interaction between the muscle layers

of rat uterus and different sensitivities to oxytocin. Bull Yamaguchi Med

School 1975;22:197–210.

[159] M.A. S, L. G, Kase N. Clinical gynecologic endocrinology and infertility.

5th edition. Williams and Wilkins, Baltimore, MA., 1994.

208

[160] Osa T, Ogasawara T. Effects in vitro of progesterone and estradiol-17b on

the contractileand electrical responses in rat myometrium. Jpn J Physiol

1984;34:427–411.

[161] Burdyga T, Borisova L, Burdyga A, Wray S. Temporal and spatial vari-

ations in spontaneous ca events and mechanical activity in pregnant rat

myometrium. European Journal of Obstetrics Gynecology and Reproduc-

tive Biology 2009;144:S25–S32.

[162] Choi HS, A. ED. The role of sarcolemmal ca2+-atpase in the regulation of

resting calcium concentration in rat ventricular myocytes. J Physiol 1998;

515:109–119.

[163] Gatto C, Hale CC, Xu W, Milanick MA. Eosin, a potent inhibitor of the

plasma membrane ca pump, does not inhibit the cardiac na-ca exchanger.

Biochemistry Jan 1995;34(3):965–972.

[164] Bassani RA, Bassani JW, Bers DM. Relaxation in ferret ventricular my-

ocytes: unusual interplay among calcium transport systems. J Physiol Apr

1994;476(2):295–308.

[165] King JF, Flenady V, Papatsonis D, Dekker G, Carbonne B. Calcium chan-

nel blockers for inhibiting preterm labour; a systematic review of the evi-

dence and a protocol for administration of nifedipine. Aust N Z J Obstet

Gynaecol Jun 2003;43(3):192–198.

[166] Maigaard S, Forman A, Brogaard-Hansen KP, Andersson KE. Inhibitory

effects of nitrendipine on myometrial and vascular smooth muscle in human

pregnant uterus and placenta. Acta Pharmacol Toxicol Copenh Jul 1986;

59(1):1–10.

[167] Balke CW, Goldman L. Excitation contraction coupling in cardiac muscle:

is there a purely voltage-dependent component? J Gen Physiol May 2003;

121(5):349–352.

[168] Iino M, Kobayashi T, Endo M. Use of ryanodine for functional removal

of the calcium store in smooth muscle cells of the guinea-pig. Biochem

Biophys Res Commun Apr 1988;152(1):417–422.

209

[169] Gilchrist JS, Belcastro AN, Katz S. Intraluminal ca2+ dependence of ca2+

and ryanodine-mediated regulation of skeletal muscle sarcoplasmic reticu-

lum ca2+ release. J Biol Chem Oct 1992;267(29):20850–20856.

[170] Young RC, Zhang P. The mechanism of propagation of intracellular calcium

waves in cultured human uterine myocytes. Am J Obstet Gynecol May

2001;184(6):1228–1234.

[171] Burghardt RC, Barhoumi R, Sanborn BM, Andersen J. Oxytocin-induced

ca2+ responses in human myometrial cells. Biol Reprod Apr 1999;

60(4):777–782.

[172] Westerblad H, Allen DG. Intracellular calibration of the calcium indicator

indo-1 in isolated fibers of xenopus muscle. Biophys J Aug 1996;71(2):908–

917.

[173] Mitsui M, Karaki H. Dual effects of carbachol on cytosolic ca2+ and con-

traction in intestinal smooth muscle. Am J Physiol May 1990;258(5 Pt

1):C787–C793.

[174] Kitazawa T, Hirama R, Masunaga K, Nakamura T, Asakawa K, Cao

J, Teraoka H, Unno T, ichi Komori S, Yamada M, Wess J, Taneike T.

Muscarinic receptor subtypes involved in carbachol-induced contraction of

mouse uterine smooth muscle. Naunyn Schmiedebergs Arch Pharmacol Jun

2008;377(4-6):503–513.

[175] Earley L, Wray S. Effects of hypoxia on force produced by agonists and

depolarization and arising spontaneously in the rat uterus. J Reprod Fertil

Nov 1993;99(2):539–544.

[176] Bae J, Peters-Golden M, Loch-Caruso R. Stimulation of pregnant rat uter-

ine contraction by the polychlorinated biphenyl (pcb) mixture aroclor 1242

may be mediated by arachidonic acid release through activation of phos-

pholipase a2 enzymes. J Pharmacol Exp Ther May 1999;289(2):1112–1120.

[177] Shmigol AV, Eisner DA, Wray S. Simultaneous measurements of changes

in sarcoplasmic reticulum and cytosolic. J Physiol Mar 2001;531(Pt 3):707–

713.

210

[178] Zraly Z, Kummer V, Veznik Z. Use of carbachol for dilatation of the cervix

in heifers. Theriogenology 1980;13(3):217–220.

[179] Shmygol A, Wray S. Modulation of agonist-induced ca2+ release by sr ca2+

load: direct sr and cytosolic ca2+ measurements in rat uterine myocytes.

Cell Calcium Mar 2005;37(3):215–223.

[180] Holda JR, Klishin A, Sedova M, Hser J, Blatter LA. Capacitative calcium

entry. News Physiol Sci Aug 1998;13:157–163.

[181] Putney Jr J, McKay R. Capacitative calcium entry channels. Bioessays

1999;21(1):38–46.

[182] Embrey MP. Letter: prostaglandin-induced abortion and cervical incom-

petence. Br Med J May 1975;2(5969):497.

[183] Keirse MJ. Therapeutic uses of prostaglandins. Baillieres Clin Obstet

Gynaecol Dec 1992;6(4):787–808.

[184] Longbottom ER, Luckas MJ, Kupittayanant S, Badrick E, Shmigol T, Wray

S. The effects of inhibiting myosin light chain kinase on contraction and

calcium signalling in human and rat myometrium. Pflugers Arch Jun 2000;

440(2):315–321.

[185] Sanborn BM, Rao BR, Korenman SG. Interaction of 17 -estradiol and

its specific uterine receptor. evidence for complex kinetic and equilibrium

behavior. Biochemistry Dec 1971;10(26):4955–4962.

[186] Puri CP, Garfield RE. Changes in hormone levels and gap junctions in

the rat uterus during pregnancy and parturition. Biol Reprod Nov 1982;

27(4):967–975.

[187] Sanborn BM, Dodge K, Monga M, Qian A, Wang W, Yue C. Molecular

mechanisms regulating the effects of oxytocin on myometrial intracellular

calcium. Adv Exp Med Biol 1998;449:277–286.

[188] Sims SM, Daniel EE, Garfield RE. Improved electrical coupling in uterine

smooth muscle is associated with increased numbers of gap junctions at

parturition. J Gen Physiol Sep 1982;80(3):353–375.

211

[189] CASTEELS R, KURIYAMA H. Membrane potential and ionic content in

pregnant and non-pregnant rat myometrium. J Physiol Mar 1965;177:263–

287.

[190] Garfield RE. Classic illustration from gap junctions: their presence and

necessity in myometrium during parturition. by r. e. garfield, et al, science

1977. Eur J Obstet Gynecol Reprod Biol Oct 1988;29(2):179–180.

[191] Word RA, Stull JT, Casey ML, Kamm KE. Contractile elements and myosin

light chain phosphorylation in myometrial tissue from nonpregnant and

pregnant women. J Clin Invest Jul 1993;92(1):29–37.

[192] Shynlova O, Kwong R, Lye SJ. Mechanical stretch regulates hypertrophic

phenotype of the myometrium during pregnancy. Reproduction Jan 2010;

139(1):247–253.

[193] Celik H, Ayar A, Sapmaz E. Effects of erythromycin on stretch-induced

contractile activity of isolated myometrium from pregnant women. Acta

Obstet Gynecol Scand Aug 2001;80(8):697–701.

[194] Shynlova OP, Oldenhof AD, Liu M, Langille L, Lye SJ. Regulation of c-fos

expression by static stretch in rat myometrial smooth muscle cells. Am J

Obstet Gynecol Jun 2002;186(6):1358–1365.

[195] Oldenhof AD, Shynlova OP, Liu M, Langille BL, Lye SJ. Mitogen-

activated protein kinases mediate stretch-induced c-fos mrna expression

in myometrial smooth muscle cells. Am J Physiol Cell Physiol Nov 2002;

283(5):C1530–C1539.

[196] Dalrymple A, Mahn K, Poston L, Songu-Mize E, Tribe RM. Mechanical

stretch regulates trpc expression and calcium entry in human myometrial

smooth muscle cells. Mol Hum Reprod Mar 2007;13(3):171–179.

[197] Rendt JM, Toro L, Stefani E, Erulkar SD. Progesterone increases ca2+

currents in myometrial cells from immature and nonpregnant adult rats.

Am J Physiol Feb 1992;262(2 Pt 1):C293–C301.

[198] Tezuka N, Ali M, Chwalisz K, Garfield RE. Changes in transcripts encod-

ing calcium channel subunits of rat myometrium during pregnancy. Am J

Physiol Oct 1995;269(4 Pt 1):C1008–C1017.

212

[199] Collins PL, Moore JJ, Lundgren DW, Choobineh E, Chang SM, Chang

AS. Gestational changes in uterine l-type calcium channel function and

expression in guinea pig. Biol Reprod Nov 2000;63(5):1262–1270.

[200] Mershon JL, Mikala G, Schwartz A. Changes in the expression of the l-type

voltage-dependent calcium channel during pregnancy and parturition in the

rat. Biol Reprod Nov 1994;51(5):993–999.

[201] Byron KL, Brueggemann LI. Labour pains: giving birth to new mechanisms

for the regulation of myometrial contractility. J Physiol May 2009;587(Pt

10):2109–2110.

[202] Greenwood IA, Yeung SY, Tribe RM, Ohya S. Loss of functional k+ chan-

nels encoded by ether--go-go-related genes in mouse myometrium prior to

labour onset. J Physiol May 2009;587(Pt 10):2313–2326.

[203] Smith RC, McClure MC, Smith MA, Abel PW, Bradley ME. The role of

voltage-gated potassium channels in the regulation of mouse uterine con-

tractility. Reprod Biol Endocrinol 2007;5:41.

[204] Song M, Helguera G, Eghbali M, Zhu N, Zarei MM, Olcese R, Toro L,

Stefani E. Remodeling of kv4.3 potassium channel gene expression under

the control of sex hormones. J Biol Chem Aug 2001;276(34):31883–31890.

[205] Lukas TJ, Haiech J, Lau W, Craig TA, Zimmer WE, Shattuck RL, Shoe-

maker MO, Watterson DM. Calmodulin and calmodulin-regulated protein

kinases as transducers of intracellular calcium signals. Cold Spring Harb

Symp Quant Biol 1988;53 Pt 1:185–193.

[206] Moore F, Bernal AL. Myosin light chain kinase and the onset of labour in

humans. Exp Physiol Mar 2001;86(2):313–318.

[207] Szal SE, Repke JT, Seely EW, Graves SW, Parker CA, Morgan KG. [ca2+]i

signaling in pregnant human myometrium. Am J Physiol Jul 1994;267(1

Pt 1):E77–E87.

[208] Weber LP, Lierop JEV, Walsh MP. Ca2+-independent phosphorylation of

myosin in rat caudal artery and chicken gizzard myofilaments. J Physiol

May 1999;516 ( Pt 3):805–824.

213

[209] Horowitz A, Clment-Chomienne O, Walsh MP, Morgan KG. Epsilon-

isoenzyme of protein kinase c induces a ca(2+)-independent contraction in

vascular smooth muscle. Am J Physiol Aug 1996;271(2 Pt 1):C589–C594.

[210] Garfield RE, Ali M, Yallampalli C, Izumi H. Role of gap junctions and

nitric oxide in control of myometrial contractility. Semin Perinatol Feb

1995;19(1):41–51.

[211] Cole WC, Garfield RE, Kirkaldy JS. Gap junctions and direct intercellular

communication between rat uterine smooth muscle cells. Am J Physiol Jul

1985;249(1 Pt 1):C20–C31.

[212] Cole WC, Garfield RE. Evidence for physiological regulation of myometrial

gap junction permeability. Am J Physiol Sep 1986;251(3 Pt 1):C411–C420.

[213] Miller SM, Garfield RE, Daniel EE. Improved propagation in myometrium

associated with gap junctions during parturition. Am J Physiol Jan 1989;

256(1 Pt 1):C130–C141.

[214] Sakai N, Tabb T, Garfield RE. Modulation of cell-to-cell coupling between

myometrial cells of the human uterus during pregnancy. Am J Obstet

Gynecol Aug 1992;167(2):472–480.

[215] Benson AP, Clayton RH, Holden AV, Kharche S, Tong WC. Endogenous

driving and synchronization in cardiac and uterine virtual tissues: bifurca-

tions and local coupling. Philos Transact A Math Phys Eng Sci May 2006;

364(1842):1313–1327.

[216] Fitzhugh R. Impulses and physiological states in theoretical models of nerve

membrane. Biophys J Jul 1961;1(6):445–466.

[217] Nagumo J, Arimoto S, Yoshizawa S. An active pulse transmission line

simulating nerve axon. Proceedings of the IRE 1962;50(10):2061–2070.

[218] Rihana S, Lefranois E, Marque C. A two dimension model of the uterine

electrical wave propagation. Conf Proc IEEE Eng Med Biol Soc 2007;

2007:1109–1112.

[219] Rihana S, Marque C. Preterm labor–modeling the uterine electrical activity

from cellular level to surface recording. Conf Proc IEEE Eng Med Biol Soc

2008;2008:3726–3729.

214

[220] Kuriyama H, Suzuki H. Changes in electrical properties of rat myometrium

during gestation and following hormonal treatments. J Physiol 1976;

260:315–333.

[221] Wesselink WA, Holsheimer J, Boom HB. A model of the electrical behaviour

of myelinated sensory nerve fibres based on human data. Med Biol Eng

Comput Mar 1999;37(2):228–235.

[222] FitzHugh R. Computation of impulse initiation and saltatory conduction

in a myelinated nerve fiber. Biophys J Jan 1962;2:11–21.

[223] Mori Y, Fishman GI, Peskin CS. Ephaptic conduction in a cardiac strand

model with 3d electrodiffusion. Proc Natl Acad Sci U S A Apr 2008;

105(17):6463–6468.

[224] Aslanidi OV, Stewart P, Boyett MR, Zhang H. Optimal velocity and safety

of discontinuous conduction through the heterogeneous purkinje-ventricular

junction. Biophys J Jul 2009;97(1):20–39.

[225] Stewart P, Aslanidi OV, Noble D, Noble PJ, Boyett MR, Zhang H. Math-

ematical models of the electrical action potential of purkinje fibre cells.

Philos Transact A Math Phys Eng Sci Jun 2009;367(1896):2225–2255.

[226] Cabo C, Barr RC. Reflection after delayed excitation in a computer model

of a single fiber. Circ Res Aug 1992;71(2):260–270.

[227] Kurjak A, Chervenak F. Textbook of perinatal medicine. Informa Health-

Care, 2006.

[228] Specht PC. Phase-plane analysis of action potentials in uterine smooth

muscle. Pflugers Arch Nov 1976;367(1):89–95.

[229] Wolfs GM, van Leeuwen M. Electromyographic observations on the human

uterus during labour. Acta Obstet Gynecol Scand Suppl 1979;90:1–61.

[230] Smith R. The endocrinology of parturition: Basic science and clinical ap-

plication. S Karger Pub, 2001.

[231] Wolfs G, Rottinghuis H. Electrical and mechanical activity of the human

uterus during labour. Arch Gynakol Apr 1970;208(4):373–385.

215

[232] Rihana S, Terrien J, Germain G, Marque C. Mathematical modeling of

electrical activity of uterine muscle cells. Med Biol Eng Comput Jun 2009;

47(6):665–675.

[233] Allen TJ. Temperature dependence of macroscopic L-type calcium channel

currents, in single guinea pig ventricular myocytes. J Cardiovasc Electro-

physiol 1996;7(4):307–321.

[234] Shmygol A, Wray S. Functional architecture of the sr calcium store in

uterine smooth muscle. Cell Calcium Jun 2004;35(6):501–508.

[235] Lindblad DS, Murphey CR, Clark JW, Giles WR. A model of the action

potential and underlying membrane currents in a rabbit atrial cell. Am J

Physiol Oct 1996;271(4 Pt 2):H1666–H1696.

[236] Wong AY, Klassen GA. A model of calcium regulation in smooth muscle

cell. Cell Calcium Mar 1993;14(3):227–243.

[237] Wong AY, Klassen GA. Endothelin-induced electrical activity and calcium

dynamics in vascular smooth muscle cells: a model study. Ann Biomed Eng

1996;24(5):547–560.

[238] Anderson NC, Ramon F, Snyder A. Studies on calcium and sodium in

uterine smooth muscle excitation under current-clamp and voltage-clamp

conditions. J Gen Physiol Sep 1971;58(3):322–339.

[239] Thomas RC. Electrogenic sodium pump in nerve and muscle cells. Physiol

Rev Jul 1972;52(3):563–594.

[240] Daniel E, Sehdev H, Robinson K. Mechanisms for activation of smooth

muscle. Physiol Rev 1962;42(Suppl 5):228–260.

[241] Casteels R. The action of ouabain on the smooth muscle cells of the guinea-

pig’s taenia coli. J Physiol May 1966;184(1):131–142.

[242] Matthews E, Sutter M. Ouabain-induced changes in the contractile and

electrical activity, potassium content, and response to drugs, of smooth

muscle cells. Canadian journal of physiology and pharmacology 1967;

45(3):509.

216

[243] Vassort G. Voltage-clamp analysis of transmembrane ionic currents in

guinea-pig myometrium: evidence for an initial potassium activation trig-

gered by calcium influx. J Physiol Nov 1975;252(3):713–734.

[244] Miyoshi H, Urabe S, Shimazutsu R, Samura O, Fujiwara H, Kudo Y. Mag-

nesium sulphate suppresses atp receptors in myometrial cells to inhibit uter-

ine contractility for tocolysis. American Journal of Obstetrics and Gyne-

cology 2006;195(6):S96.

[245] Kosterin SA, Burdyga TV, Fomin VP, Grover AK. Mechanisms of Ca2+

transport in myometrium In control of uterine contractility. Boca Raton,

FL: CRC press Inc., 1994.

[246] Luckas MJ, Taggart MJ, Wray S. Intracellular calcium stores and agonist-

induced contractions in isolated human myometrium. Am J Obstet Gynecol

Aug 1999;181(2):468–476.

[247] Marshall J. The physiology of the myometrium, In the Uterus. Baltimore:

Williams and Wilkins 89-109, 1973.

[248] MARSHALL JM. Regulation of activity in uterine smooth muscle. Physiol

Rev Suppl Jul 1962;5:213–227.

[249] Sigger JN, Harding R, Jenkin G. Relationship between electrical activ-

ity of the uterus and surgically isolated myometrium in the pregnant and

nonpregnant ewe. J Reprod Fertil Jan 1984;70(1):103–114.

[250] Shintani Y, Hirano K, Nishimura J, Nakano H, Kanaide H. Enhanced

contractile response to thrombin in the pregnant rat myometrium. Br J

Pharmacol Dec 2000;131(8):1619–1628.

[251] Douglas AJ, Clarke EW, Goldspink DF. Influence of mechanical stretch on

growth and protein turnover of rat uterus. Am J Physiol May 1988;254(5

Pt 1):E543–E548.

[252] Riemer RK, Heymann MA. Regulation of uterine smooth muscle function

during gestation. Pediatr Res Nov 1998;44(5):615–627.

[253] Kasai Y, Tsutsumi O, Taketani Y, Endo M, Iino M. Stretch-induced en-

hancement of contractions in uterine smooth muscle of rats. J Physiol Jul

1995;486 ( Pt 2):373–384.

217

[254] Dupont G, Pontes J, Goldbeter A. Modeling spiral ca2+ waves in single

cardiac cells: role of the spatial heterogeneity created by the nucleus. Am

J Physiol Oct 1996;271(4 Pt 1):C1390–C1399.

[255] Tea BS, Dam TV, Moreau P, Hamet P, deBlois D. Apoptosis during regres-

sion of cardiac hypertrophy in spontaneously hypertensive rats. temporal

regulation and spatial heterogeneity. Hypertension Aug 1999;34(2):229–235.

[256] Decking UKM. Spatial heterogeneity in the heart: recent insights and open

questions. News Physiol Sci Dec 2002;17:246–250.

[257] Huo Y, Kaimovitz B, Lanir Y, Wischgoll T, Hoffman JIE, Kassab GS.

Biophysical model of the spatial heterogeneity of myocardial flow. Biophys

J May 2009;96(10):4035–4043.

[258] Toescu EC. Temporal and spatial heterogeneities of ca2+ signaling: mech-

anisms and physiological roles. Am J Physiol Aug 1995;269(2 Pt 1):G173–

G185.

[259] Young RC, Zhang P. Functional separation of deep cytoplasmic calcium

from subplasmalemmal space calcium in cultured human uterine smooth

muscle cells. Cell Calcium Jul 2004;36(1):11–17.

[260] Draeger A, Wray S, Babiychuk EB. Domain architecture of the smooth-

muscle plasma membrane: regulation by annexins. Biochem J Apr 2005;

387(Pt 2):309–314.

[261] Noble K, Zhang J, Wray S. Lipid rafts, the sarcoplasmic reticulum and

uterine calcium signalling: an integrated approach. J Physiol Jan 2006;

570(Pt 1):29–35.

[262] Floyd R, Wray S. Calcium transporters and signalling in smooth muscles.

Cell Calcium 2007;42(4-5):467–476.

[263] Ozaki H, Yasuda K, Kim YS, Egawa M, Kanzaki H, Nakazawa H, Hori M,

Seto M, Karaki H. Possible role of the protein kinase c/cpi-17 pathway

in the augmented contraction of human myometrium after gestation. Br J

Pharmacol Dec 2003;140(7):1303–1312.

218

[264] Sanborn BM, Yue C, Wang W, Dodge KL. G protein signalling pathways

in myometrium: affecting the balance between contraction and relaxation.

Rev Reprod Sep 1998;3(3):196–205.

[265] Breuiller-Fouch M, Tertrin-Clary C, Hluy V, Fournier T, Ferr F. Role of

protein kinase c in endothelin-1-induced contraction of human myometrium.

Biol Reprod Jul 1998;59(1):153–159.

[266] Eude I, Paris B, Cabrol D, Ferr F, Breuiller-Fouch M. Selective protein

kinase c isoforms are involved in endothelin-1-induced human uterine con-

traction at the end of pregnancy. Biol Reprod Nov 2000;63(5):1567–1573.

[267] Liberto GD, Dallot E, Parco IEL, Cabrol D, Ferr F, Breuiller-Fouch M. A

critical role for pkc zeta in endothelin-1-induced uterine contractions at the

end of pregnancy. Am J Physiol Cell Physiol Sep 2003;285(3):C599–C607.

[268] Morrison JJ, Dearn SR, Smith SK, Ahmed A. Activation of protein ki-

nase c is required for oxytocin-induced contractility in human pregnant

myometrium. Hum Reprod Oct 1996;11(10):2285–2290.

[269] Phillippe M. Protein kinase c, an inhibitor of oxytocin-stimulated phasic

myometrial contractions. Biol Reprod Apr 1994;50(4):855–859.

[270] Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes

of preterm birth. Lancet Jan 2008;371(9606):75–84.

[271] Creasy RK. Preventing preterm birth. N Engl J Med Sep 1991;325(10):727–

729.

[272] Simhan HN, Caritis SN. Prevention of preterm delivery. N Engl J Med

Aug 2007;357(5):477–487.

[273] Bernal AL, Europe-Finner GN, Phaneuf S, Watson SP. Preterm labour: a

pharmacological challenge. Trends Pharmacol Sci Apr 1995;16(4):129–133.

219

Appendix A

Single cell uterine model

A.1 Model Equations – Membrane currents and

Sarcoplasmic reticulum control

Hyperpolarisation-activated current – Ih

Ih = gh y(V − Eh ) (2.12)

Eh =RT

Fln

([ K+ ]o + ( PNa / PK ) [ Na+ ]o

[ K+ ]i + ( PNa / PK ) [ Na+ ]i

)(2.13)

y∞ =1

1 + exp

(V + 105.39

8.66

) (2.14)

τy =1

3.5x10−6 exp (−0.0497V) + 0.04 exp (0.0521V)(2.15)

dy

dt=

y∞ − y

τy

(2.16)

220

L-type calcium current – ICaL

ICaL = gCaL d2 fCa (0.8 f1 + 0.2 f2 )(V − ECaL ) (2.17)

fCa =1

1 +[ Ca2+ ]i

Km,CaL

(2.18)

d∞ =1

1 + exp

(− (V + 22)

7

) (2.19)

f∞ =1

1 + exp

(V + 33

7

) (2.20)

τd = 2.29 +5.7

1 +

(V + 29.97

9

)2 (2.21)

τ f1 = 12 ms (2.22)

τ f2 = 90.97− 90.97(

1 + exp

(V + 13.96

45.38

))(1 + exp

(− (V + 9.5)

3.39

)) (2.23)

dd

dt=

d∞ − d

τd

(2.24)

d f1

dt=

f∞ − f1

τ f1

(2.25)

d f2

dt=

f∞ − f2

τ f2

(2.26)

221

T-type calcium current – ICaT

ICaT = gCaT b2g(V − ECaT ) (2.27)

b∞ =1

1 + exp

(− (V + 54.23)

9.88

) (2.28)

g∞ = 0.02 +0.98

1 + exp

(V + 72.98

4.64

) (2.29)

τb = 0.45 +3.9

1 +

(V + 66

26

)2 (2.30)

τg = 150− 150(

1 + exp

(V − 417.43

203.18

))(1 + exp

(− (V + 61.11)

8.07

)) (2.31)

db

dt=

b∞ − b

τb

(2.32)

dg

dt=

g∞ − g

τg

(2.33)

222

Fast sodium current – INa

INa = gNa m3h(V − ENa ) (A.1)

ENa =RT

Fln

([Na]o

[Na]i

)(2.35)

m∞ =1

1 + exp

(− (V + 35.96)

9.24

) (2.36)

h∞ =1

1 + exp

(V + 57

8

) (2.37)

τm = 0.25 +7

1 + exp

(V + 38

10

) (2.38)

τh = 0.9 +1002.85

1 +

(V + 47.5

1.5

)2 (2.39)

dm

dt=

m∞ −m

τm

(2.40)

dh

dt=

h∞ − h

τh

(2.41)

Store-operated non-selective cation current – ISOC

ISOC = ISOCCa + ISOCNa (2.42)

ISOCNa = gSOCNa PSOC (V − ENa ) (2.43)

ISOCCa = gSOCCa PSOC (V − ECa ) (2.44)

PSOC =

(1 +

[ Ca2+ ]uKm,SOC

)−1

(2.45)

223

Delayed rectifying potassium current – IK1

IK1 = gK1 q(0.38 r1 + 0.62 r2 )(V − EK ) (2.46)

EK =RT

Fln

([K]o

[K]i

)(2.47)

q∞ =1

1 + exp

(− (V − 7.7)

23.7

) (2.48)

r∞ =1

1 + exp

(V + 63

6.3

) (2.49)

τq =500

1 +

(V + 60.71

15.79

)2 (2.50)

τ r1 =5× 104

1 +

(V + 62.71

35.86

)2 (2.51)

τ r2 = 3× 104 +2.2× 105

1 + exp

(V + 22

4

) (2.52)

dq

dt=

q∞ − q

τq

(2.53)

d r1

dt=

r∞ − r1

τ r1

(2.54)

d r2

dt=

r∞ − r2

τ r2

(2.55)

224

Delayed rectifying potassium current – IK2

IK2 = gK2 p(0.75 k1 + 0.25 k2 )(V − EK ) (2.56)

EK =RT

Fln

([K]o

[K]i

)

p∞ =1

1 + exp

(− (V − 4.2)

23

) (2.57)

k∞ =1

1 + exp

((V + 21.2)

5.7

) (2.58)

τp =100

1 +

(V + 64.1

28.67

)2 (2.59)

τ k1 = 1× 106 − 1× 106

(1 + exp

(V − 315

50

))(1 + exp

(− (V + 74.9)

8

)) (2.60)

τ k2 = 2.5× 106 − 2.5× 106

(1 + exp

(V − 132.87

25.40

))(1 + exp

(− (V + 24.92)

2.68

))

(2.61)

dp

dt=

p∞ − p

τp

(2.62)

d k1

dt=

k∞ − k1

τ k1

(2.63)

d k2

dt=

k∞ − k2

τ k2

(2.64)

225

A-type transient potassium current – IKA

IKA = gKA s x(V − EK ) (2.65)

EK =RT

Fln

([K]o

[K]i

)

s∞ =1

1 + exp

(− (V + 27.79)

7.57

) (2.66)

x∞ = 0.02 +0.98

1 + exp

(V + 69.5

6

) (2.67)

τ s =17

1 +

(V + 20.52

35

)2 (2.68)

τ x = 7.5 +10

1 +

(V + 34.18

120

)2 (2.69)

d s

dt=

s∞ − s

τ s

(2.70)

d x

dt=

x∞ − x

τ x

(2.71)

226

Calcium-activated potassium current – IKCa

IKCa =Am NBKCa PKCa iKCa

100(2.72)

PKCa = 0.17 pf + 0.83 ps (2.73)

d pf

dt=

po − pf

τpf

(2.74)

d ps

dt=

po − ps

τ ps

(2.75)

po =

(1 + exp

(−V − V1/2KCa

18.25

))−1

(2.76)

V1/2KCa = −41.7log10( [ Ca2+ ]i )− 128.2 (2.77)

iKCa = 104 PBKCa VF2

RT

[K]o − [K]i exp(VF/RT)

1− exp(VF/RT)(2.78)

Background K+ leakage current – IKleak

IKleak = gK−leak (V − EK ) (2.79)

227

Non-selective cation current – INSCC

INSCC = INSCa + INSNa + INSK + INSleak (2.80)

INSCa = 0.15 fMg g( [ Ca2+ ]o ) gNS (V − ENS ) (2.81)

INSNa = fMg g( [ Na+ ]o ) gNS (V − ENS ) (2.82)

INSK = 1.19 fMg g( [ K+ ]o ) gNS (V − ENS ) (2.83)

INSleak = fMg gleak (V − ENS ) (2.84)

g( [X]o ) =1

g s

0.03

1 +

(150

[X]o + 10−8

)2 , g s =

0.000525 for Ca2+

0.0123 otherwise(2.85)

fMg = 0.1 +0.9

1 +

([Mg2+]o

Kd,Mg

)1.3 (2.86)

ENS =RT

F· ln

PNa

PCs

[ Na+ ]o +PK

PCs

[ K+ ]o + PCa′4 PCa

PCs

[ Ca2+ ]o

PNa

PCs

[ Na+ ]i +PK

PCs

[ K+ ]i + PCa′4 PCa

PCs

[ Ca2+ ]i

(2.87)

PCa′ =

PCa

1 + exp

(VF

RT

) (2.88)

228

Calcium-activated chloride current – ICl

ICl =

gCl gfCa

√c1(V − ECl ) if V > 0

gCl gfCa c2 n(V − ECl ) otherwise(2.89)

ECl =RT

F zCl

ln

([Cl]i

[Cl]o

)(2.90)

gCl =

gCl (19.26 + 365.8 exp(0.0564V)) if V < −20.0

gCl (0.752 + 0.0349 exp(−0.414V)) otherwise(2.91)

gfCa =[ Ca2+ ]i

[ Ca2+ ]i + Kd,Cl

(2.92)

c∞ =1

1 + exp

(44.08− V

13.7

) (2.93)

n∞ =1

1 + exp

(V + 56.97

20.29

) (2.94)

τ c1 = 26.14− 16.88

1 + exp

(− (V − 48.14)

14.85

) (2.95)

τ c2 = 50.79 +81.54

1 + exp

(− (V + 43.51)

9.5

) (2.96)

τn = 0.69 ms (2.97)

d c1

dt=

c∞ − c1

τ c1

(2.98)

d c2

dt=

c∞ − c2

τ c2

(2.99)

d n

dt=

n∞ − n

τn

(2.100)

229

Plasma membrane Ca2+ -ATPase current– ICapump

ICapump =zCa F Vc

Cm BCa Ac

· JpCa

1 +

(Km,pCa

[ Ca2+ ]i

) hPCa(2.101)

Na+ / K+ exchanger current – INaK

INaK = gNaK fNaK kNaK nNaK (2.102)

fNaK =1

1 + 0.125 exp

(− 0.1VF

RT

)+

0.00219 exp

([ Na+ ]o

49.71

)exp

(− 1.9VF

RT

)

(2.103)

kNaK =1

1 +

(Kd,K

[ K+ ]o

)nK(2.104)

nNaK =1

1 +

(Kd,Na

[ Na+ ]i

)nNa(2.105)

230

Sodium/Calcium exchanger current – INaCa

INaCa =JNaCa zCa Vc F

Ac Cm

(2.106)

JNaCa = (Allo)(∆E) (2.107)

∆E =E1

E2(E3 + E4)(2.108)

Allo =1

1 +

(Km,Allo

[ Ca2+ ]i

) nallo(2.109)

E1 = JNaCa

([ Na+ ]i

3 [ Ca2+ ]o expγVF

RT

)−

JNaCa

([ Na+ ]o

3 [ Ca2+ ]i exp(γ − 1)VF

RT

)(2.110)

E2 = 1 + ksat exp(γ − 1)VF

RT(2.111)

E3 = Km,Cao [ Na+ ]i3 + Km,Nao

3 [ Ca2+ ]i +

Km,Nai3 [ Ca2+ ]o

(1 +

[ Ca2+ ]iKm,Cai

)(2.112)

E4 = [ Ca2+ ]o [ Na+ ]i3 + [ Na+ ]o

3 [ Ca2+ ]i +

[ Na+ ]o3 Km,Cai

(1 +

[ Na+ ]iKm,Nai

3)

(2.113)

Sodium-potassium-chloride co-transport – INaKCl

INaKCl = −RNaKCl zCl Am LNaKCl RFT× ln

[ Na+ ]o

[ Na+ ]i

[ K+ ]o

[ K+ ]i

([ Cl− ]o

[ Cl− ]i

)2

(2.114)

INaKCl−Na = INaKCl−K = −0.5 INaKCl−Cl (2.115)

231

Sarcoplasmic reticulum control and SR currents

Iup = Iup[ Ca2+ ]i

100 ( [ Ca2+ ]i + Km, up )(2.116)

Itr =([ Ca2+ ]u − [ Ca2+ ]r) zCa F volu

100 τ tr

(2.117)

Irel = Frel( R10

2 + Rleak )([ Ca2+ ]r − [ Ca2+ ]u ) zCa F volr100 τ rel

(2.118)

d[ Ca2+ ] u

dt=

Iup − ItrzCa F volu

(2.119)

d[ Ca2+ ]r

dt=

Itr − IrelzCa F volr

(1 +

[ CSQN ] KCSQN

( KCSQN + [ Ca2+ ]r)2

)−1

(2.120)

Ryanodine receptor

R00 = 1− R01 − R10 − R11 (2.121)

d R10

dt= Kr1 [ Ca2+ ]i

2 R00 − ( K−r1 + Kr2 [ Ca2+ ]i ) R10 + K−r2 R11 (2.122)

d R01

dt= Kr2 [ Ca2+ ]i R00 + K−r1 R11 − ( K−r2 + Kr1 [ Ca2+ ]i

2) R01 (2.123)

d R11

dt= Kr2 [ Ca2+ ]i R10 − ( K−r1 + K−r2 ) R11 + Kr1 [ Ca2+ ]i

2R01 (2.124)

232

Ionic balances

ICa−tot = ICaL + ICaT + INSCa + ISOCCa − 2 INaCa + ICapump (2.125)

d [ Ca2+ ]i

dt= −( ICa−tot + Iup − Irel )

Ac Cm BCa

Vc zCa F×

FBCa

(1 +

[ SCM ]Kd

(Kd + [ Ca2+ ]i )2+

[ BF ] Kd,B

( Kd,B + [ Ca2+ ]i )2

)−1

(2.126)

INa−tot = −0.5 INaKCl−Cl + INa + ISOCNa +

3( INaK + INaCa ) + INSNa + PNa · Ih (2.127)

d [ Na+ ]i

dt= − INa−tot

Ac Cm

Vc F(2.128)

IK−tot = INaKCl−K + IK1 + IK2 + IKA + IKCa + INSK + IKleak − 2 INaK

(2.129)

d [ K+ ]i

dt= − IK−tot

Ac Cm

Vc F(2.130)

ICl−tot = INaKCl−Cl + ICl (A.2)

d [ Cl− ]i

dt= − ICl−tot

Ac Cm

Vc F zCl

(2.132)

Itot = ICaL + ICaT + INa + IK1 + IK2 + IKA + IKCa + Ih + ICl +

INSCC + INaK + INaCa + IKleak + ISOC + ICapump (2.133)

dV

dt=− Itot + Ist

Cm

(2.134)

233

A.2 Model Equations – Kinetic cross-bridge model

and force generation

Kinetic cross-bridge cycling model

dM

dt= −K1M + K2Mp + K7AM (2.135)

dMp

dt= K4AMp−K1M− (K2 + K3)Mp (2.136)

dAMp

dt= K3Mp + K6AM− (K4 + K5)AMp (2.137)

dAM

dt= K5AMp− (K7 + K6)AM (2.138)

MLCK =MLCKmax

1 +(

[ Ca2+ ]iKMLCK

) pM(2.139)

K1 = CmsMLCK

1 +(

KCa,MLCK

[ Ca2+ ]i

) nM(2.140)

K6 = K1 (2.141)

Fraction phosphorylation = AMp + Mp (2.142)

Fraction stress = AMp + AM (2.143)

234

Mechanical production model

d ls

dt=

1

µs

( Fs − ks (exp( αs ( ls − ls0 )/ ls0 )− 1)) (2.144)

d la

dt=

Fa exp(β(( la − lopt )/ lopt )2)− ( fAMp AMp vx )

fAM AM + fAMp AMp(2.145)

d lx

dt=

( kx1 AMp + kx2 AM)( lc − la − ls )− ( fAMp AMp vx )

fAM AM + fAMp AMp(2.146)

lx = lc − la − ls (2.147)

Fp = kp (exp( αp ( lc − lc0 )/ lc0 )− 1 (2.148)

Fx = ( kx1 AMp + kx2 AM) lx exp(−β(( la − lopt )/ lopt )2) (2.149)

Fa = ( fAMp AM + fAMp AMp)d la

dt+ ( fAMp AMp vx ))×

exp(−β(( la − lopt )/ lopt )2) (2.150)

Fa = µs

d ls

dt+ ks (exp( αs ( ls − ls0 )/ ls0 )− 1) (2.151)

Fa = Fs = Fx (2.152)

Ft = Fa + Fp (2.153)

1-D strand model

∂V

∂t=

∂D

∂ x

∂V

∂ x− Itot

C(7.2)

235

A.3 Parameter values for ionic currents

Parameters Values DefinitionHyperpolarisation-activated current, Ihgh 0.0542 nS/pF Maximum conductance of IhPNa /PK 0.35 Na+ and K+ permeability ratioL-type calcium current, ICaL

gCaL 0.6 nS/pF Maximum conductance of ICaL

ECaL 45 mV Reversal potential of ICaL

Km,CaL 0.6 µM Half saturation concentration of ICaL

T-type calcium current, ICaT

gCaT 0.0174 nS/pF Maximum conductance of ICaT

ECaT 42 mV Reversal potential of ICaT

Fast sodium current, INa

gNa 0.12 nS/pF Maximum conductance of INa

Store-operated non-selective cation current – ISOC

gSOCCa 8.3×10−5 nS/pF Maximum conductance of theCa2+ component of ISOC

gSOCNa 5.75×10−4 nS/pF Maximum conductance of theNa+ component of ISOC

Km,SOC 100 nM Half activation constant of ISOC

Delayed rectifying potassium current – IK1

gK1 0.2275 nS/pF Maximum conductance of IK1

Delayed rectifying potassium current – IK2

gK2 0.07875 nS/pF Maximum conductance of IKA

A-type transient potassium current – IKA

gKA 0.189 nS/pF Maximum conductance of IKA

Calcium-activated potassium current – IKCa

NBKCa 5.94×106 cm−2 Channel densityτ pf

0.84 ms Fast activation time constantτ ps 35.9 ms Slow activation time constantPBKCa 3.9×10−13 cm3/s Single channel permeabilityBackground K+ leakage current – IKleak

gK−leak 0.005 nS/pF Conductance of IKleak

(Continued)

236

Parameters Values DefinitionNon-selective cation current – INSCC

gNS 0.0123 Maximum conductance of INSCC

gleak 0.009685 Maximum conductance of leakage compo-nent of INSCC

Kd,Mg 0.28 Half saturation concentrationPCa : PCs 0.89 Permeability ratio of Ca2+ and Cs+

PNa : PCs 0.9 Permeability ratio of Na+ and Cs+

PK : PCs 1.3 Permeability ratio of K+ and Cs+

Calcium-activated chloride current – ICl

gCl 0.1 pA/pF Maximal conductance of ICl

Kd,Cl 257 nM Calcium required for half activation in ICl

Plasma membrane Ca2+ -ATPase current – ICapump

JpCa 7×10−7 mM/ms Maximum flux of ICapump

Km,pCa 0.5 µN Half saturation of [ Ca2+ ]ihPCa 2 Hill coefficientNa+ / K+ exchanger current – INaK

gNak 1.7 nS/pF Maximum conductance of INaK

Kd,K 2 mM Half saturation of K+ in INaK

nK 1.5 Hill coefficient of K+ in INaK

Kd,Na 22 mM Half saturation of Na+ in INaK

nNa 2 Hill coefficient of Na+ in INaK

Sodium/Calcium exchanger current – INaCa

JNaCa 3.5×10−6 mM/ms Maximum flux of Na+ /Ca2+ exchangerKm,Allo 0.3 µM Calcium activation of INaCa

nallo 4 Hill coefficient of INaCa

ksat 0.27 Saturation of INaCa at negative potentialsγ 0.35 Energy barrier of Na+ /Ca2+ exchanger in

the electric fieldKm,Nai 30 mM Saturating concentration of [ Na+ ]iKm,Cai 7 µM Saturating concentration of [ Ca2+ ]iKm,Nao 87.5 mM Saturating concentration of [ Na+ ]oKm,Cao 1.3 mM Saturating concentration of [ Ca2+ ]oSodium-potassium-chloride cotransport current – INaKCl

LNaKCl 1.79×10−8 Cotransport coefficientnmol−2/J · s · cm−2

RNaKCl 1 Cotransport regulation(Continued)

237

Parameters Values DefinitionSR uptake current, Iup

Iup 6.68 pA Maximum SR uptake currentKm, up 1 µM Michaelis constant of the SERCA pumpSR transfer current, Itrvolr 0.07 pL Volume of the uptake compartmentτ tr 1000 ms Time constant for the internal diffusion in

ItrSR release current, IrelFrel 0.2 Scaling factor of IrelRleak 75.07×10−4 Leakage parameterτ rel 0.015 ms Time constant of IrelKCSQN 0.8 mM Binding affinity of calsequestrin[CSQN ] 15 mM Concentration of calsequestrin in the re-

lease compartmentRyanodine receptorKr1 2500 mM−2ms−1 Activation rate constantKr2 1.05 mM−1ms−1 Inactivation rate constantK−r1 0.00076 ms−1 Unbinding rate constant from activationK−r2 0.084 ms−1 Unbinding rate constant from inactivationIonic balancesFBCa 0.003 Fractional Ca2+ buffer[ SCM ] 0.3 mM Total concentration of calmodulin sites for

Ca2+

[ BF ] 0.2 mM Total concentration of other buffer sitesfor Ca2+

Kd 2.6× 10−4 mM Dissociation constantKd,B 5.29810−4 mM Dissociation constant in buffer

Table A.1: Parameter values on ionic currents

238

A.4 Parameter values for force conctracion and

1-D strand model

Parameters Values DefinitionKinetic cross-bridge modelMLCKmax 0.84 Maximum level of MLCKKMLCK 721.746 nM Half activation of MLCKpM 1 Hill coefficient for MLCKK2 0.0012387 ms−1 Myosin dephosphorylation rate constantK3 0.0001419 ms−1 Cross-bridge formation rate constantK4 0.00035475 ms−1 Cross-bridge detachment rate constantK5 0.001238 ms−1 Myosin dephosphorylation rate constantK7 3.78×10−05 ms−1 Latch state detachment constantCms 0.001 Conversion parameter from s to msnM 8.7613 Hill coefficientKCa,MLCK 256.98 nM Half activation of MLCK set by [ Ca2+ ]i

(Continued)

239

Parameters Values DefinitionMechanical production modellc 123 or 150* µm Length of the whole celllc0 40 µm Length of the cell at zero passive forcels0 30 µm Length of series viscoelastic component at

zero forcelopt 100 µ Optimal length of active contractile com-

ponentkx1 12.5 µN/µm Phosphorylated cross-bridge stiffness con-

stantkx2 8.8 µN/µm Latch bridge stiffness constantks 0.2 µN Series element stiffness constantkp 0.1 µN Parallel element stiffness constantvx 5 µm/ms Cross-bridge cycling velocityfAMp 1.3 µN ms µm −1 Friction constant for phosohorylated

cross-bridgesfAM 85.5 µN ms µm −1 Friction constant for latch bridgesµs 0.01 µN ms µm −1 Viscosity coefficient of series elementαs 4.5 Length modulation for series viscoelastic

elementαp 0.1 Length modulation for passive elementβ 7.5 Length modulation constant for active

and cross-bridge elements1-D strand modelx 0.028 µm Separation of cellsD 0.003 µm/ms Diffusion coefficient

Table A.2: Parameter values on force contraction and 1-D strandmodel. *The value of the length of the cell used depended on the simulations inan isometric condition.

240

A.5 Other parameter values

Parameters Model values DefinitionT 308 K Model temperatureF 96485 C/mol Faraday constantR 8.314 J ·K−1mol−1 Universal gas constantzNa 1 Valency of Na+

zK 1 Valency of K+

zCa 2 Valency of Ca2+

zCl -1 Valency of Cl−

[ Na+ ]o 140 mM Na+ extracellular concentration[ K+ ]o 5.4 mM K+ extracellular concentration[ Ca2+ ]o 146.4 mM Ca2+ extracellular concentration[ Mg2+ ]o 1.2 mM Mg2+ extracellular concentration[ Cl− ]o 146.4 mM Cl− extracellular concentrationCm 1 µF cm−2 Specific membrane capacitanceAm 1×10−4 pF/cm−2 Area of the cellAc /Vc 4 cm−1 Cell area to volume ratioBCa 0.022

Table A.3: Other parameter used in the model

241

A.6 Initial values

Parameters Values DefinitionV -58 mV Membrane potential[ Ca2+ ]i 0.000220 mM Intracellular concentration of Ca2+

[ K+ ]i 140 mM Intracellular concentration of K+

[ Na+ ]i 4 mM Intracellular concentration of Na+

[ Cl− ]i 130 mM Intracellular concentration of Cl−

y 0.00241 Activation gating variable of Ihd 0.01136 Activation gating variable of ICaL

f1 0.94760 Fast inactivation gating variable ofICaL

f2 0.94760 Slow inactivation gating variable ofICaL

b 0.52438 Activation gating variable of ICaT

g 0.03381 Inactivation gating variable of ICaT

m 0.13319 Activation gating variable of INa

h 0.38538 Inactivation gating variable of INa

q 0.07094 Activation gating variable of IK1

r1 0.1758 Fast inactivation gating variable ofIK1

r2 0.1758 Slow inactivation gating variable ofIK1

p 0.07596 Activation gating variable of IK2

k1 0.9964 Fast inactivation gating variable ofIK2

k2 0.9964 Slow inactivation gating variable ofIK2

s 0.03340 Activation gating variable of IKA

x 0.08139 Inactivation gating variable of IKA

pf

1

1 + exp(−V−V1/2KCa

18.25

) Activation gating variable of IKCa

(Continued)

242

Parameters Values Definition

ps

1

1 + exp(−V−V1/2KCa

18.25

) Inactivation gating variable of IKCa

c1 0.0005 Activation gating variable of posi-tive ICl

c2 0.0005 Activation gating variable of nega-tive ICl

n 0.5373 Inactivation gating variable of nega-tive ICl

[ Ca2+ ]u 0.1240 mMCa2+ concentration in the uptakecompartment

[ Ca2+ ]r 0.1015 mMCa2+ concentration in the releasecompartment

M 0.701716 Free myosin stateMp 0.0609591 Phosphorated myosin stateAMp 0.0944313 Attached cross-bridge stateAM 1−M−Mp− AMp Latched cross-bridge statels 38.4489 µm Length of series elastic componentla 84.2993 µm Length of overlapping actin and

myosin filamentslx lc - ls - la Length of series cross-bridge compo-

nent

Table A.4: Initial condition values

243