mechanisms underlying the contractile responses of

176
MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF INTESTINAL SMOOTH MUSCLES TO AZITHROMYCIN USING ANIMAL MODELS Rizwan Faisal 2014/KMU/IBMS/PhD/FS/8 PhD Thesis Pharmacology Institute of Basic Medical Sciences Khyber Medical University Peshawar October 2020

Upload: others

Post on 29-Apr-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

MECHANISMS UNDERLYING THE CONTRACTILE

RESPONSES OF INTESTINAL SMOOTH MUSCLES TO

AZITHROMYCIN USING ANIMAL MODELS

Rizwan Faisal

2014/KMU/IBMS/PhD/FS/8

PhD Thesis

Pharmacology

Institute of Basic Medical Sciences

Khyber Medical University

Peshawar

October 2020

Page 2: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

MECHANISMS UNDERLYING THE CONTRACTILE

RESPONSES OF INTESTINAL SMOOTH MUSCLES TO

AZITHROMYCIN USING ANIMAL MODELS

A thesis submitted in the partial fulfillment of the requirement for the degree of

Doctor of Philosophy

In

Pharmacology

Rizwan Faisal

2014/KMU/IBMS/PhD/FS/8

Institute of Basic Medical Sciences

Khyber Medical University

Peshawar

October 2020

Page 3: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

CERTIFICATE

This thesis by Mr. Rizwan Faisal titled “Mechanisms underlying the contractile

responses of intestinal smooth muscles to azithromycin using animal models” is

accepted in its present form, by the Department of Pharmacology, Institute of Basic

Medical Sciences, Khyber Medical University Peshawar, as satisfying thesis

requirements for award of degree of Doctor of Philosophy in Pharmacology.

Supervisor: ____________________

(Prof. Dr. Niaz Ali)

External Examiner: ____________________

(Dr…………………...)

Internal Examiner: ____________________

(Dr…………………...)

Director IBMS: ____________________

(Dr…………………...)

Dean Basic Medical Sciences,

Institute of Basic Medical Sciences. _____________________

(Prof. Dr. Jawad Ahmed)

Page 4: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

i

DEDICATION

This work is dedicated to my two beautiful souls; my parents. You have made me

stronger, better and more fulfilled than I could have ever imagined. Your positivity and

immense love has made me a better version of me. I hope I have brought pride to my

family which is a source of self- contentment and harmony at my end.

Page 5: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

ii

DECLARATION

I hereby declare that the work accomplished in this thesis is my own research effort

carried out in the department of Pharmacology, Institute of Basic Medical Sciences,

Khyber Medical University Peshawar. The thesis has been written and composed by

me.

The work in this thesis has neither been previously submitted for examination leading

to the award of a degree nor does it contains any material from the published

resources that can be considered as the violation of the international copyright law.

I also declare that I am aware of the terms ‘copyright’ and ‘plagiarism’. I will be solely

responsible for the consequences of violation to these rules (if any) found in the thesis.

The thesis has been checked for plagiarism through turnitin software.

Name: Rizwan Faisal

Signature: _____________

Date: November 16, 2021

Page 6: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

iii

ABSTRACT

Background: Azithromycin is one of the most widely prescribed antibiotics worldwide

for various clinical conditions. So far there are controversial reports regarding its

actions on intestinal smooth muscles. The mechanisms by which Azithromycin affects

intestinal contractility are still unexplored.

Objectives:

i. To determine the effects of Azithromycin on the contractility of intestinal

smooth muscles in vitro model of rabbits and rats using Power Lab system

ii. To investigate the mechanisms of Azithromycin affecting the intestinal

contractility through receptors and voltage gated ion channels by using Power

Lab system

Methodology: Rabbit’s jejunal and rat’s ileal preparations were mounted in organ

bath containing 15 ml Tyrode’s solution, constantly aerated with carbogen gas.

Azithromycin was tested on rabbit’s jejunal and rat’s ileal preparations in

concentrations of 0.01, 0.03, 0.1, 0.3, 1, 3, 5, 10 and 15µM µg/ml and response for

each concentration was recorded for 2 min. Spasmogenic activity of Azithromycin was

observed. To explore its mechanism of action, response of Azithromycin was noted in

the presence of 0.3µM Atropine, 3µM Loratadine, 0.3µM Ondansetron, 3µM

Metaclopramide, 0.3µM Verpamil, 0.3µM Propranolol, 3µM Amiodarone and 0.3µM

Atropine, Ondansetron, Verapamil and Propranolol.

Results: Mean (%) Emax for AZM was 67.6±1.6 and 54.0±2.1 for rabbits’ jejunal and

rats’ ileal preparations, respectively. Mean (%) Emax in the presence of Atropine for

rabbits’ jejunal preparations was 2.4±0.1 and for rats’ ileal preparations it was

11.4±1.3. Similarly, mean (%) Emax was 67.9 ± 2.0 for rabbits’ jejunal preparations and

was 50.7±1.9 for rats’ ileal preparations in the presence of Loratadine. Mean (%) Emax

in the presence of Ondansetron was 27.5±0.5 for rabbits’ jejunal preparations and

34.0±2.9 for rats’ ileal preparations. In the presence of Metoclopramide, mean (%)

Page 7: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

iv

Emax was 88.4±1.2 for rabbits’ jejunal preparations and 79.1±3.8 for rats’s ileal

preparations. Mean (%) Emax in the presence of Verapamil for rabbits’ jejunal

preparations was 13.6±1.2 and for rats’ ileal preparations it was 22.3±2.5. Mean (%)

Emax in the presence of Propranolol was 10.2±2.1 for rabbits’ jejunal preparations and

15.6±1.4 for rats’ ileal preparations. Mean (%) Emax in the presence of Amiodarone

was 68.4±1.3 for rabbits’ jejunal preparations and 58.0±3.4 for rats’ ileal preparations.

The spasmogenic response of Azithromycin was completely lost when it was given in

the presence of Atropine, Ondansetron, Verapamil and Propranolol.

Conclusion: Results reveal that the spasmogenic response of Azithromycin is mainly

mediated through muscarinic receptors. However, we found involvement of mixed

pathways including serotonergic receptors, voltage gated calcium channels and

voltage gated sodium channels.

Keywords: Azithromycin, Acetylcholine, Atropine, Voltage gated sodium channels,

Voltage gated calcium channels, Voltage gated potassium channels

Page 8: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

5

ACKNOWLEDGMENT

Thanks to Allah, Almighty for all his blessings upon me throughout my life and who

enabled me to complete my thesis.

I am deeply obliged to my supervisor Prof. Dr. Niaz Ali who guided me during my

research and thesis. His keen interest and valuable suggestions made this work to end

in a refined manner.

It is my privilege to thank my family, my wife, who has been extremely supportive

throughout and has made countless sacrifices to help me reached to this status. My

children, Rameen Karim and Hania Karim, they had been my inspiration to achieve

greatness, without them, I would not have reached my goal where I am standing now.

My mother, brother, sister in law and nephew Alyaan Karim; deserves special thanks

for their continued support and encouragement. I have been blessed with such kind

and affectionate family who had always faith in me.

A very special thanks to my father-in-law, Prof. Dr. Zabta Khan Shinwari, who has

always been my true role model and mentor, his encouraging words and assistance in

every step of my research work has always given me strength.

Lastly, I am indebted to my endear colleague, PhD scholar, Muhammad Nabi who

untiringly assisted me in Power Lab activity. I am also very thankful to Amjad Khan,

Pharmacology laboratory technician, whose association and facilitation helped me in

performing various activities related with my research work and stood firm by my side,

which is a true asset.

RIZWAN FAISAL

Page 9: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

6

Page 10: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

7

TABLE OF CONTENTS

DEDICATION i

DECLARATION

ii

ABSTRACT

iii

ACKNOWLEDGEMENTS

v

TABLE OF CONTENTS vi

LIST OF TABLES

x

LIST OF FIGURES

xi

LIST OF ABBREVIATIONS xiv

1. INTRODUCTION

1

1.1 Neurotransmitters

2

1.1.1 Acetylcholine

2

1.1.2 Histamine

3

1.1.3 Dopamine

6

1.1.4 Serotonin

7

1.2 Voltage gated ion channels

10

Page 11: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

8

1.2.1 Sodium channels

10

1.2.2 Calcium channels

11

1.2.3 Potassium channels

12

1.3 Hormonal control of gastrointestinal motility

16

1.4 Intrinsic and extrinsic innervation of gastrointestinal motility

17

1.4.1 Parasympathetic control of gastrointestinal tract

17

1.4.2 Sympathetic control of gastrointestinal tract

18

1.4.3 Role of gut microbiota in gastrointestinal motility

18

1.5 Animal models for in vitro preparation

21

1.5.1 History of experimental animals

21

1.5.2 Purpose of animal models

21

1.5.3 Animal models for gastrointestinal disorders

22

1.5.3.1 In vitro animal models for GI motility

23

1.5.3.2 Rabbits

23

1.5.3.3 Rats

24

1.5.3.4 Advantages of in vitro techniques

25

1.6 Current prokinetic agents

25

Page 12: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

9

1.6.1 Cholinergic agonists

26

1.6.2 Serotonergic agonists

26

1.6.3 Dopaminergic antagonists

27

1.6.4 Erythromycin

28

1.6.5 Miscellaneous drugs

29

1.7 Azithromycin

29

1.7.1 Source

29

1.7.2 Structure

29

1.7.3 History

32

1.7.4 Administration

32

1.7.5 Mechanism of action

32

1.7.6 Half-life and tissue penetration

34

1.7.7 Clinical uses

34

1.7.8 Advantages of Azithromycin over Erythromycin

35

1.7.9 Advantages of Azithromycin over other antibiotics

35

1.8 Pharmacological actions of Azithromycin

35

1.8.1 Antibacterial action

35

Page 13: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

10

1.8.2 Immunomodulatory action

36

1.8.3 Anti-inflammatory action

37

1.9 Research studies of azithromycin related with smooth muscle contractility

37

1.9.1 Effect of Azithromycin on aortic strips

37

1.9.2 Effect of Azithromycin on uterine smooth muscles

38

1.9.3 Effect of Azithromycin on respiratory smooth muscles

38

1.9.4 Effect of Azithromycin on GI smooth muscles

41

1.9.4.1 Azithromycin and esophageal smooth muscle contractility

41

1.9.4.2 Effect of Azithromycin on gall bladder smooth muscles

41

1.9.4.3 Azithromycin and gastroparesis

42

1.9.4.4 Gastric motility disorders

44

1.9.4.5 Azithromycin and intestinal contractility

45

Page 14: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

11

2. MATERIAL AND METHODS

48

2.1 Animals

48

2.2 Chemicals and drugs

48

2.3 Preparation of isolated tissue

49

2.3.1 Rabbit’s jejunal and rat’s ileal preparation

49

2.4 Effects of drugs on rabbit’s jejunal and rat’s ileal preparation

52

2.4.1 Effect of azithromycin on rabbit’s jejunal and rat’s ileal preparation

52

2.4.2 Validation of curves

52

2.4.2.1 Acetylcholine

52

2.4.2.2 Atropine

52

2.4.2.3 Loratadine

53

2.4.2.4 Ondansetron

53

2.4.2.5 Metoclopramide

54

2.4.2.6 Verapamil

54

2.4.2.7 Propranolol

54

2.4.2.8 Amiodarone

54

2.4.3 Effect of Azithromycin in the presence of receptor and channel

blockers 55

Page 15: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

12

2.4.3.1 Effect of Azithromycin in the presence of Atropine

55

2.4.3.2 Effect of Azithromycin in the presence of Loratadine

55

2.4.3.3 Effect of Azithromycin in the presence of Ondansetron

55

2.4.3.4 Effect of Azithromycin in the presence of Metoclopramide

56

2.4.3.5 Effect of Azithromycin in the presence of Verapamil

56

2.4.3.6 Effect of Azithromycin in the presence of Propranolol

56

2.4.3.7 Effect of Azithromycin in the presence of Amiodarone

57

2.4.3.8 Effect of Azithromycin in the presence of Atropine, Ondansetron,

Verapamil and Propranolol

57

2.5 Statistical analysis

57

3. RESULTS

58

3.1 Validation of curves

58

3.1.1 Atropine

58

3.1.2 Loratadine

61

3.1.3 Ondansetron

64

3.1.4 Metoclopramide

67

3.1.5 Verapamil

70

Page 16: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

13

3.1.6 Propranolol

73

3.1.7 Amiodarone

76

3.2 Effect of Azithromycin on rabbit’s jejunal and rat’s ileal preparation

79

3.3 Effect of Azithromycin in the absence and presence of antagonists

82

3.3.1 Effect of Azithromycin in the absence and presence of Atropine

82

3.3.2 Effect of Azithromycin in the absence and presence of Loratadine

87

3.3.3 Effect of Azithromycin in the absence and presence of Ondansetron

92

3.3.4 Effect of Azithromycin in the absence and presence of

Metoclopramide 97

3.3.5 Effect of Azithromycin in the absence and presence of Verapamil

102

3.3.6 Effect of Azithromycin in the absence and presence of Propranolol

107

3.3.7 Effect of Azithromycin in the absence and presence of Amiodarone

112

3.3.8 Effect of Azithromycin in the absence and presence of Atropine,

Ondansetron, Verapamil and Propranolol

117

4. DISCUSSION

122

5. CONCLUSION

130

REFERENCES

131

PUBLICATION

Page 17: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

14

LIST OF TABLES

Table 3.1 Effect of Atropine on rabbits’ jejunal preparations and rats’ ileal

preparations

58

Table 3.2 Effect of Loratadine on rabbits’ jejunal preparations and rats’ ileal

preparations

61

Table 3.3 Effect of Ondansetron on rabbits’ jejunal preparations and rats’

ileal preparations

64

Table 3.4 Effect of Metaclopramide on rabbits’ jejunal preparations and

rats’ ileal preparations

67

Table 3.5 Effect of Verapamil on rabbits’ jejunal preparations and rats’ ileal

preparations

70

Table 3.6 Effect of Propranolol on rabbits’ jejunal preparations and rats’

ileal preparations

73

Table 3.7 Effect of Amiodarone on rabbits’ jejunal preparations and rats’

ileal preparations

76

Table 3.8 Effect of Azithromycin on rabbit’s jejunal preparations and rat’s

ileal preparations

79

Table 3.9 Effect of Azithromycin in the absence and presence of Atropine

on rabbit’s jejunal preparations and rat’s ileal preparations

82

Table 3.10 Effect of Azithromycin in the absence and presence of Loratadine

on rabbit’s jejunal preparations and rat’s ileal preparations

87

Table 3.11 Effect of Azithromycin in the absence and presence of

Ondansetron on rabbit’s jejunal preparations and rat’s ileal

preparations

92

Table 3.12 Effect of Azithromycin in the absence and presence of

Metaclopramide on rabbit’s jejunal preparations and rat’s ileal

preparations

97

Table 3.13 Effect of Azithromycin in the absence and presence of Verapamil

on rabbit’s jejunal preparations and rat’s ileal preparations

102

Table 3.14 Effect of Azithromycin in the absence and presence of

Propranolol on rabbit’s jejunal preparations and rat’s ileal

preparations

107

Table 3.15 Effect of Azithromycin in the absence and presence of

Amiodarone on rabbit’s jejunal preparations and rat’s ileal

preparations

112

Table 3.16 Effect of Azithromycin in the absence and presence of Atropine,

Ondansetron, Verapamil & Propranolol (AOVP) on rabbit’s

jejunal preparations and rat’s ileal preparations

117

Page 18: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

15

LIST OF FIGURES

Figure 1.1 Chemical structure of acetylcholine 4

Figure 1.2 Gq-coupled receptor signal transduction pathway 4

Figure 1.3 Chemical structure of histamine 5

Figure 1.4 Gs and Gi-coupled receptor signal transduction pathway 5

Figure 1.5 Chemical structure of dopamine and serotonin 8

Figure 1.6 Ligand gated ion channels 9

Figure 1.7 Voltage Gated Sodium Channels 13

Figure 1.8 Voltage gated calcium channel 14

Figure 1.9 Voltage-gated Potassium channel 15

Figure 1.10 Intrinsic and Extrinsic Innervation of Gut motility 19

Figure 1.11 Bacterial distribution in the GI tract 20

Figure 1.12 Chemical structure of azithromycin 30

Figure 1.13 3D structure of azithromycin 31

Figure 1.14 Mechanism of action of Azithromycin 33

Figure 2.1 Small and Large intestines of a rabbit 50

Figure 2.2 Isolated rabbit’s jejunal preparation 51

Figure 3.1a Effect of Atropine on spontaneous rabbits’ jejunal preparations 59

Figure 3.1b Effect of Atropine on spontaneous rats’ ileal preparations 60

Figure 3.2a Effect of Loratadine on spontaneous rabbits’ jejunal preparations 62

Figure 3.2b Effect of Loratadine on spontaneous rats’ ileal preparations 63

Figure 3.3a Effect of Ondansetronon spontaneous rabbits’ jejunal preparations 65

Figure 3.3b Effect of Ondansetron on spontaneous rats’ ileal preparations 66

Figure 3.4a Effect of Metoclopramide spontaneous rabbits’ jejunal

preparations

68

Figure 3.4b Effect of Metoclopramide on spontaneous rats’ ileal preparations 69

Figure 3.5a Effect of Verapamil spontaneous rabbits’ jejunal preparations 71

Figure 3.5b Effect of Verapamil on spontaneous rats’ ileal preparations 72

Figure 3.6a Effect of Propranolol spontaneous rabbits’ jejunal preparations 74

Figure 3.6b Effect of Propranolol on spontaneous rats’ ileal preparations 75

Figure 3.7a Effect of Amiodarone spontaneous rabbits’ jejunal preparations 77

Figure 3.7b Effect of Amiodarone on spontaneous rats’ ileal preparations 78

Figure 3.8a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations

80

Figure 3.8b Effect of Azithromycin on rat’s ileal preparations 81

Figure 3.9a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Atropine

83

Figure 3.9b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Atropine

84

Figure 3.9c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of Atropine

85

Page 19: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

16

Figure 3.9d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Atropine

86

Figure 3.10a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Loratadine

88

Figure 3.10b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Loratadine

89

Figure 3.10c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of Loratadine

90

Figure 3.10d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Loratadine

91

Figure 3.11a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Ondansetron

93

Figure 3.11b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Ondansetron

94

Figure 3.11c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of Ondansetron

95

Figure 3.11d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Ondansetron

96

Figure 3.12a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Metaclopramide

98

Figure 3.12b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Metaclopramide

99

Figure 3.12c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of

Metaclopramide

100

Figure 3.12d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Metaclopramide

101

Figure 3.13a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Verapamil

103

Figure 3.13b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Verapamil

104

Figure 3.13c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of Verapamil

105

Figure 3.13d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Verapamil

106

Figure 3.14a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Propranolol

108

Figure 3.14b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Propranolol

109

Figure 3.14c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of Propranolol

110

Figure 3.14d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Propranolol

111

Page 20: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

17

Figure 3.15a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Amiodarone

113

Figure 3.15b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Amiodarone

114

Figure 3.15c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of Amiodarone

115

Figure 3.15d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Amiodarone

116

Figure 3.16a Effect of Azithromycin on spontaneous rabbit’s jejunal

preparations in the absence and presence of Atropine,

Ondansetron, Verapamil & Propranolol

118

Figure 3.16b Effect of Azithromycin on rat’s ileal preparations in the absence

and presence of Atropine, Ondansetron, Verapamil & Propranolol

119

Figure 3.16c Comparison of Azithromycin’s effect on spontaneous rabbit’s

jejunal preparations in the absence and presence of Atropine,

Ondansetron, Verapamil & Propranolol

120

Figure 3.16d Comparison of Azithromycin’s effect on rat’s ileal preparations in

the absence and presence of Atropine, Ondansetron, Verapamil &

Propranolol

121

Page 21: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

18

LIST OF ABBREVIATIONS

ACh Acetylcholine ASMs Airway smooth muscles BA Bile Acids BAL Bronchoalveolar Lavage BALF Broncheoalveolar Lavage Fluid B-ARs Beta-adrenoceptors BOS Bronchiolitis Obliterans Syndrome Ca++ Calcium cAMP Cyclic Adenosine monophosphate CCL5 Chemokine ligand 5 CF Cystic Fibrosis CK Creatinine kinase COPD Chronic obstructive pulmonary disease Cyt P450 Cytochrome P450 DA Dopamine DAG DPB

Diacylglycerol Diffuse Pan bronchiolitis

DRC Dose Response Curve EC SR

Enterochromafin Sarcoplasmic Reticulum

ERK Extracellular regulated protein kinases ERY Erythromycin FDA Federal Drug Administration FEV1 Forced expiratory volume in one second GBCI Gallbladder Contraction Index GERD Gastroesophageal Reflux Disease GFR GGT

Glomerular filtration rate Gamma glutamyl transferase

GI Gastrointestinal GPCR Gαq

G protein coupled receptor Gq alpha sub unit

HB 5-HT

Hemoglobin Serotonin

ICC Interstitial cells of Cajal IFN-γ IP3

Interferon-gamma Inositol triphosphate

IL-1β Interleukin-1 beta Kv Potassium channel LPS Lipopolysaccharide LTx Lung Transplantation LVDCCs L-type voltage dependent calcium channels MCs Mast cells MLCK Myosin light chain kinase MMCs Migrating Motor Contractions Nav Sodium channel (voltage gated)

Page 22: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

19

NE Norepinephrine

NF-κB Nuclear factor Kappa-B NZW New Zealand white PCV Packed cell volume PKA Protein kinase A PKC PIP2

Protein kinase C Phosphatidylinositol 4,5-bisphosphate

R848 Resiquimod RCT Sp 1 STIM

Randomized control trial Specificity protein 1 Stromal interaction molecule

TEC Total erythrocyte count TLC Total leukocyte count TRs TRPC

Tracheal rings Transient receptor potential channel

VGCCs

Voltage gated calcium channels

1 INTRODUCTION

Intestinal motility has a complex function and is regulated by sensory and motor system

of the enteric nervous system (ENS) which in turn are linked and controlled by central

and autonomic nervous systems (Furness et al., 2014). ENS performs various functions

like food propulsion, regulation of blood flow and immunological defense (Fleming et

al., 2020). The intestine is made up of smooth muscles, in most areas of the tract they

are aligned in circular and longitudinal layers (Campbell, 2015). The function of these

smooth muscles is to produce tonic contractions that maintain the dimension of organ

against a load such as imposed by a bolus of food. They also produce powerful

contractions which results in shortening of muscles to push the bolus along

gastrointestinal tract (GIT) (Bitar, 2003). There are gap junctions between adjacent

smooth muscle cells which keep cells electrically coupled. When a portion of smooth

muscle is depolarized, that depolarization spreads to adjacent portions of smooth

muscle which leads to a well-coordinated series of contraction (Haffen et al., 2020).

The receptors responsible for the motility are present on smooth muscle cells and on

the neurons in the intrinsic and extrinsic parts of the ENS (Roa et al., 2016).

Page 23: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

20

Neurotransmitters and their receptors perform a key role in the regulation of intestinal

motility. Mostly, the neurotransmitters (e.g. dopamine, acetylcholine, serotonin) found

in the ENS are similar to those present in the central nervous system (CNS) (Nezami et

al., 2010). Initially, most of studies on neurotransmitters were conducted in exploring

their role in various disorders of the CNS, “fight or flight” responses, signal

transmission through a chemical synapse and controlling blood flow throughout the

body (Mittal et al., 2017). Later on, researchers identified the significant involvement

and regulation of the neurotransmitters in the regulation of GI physiology as well

(Reynolds, 2016).

Certain receptor agonists and antagonists interacts directly to transform excitatory or

inhibitory signals of intestinal tract. In view of this complexity it is not surprising that

our understanding regarding the mechanisms of actions of different neurotransmitters

and drugs which effects the gut motility is still inadequate. However in recent past,

considerable advancements have been attained, and drug therapy for gut dysmotility is

emerging (Mittal et al., 2017).

1.1 Neurotransmitters involved in gut motility:

1.1.1 Acetylcholine:

Acetylcholine (ACh) is the main excitatory neurotransmitter of the ENS, and

muscarinic type of cholinoreceptor mediates the excitatory effects of ACh on intestinal

smooth muscles (Jonge et al., 2013). The excitatory effects of parasympathetic nerves

acting on intestinal smooth muscles is mediated by ACh indirectly through an effect on

the ENS (Ehlert et al., 2012; Gao et al., 2016). By depolarizing smooth muscles ACh

starts their contraction via stimulating muscarinic receptors.

ACh is an esterified form of choline and acetic acid. ACh has choline molecule which

is acetylated at the oxygen atom (Figure 1.1). It has highly charged

polar ammonium group (Suryanarayanan, 2014). ACh has been proved to amplify

spontaneous contractions in the small intestine of rabbits (Parthasarathy et al., 2015).

There are five different muscarinic receptor subtypes (M1-M5) (Radu et al., 2017), M3

is the subtype of receptor that is mainly found in the smooth muscles. These receptors

are excitatory in nature and are directly involved in mediation of gastrointestinal (GI)

smooth muscle’s contraction (Ehlert et al., 2012; Gao et al., 2016).

Page 24: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

21

M3-receptors are coupled to Gq-receptors and stimulation of M3/Gq‐coupled receptors

activates phospholipase C (PLC) which breaks the phospholipid constituent of the

plasma membrane, phosphatidylinositol-4,5-bisphosphate (P1P2), in to two secondary

messengers, inositol-1,4,5-triphosphate (IP3) and diacylglycerol (DAG). DAG is

restricted to the membrane, where it stimulates a phospholipid and calcium sensitive

protein kinase called protein kinase C. IP3 diffuses through the cytoplasm to generate

release of Ca2+ from sarcoplasmic reticulum (SR) and it ultimately results in smooth

muscle contraction of GIT (Figure 1.2) (Montgomery et al., 2016).

ACh is preserved in storage vesicles present in the cholinergic neurons. ACh is released

into the neuromuscular junction (NMJ) after arrival of a nerve impulse. In NMJ it binds

to a receptor in post synaptic membrane of a muscle fiber, permeability of the

membrane is changed which causes the channels to open up and with the result sodium

ions (positively charged) starts their flow inside the muscle cells. When sodium

channels along endplate membrane are fully activated after successive nerve impulses,

muscle cell contracts (Jimsheleishvili et al., 2019). This is the logic for using ACh

agonists for hypomotility and for acute intestinal pseudo obstruction in which

increasing cholinergic signaling is the first line treatment.

1.1.2 Histamine:

Histamine is an amine widely present in the human body especially in the skin, lungs,

and GIT (Fabisiak et al., 2017). Histamine is a short acting neuromodulator, it regulates

the release of acetylcholine, serotonin and norepinephrine (Kapalka, 2019). Mast cells

are responsible for most of the histamine production and to a lesser extent it is also

produced by basophils, gastro enterochromoffin (EC) cells and histaminergic neurons.

Histamine is believed to affect at least 3 major functions in GIT: regulation of GI

motility, amplification of gastric acid secretion, and adjustment of mucosal ion

secretion (Fabisiak et al., 2017).

Histamine is formed by the decarboxylation of the histidine (Figure 1.3). It is an organic

molecule with four histamine receptors. H1, H2, H3 & H4, they are G protein-coupled

receptors. Histamine shows various effects because different histamine receptors are

present on different cell types and they work by different mechanisms of intracellular

signaling. H1-receptors are mainly found in mast cells and are involved in allergic

conditions. H2-receptors are found in heart, brain, parietal cells and neutrophils. H3-

Page 25: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

22

receptors are mainly present in peripheral nervous system and brain. H4-receptors are

found in blood cells (monocytes, eosinophils, mast cells) (Thangam et al., 2018). As

far as GIT is concerned, H1 and H4 subtypes of histamine receptors are mainly found in

intestinal smooth muscles (Mittal et al., 2017). Histamine is also responsible for

increase in intestinal motility. It has been revealed that overproduction of histamine by

mast cells causes diarrhea (Fabisiak et al., 2017).

Histamine has many effects on the body, one of the effects is contraction of smooth

muscle fibers in the stomach, intestines, lungs and uterus, (Bowen, 2019). Coupling of

H1-receptors are dual in nature. They are activated through Gq protein alpha sub unit

family (Gαq/11), which in turn activates phospholipase C (PLC) and ultimately

intracellular calcium (Ca++) levels are increased, resulting in smooth muscle contraction

of GIT (Figure 1.4) (Thangam et al., 2018). They are also associated to Gi/o proteins,

resulting in decrease production of cAMP (Monczor et al., 2016). Decrease in cAMP

levels result in increase in the smooth muscle contraction (Figure 1.2) (Bilington et al.,

2003; Mizuta et al., 2013; Kuo et al., 2015).

Figure 1.1: Chemical structure of Acetylcholine (Steven et al., 2011)

Page 26: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

23

Figure 1.2: Gq-coupled receptor signal transduction pathway (Zhag et al., 2016)

Figure 1.3: Chemical structure of Histamine (Parsons et al., 2006)

Page 27: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

24

Figure 1.4: Gs and Gi-coupled receptor signal transduction pathway

(Rosenbaum et al., 2014)

1.1.3 Dopamine:

Dopamine (DA), also called hydroxytyramine, a nitrogen-containing organic

compound (Figure 1.5). It also act as a neurotransmitter. Dopamine receptors are

involved in the regulation of motor activity and several neurological disorders.

Dopamine receptors directly regulate neurotransmission of other neurotransmitters,

release of cyclic adenosine monophosphate, cell proliferation, and differentiation

(Mishra et al., 2018).

An important factor in the regulation of GI motility is the dopaminergic mechanisms.

Enteric neurons and epithelial cells in intestine produces 50% of dopamine (Xue et al.,

2018). It is a critical neurotransmitter whose release from enteric dopaminergic neurons

and subsequent binding to DA receptors in GI smooth muscle inhibits the GI motility

mainly by activating D2 receptors (Giudicessi et al., 2018; Li et al., 2006). There are 5

sub types of DA (D1-5) which are further classified into 2 subclasses. D1 and D5 are

placed in “D1-like” DA receptors and D2, D3, and D4 are placed in “D2-like”DA

receptors.

Page 28: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

25

The D2 receptor has 7 transmembrane spanning domains. D1 like dopamine receptors

activate adenylyl cyclase while D2 like dopamine receptors inhibit adenylyl cyclase

(Xue et al., 2018). Various subtypes of DA receptors are found in the intestinal smooth

muscles but D2 is found abundantly (Ayano, 2016). Stimulation of Gs-coupled receptors

results in activation of adenylyl cyclase activity leading to an increase in cyclic AMP

(cAMP) levels which in turn activates protein kinase A (PKA). PKA phosphorylates

the myosin light chain kinase (MLCK), promoting smooth muscle relaxation (Figure

1.4) (Mizuta et al., 2013).

Metoclopramide (D2-receptor antagonist), blocks D2 receptors in the chemoreceptor

trigger zone as well as in the GI smooth muscles (Zabirowicz et al., 2019). It

antagonizes dopamine mediated relaxation on smooth muscle of GIT and exerts its

prokinetic effect. Metoclopramide increases the tone and amplitude of esophageal and

gastric contractions and thus increases gastric emptying (Mikami et al., 2016). The

resting tone of the lower esophageal sphincter is increased and at the same time relaxes

pyloric sphincter and duodenal bulb resulting in increased peristalsis (Isola and Adams,

2020).

1.1.4 Serotonin (5-HT):

Serotonin, also called 5-hydroxytryptamine (5-HT), is derived from the amino

acid tryptophan (Figure 1.5). It is a potent neurotransmitter which acts as a

vasoconstrictor (Kim et al., 2018). 5-HT as a chemical messenger is most widely

studied (McCorvy et al., 2015). 5-HT when combines with serotonergic receptors,

intestinal motility is increased (Kindig et al., 2015). 5-HT has various subtypes but 5-

HT3 & 5-HT4 are predominantly present in the intestinal smooth muscles (Mittal et al.,

2017). All 5-HT receptors are membrane-bound G-protein-coupled receptors (GPCRs)

except 5HT3 which is a ligand gated ion channel. 5-HT receptors activate intracellular

pathways by means of G proteins and produces a response. (McCorvy et al., 2015;

Giulietti et al., 2014).

5HT3-receptors belong to a group of transmembrane ion channel proteins which upon

binding with ligand, such as neurotransmitter, results in conformational change. This

allows potassium (K+), sodium (Na+) and Ca2+ions to move across the cell membrane.

Being an excitatory neurotransmitter it results in depolarization which leads to smooth

Page 29: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

26

muscle contraction (Figure 1.6) (Thompson et al., 2006). 5-HT4 receptor amplify the

peristaltic reflex pathways by exerting its action presynaptically on nerve terminals to

increase ACh release (Terry et al., 2016).

Researchers believed that 5-HT regulates the GI function because most of 5-HT is

synthesized and then stored in GIT. Moreover the agonists and antagonists alter the

intestinal behavior (Bomstein, 2012). Nearly 90–95% of the human body’s 5-HT is

synthesized and stored in EC cells of the intestinal mucosa where it controls motility,

secretion and sensation (Fuentes et al., 2016).

5-HT remained focus of clinical studies for intestinal motility disorders. 5-HT is a

regulatory neurotransmitter of enterocytes, enteric neurons and smooth muscles. It also

controls the development and long term consequences of central nervous system (CNS)

and ENS. Mucosal EC cells behave as sensory transducers to the mucosal signaling

molecule. 5-HT is secreted into the wall of GIT and starts peristaltic movements and

secretory reflexes in response to mechanical stimulation. EC cells release 5-HT which

affect both sensory motor and secretomotor neurons and also enterocytes and smooth

muscles. 5-HT has a key role in mediation of signals to CNS (Israelyan et al, 2019).

A. Dopamine

Page 30: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

27

B: Serotonin

Figure 1.5: Chemical structure of dopamine and serotonin (Steven et al., 2011)

Page 31: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

28

Figure 1.6: Ligand gated ion channels (Pacheco, 2007)

Page 32: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

29

1.2 Voltage gated ion channels involved in gut motility:

Besides neurotransmitters there are also voltage-gated ion channels (VGICs) which

control intestinal motility as well as alter the normal physiology (Radulovic et al, 2015).

VGICs are the main proteins which are permeable to ions and gated by changes in the

voltage of transmembrane. They are responsible for cell’s excitability. Muscle

contraction and synaptic vesicle exocytosis are the major cellular events that are

initiated by action potential generated by VGICs in muscles, neurons and secretory cells

(Lipscombe et al, 2014). Ion channels are often modified at expression level and

pharmacologically targeted in functional bowel disease (Fuentes et al., 2016).

VGICs open and close in reaction to electrical potential changes across the cell

membrane in which the channel is located and are structurally and functionally

homologous. Ion channels are dissimilar from other kind of membrane proteins, named

as transporters. Transporters mostly perform transmembrane transport of various

metabolites. Ion channels on the other hand are responsible for transducing ion-coded

information (Zhang et al., 2018). Ion channels are present in gut and their cell

particularity and position shows their role in normal and abnormal function in bowel

diseases. Voltage gated ion channels in smooth muscles and epithelial cells controls the

different phases of digestion like fluid secretion, absorption and motility (Fuentes et al.,

2016). A combination of the sodium, calcium, potassium and non-selective cation

channels maintain the resting potential membrane potential (Beyder et al., 2012).

VGICs are also possible targets for intervention in motility disorders of intestinal tract

(Kindig et al., 2015).

VGICs respond to altered membrane potential which results in particular ion passage.

This VGICs family include voltage-gated sodium channels (NaV), voltage-gated

potassium channels (KV) and voltage-gated calcium channels (CaV) (Moran et al.,

2015).

1.2.1 Sodium channels:

These channels are present in both the smooth muscle cells (SMC) and interstitial cells

of Cajal (ICC) (Beyder et al., 2012). NaV generate and spread the electrical signals in

cell (Moron et al., 2015). Sodium channels are made up of alpha (α) subunits that forms

Page 33: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

30

a transmembrane pore and an adjuvant beta (β) subunit. The β subunits govern channel

opening, expression and relation with cell bonding molecules with the cytoskeleton and

the extracellular molecules (Fuentes et al., 2016).

Highly sensitive sodium channels transporting sodium ions inside the cell activates and

inactivates the cells in milliseconds. They are classified in two groups according to their

sensitivity to tetrodotoxin (TTX); α-subunits that are sensitive to tetrodotoxin (TTX-S)

and those subunits that are resistant to tetrodotoxin (TTX-R) (Rocha HAC et al, 2014).

There are many isoforms of sodium channels present in the GI SMCs and ICC (Figure

1.7) (Beyder et al., 2012).

Depolarization opens the sodium channels of the axonal plasma membrane resulting in

pouring in of sodium ions. There is slow opening of potassium channels permitting

outpouring of potassium ions (K+), bringing back the resting membrane potential.

Calcium channels are voltage-dependent and are responsible for depolarization in some

cells. There are sensing regions on the sodium channel proteins having positive charge,

which upon depolarization moves towards negatively charged outer surface of

membrane. This allows transit of Na ions after opening of channel. Within a millisecond

of channel activation, the voltage perceiving region reverts back and a channel disabling

portion moves back to stop the channel and the channel proteins returns back to its

normal resting potential (Martin et al., 2015).

1.2.2 Calcium channels:

Calcium channels are of two types; L-type (CaV1) low-voltage-gated channels and T-

type (CaV3). L-type Ca2+ channels are also known as the high voltage gated channels

the while T-type Ca2+ channels are low-voltage-gated. Upon depolarization L-type

channels are slowly activated than T-type calcium channels but shows higher

communication in SMC. Calcium ions required for initiation of excitation and

contraction is made available by these channels (Beyder et al., 2012). Cytosolic calcium

is increased following calcium channel opening after membrane depolarization in

SMC. In gut, periodic motor patterns as in large and small intestine, mediators and

neurotransmitters operate on a structure in which slow waves originate in ICC and make

easier the achievement of the attainment of the stimulation threshold of calcium

channels intermittently and creation of action potential (Curro, 2016).

Page 34: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

31

CaV generate action potentials in a manner exactly like voltage-dependent Na+ channels.

Ca2+ channels can also modulate the shape of action potentials generated by voltage

gated NaV. The activity of calcium channels monitors a wide variety of biochemical

reactions inside the cells and they do so by influencing the concentration of calcium

inside the cell. The release of neurotransmitters at the level of synapse is the most

crucial of these biochemical reactions. Due to the wide variety of function, 16 different

calcium channel genes have been discovered. These calcium channels are different in

regard of their activation and inactivation characteristics as well as narrow

modifications in the chemical and electrical operation mediated by calcium.

Consequently the drugs that inhibit these CaV are of utmost importance in resolving a

number of ailments from heart diseases to anxiety disorders (Figure 1.8) (Freeman,

2018).

1.2.3 Potassium channels:

Half of the ion channels superfamily is made up of potassium (K+) and voltage-sensitive

potassium (KV) channels are the largest group. The wide variety of K+ channels detected

in ICCs and SMCs of the GIT reflect their rich diversity. When the intracellular

K+ concentration becomes 10-fold higher than extracellular, K+ channel open and

produces an outward K+ flux that turns the membrane potential in the negative direction

toward K+ reversal potential. As a result K+ channels are responsible for the

maintenance of resting potential, plateau current and repolarization of slow waves.

(Beyder et al., 2012). Many signaling pathways target these channels including some

kinases such as protein kinase C (PKC), protein kinase A (PKA), extracellular

regulated protein kinases (ERK) and many others (Li et al., 2014).

K+ channels are voltage gated ion channels that are numerous in number and most

variant. Almost 100 potassium channel genes are recognized so far. These genes are

divided into several groups depending upon their different gating properties. Since

channels are inactivated in minutes such as squid axon K+ channel that was studied by

Hodgkin and Hurley where others are inactivated within millisecond just like sodium

channel. Rate of action potential firing and its duration is dependent upon these

properties, thereby effecting axonal conduction and synaptic transmission. K+ channels

play an important role in generation of resting membrane potential (Figure 1.9) (Purves

et al., 2014).

Page 35: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

32

Figure 1.7: Voltage Gated Sodium Channels (Molnar et al., 2016)

Page 36: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

33

Figure 1.8: Voltage gated calcium channel (Alahamd et al., 2020)

Page 37: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

34

Figure 1.9: Voltage-gated Potassium channel (Lordfred, 2015)

Page 38: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

35

1.3 Hormonal control of gastrointestinal motility:

The GI hormones are a group of polypeptides produced by endocrine cells. The GI

hormones perform a key role in the regulation of the motor activity of the GIT. They

may exert indirect effects (neutrally mediated) or direct effects through motor activity

of smooth muscles. Motor activity of GIT is stimulated by gastrin, CCK and motilin

while secretin, vasoactive inhibitory peptide (VIP) and glucagon like peptide-1 inhibit

the motor activity.

GI functions are normally regulated by ENS which in turn is linked to autonomic and

central nervous systems (Furness et al., 2014). Biological actions of the GI hormones

are mainly mediated by binding to G-protein coupled receptors (hormone specific),

located on plasma membrane of target cell. This intracellular signal transduction

pathway entails adenylate cyclases, Ras family of GTP-binding proteins and ion

channels (Bhagavan et al., 2011).

Gastrin is found in the G cells of the gastric antrum and duodenum. It is mainly

responsible for increasing gastric motility, mucosal growth of the stomach, and

secretion of hydrochloric acid (HCl) (Prosapio et al., 2020). CCK also plays a very key

role in the regulation of digestive processes. It inhibits gastric emptying and secretion,

stimulates the intestinal and gall bladder contractions. It produces its actions in

mammals by acting on Cholecystokinin A and Cholecystokinin B receptors (Le et al.,

2019). Motilin is another hormone produced by endocrine cells of the mucosa of

duodenum and jejunum. Its receptors are also a part of the G protein-coupled receptors

family. It controls the interdigestive migrating contractions. Its levels cyclically

increase after every 90-120 min throughout the interdigestive fasting period. Motilin

release disappears after meal ingestion. These cyclical plasma motilin peaks results in

powerful peristaltic contractions originating from stomach and travels to small intestine

(Ohno et al., 2010).

Secretin is secreted in the duodenum by the S cells. Secretin is found to decrease gastric

contractions and increase gastric emptying time. Secretin receptors are G protein-

coupled receptors (Brandler et al., 2020). VIP is found in the digestive, reproductive,

respiratory and central and peripheral nervous system as neuroendocrine releasing

agent and as a neurotransmitter. VIP decreases the motor activity of GIT, thus

Page 39: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

36

decreasing the GI contractility (Bhagavan et al., 2011). GLP-1 is released in response

to meal from the gut to increases insulin secretion. GLP-1 is also involved in other GI

functions such as decrease in secretion of gastric acid, gastric wall tone and GI transit.

It has also shown to decline gastric and intestinal contractions. The mechanisms by

which GI motility is affected by GLP-1 are not completely understood (Brandler et al.,

2020).

1.4 Intrinsic and extrinsic innervation of gastrointestinal motility:

Various functions of the gut are regulated by intrinsic neurons of ENS and/or by

extrinsic parasympathetic (through pelvic and vagus nerves), sympathetic and sensory

neurons (Figure 1.10). Various studies have shown that ENS can act independently to

regulate motility reflexes even in extrinsically denervated intestine. However, CNS

performs a key role in regulating motility of esophagus and stomach while pathways of

extrinsic peripheral nerve coordinate with distant parts of GIT. The intestines have

substantial degree of autonomy to perform their functions without extrinsic neural

inputs (Uesaka et al., 2016).

Intestinal smooth muscles are innervated both by sympathetic and parasympathetic.

The parasympathetic system exerts its action through muscarinic receptors while α- and

β-adrenoceptors are used by sympathetic nervous system to mediate its inhibitory

effects.

1.4.1 Parasympathetic control of the gastrointestinal tract:

Preganglionic fibers are supplied by the parasympathetic system to ENS which synapse

with Auerbach’s and Meissner’s plexus. In these plexus, fibers from the cell bodies

reach the intestine to regulate motility and mucosal secretary cells. The parasympathetic

system, through ACh release is mainly involved in maintaining intestinal contractility

normal (Peddiready, 2011). Acetylchoine is the principal neurotransmitter at post-

ganglionic neurons. Vagus nerve is the key facilitator of the parasympathetic nervous

system. GIT is innervated not only by vagal sensory fibers but also by vagal motor

fibers One of the most vital functions of the vagus nerve is to bring information from

different organs including gut to brain (Uesaka et al., 2016).

Page 40: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

37

1.4.2 Sympathetic control of the gastrointestinal tract:

The sympathetic innervation of the gut arises from pre-vertebral ganglia of the

abdomen. Sympathetic post-ganglionic neurons which innervate the gastric region

reside within the celiac ganglion, while the neurons which innervate the intestine reside

in the pelvic ganglion (colon) or inferior mesenteric region (colon), the superior

mesenteric ganglion (small intestine) (Browning et al., 2011). Norepinephrine is the

neurotransmitter at the post ganglionic neurons of the sympathetic nervous system. The

sympathetic nervous system primarily exerts inhibitory effects on the GI motility, it

decreases the peristalsis and increases the contractility of sphincters (Uesaka et al.,

2016).

1.4.3 Role of gut microbiota in gastrointestinal motility:

Humans have enormous microbial ecosystem which is responsible for many GI

activities like digestion, metabolism, detoxification, prevention of bacterial

attachment to gut wall and control of GI homeostasis. High microbiotal concentration

are found in the stomach and it decreases progressively in the colon (Dieterich et al.,

2018). Due to certain differences in various regions of the GIT especially in the pH,

the type of bacteria and other microbes vary drastically. As stomach has strong acidic

environment, therefore, only few bacterial species grow and survive in it while other

gut regions have diverse bacterial species (Figure 1.11). The large intestine mainly

contains anaerobic and gram negative bacteria while small intestine contains aerobic

and gram positive bacteria (Ghoshal et al., 2012).

Recently, a lot of progress has been made in evaluating the role of gut microbiota in

the regulation of GI motility (Raja et al., 2018). Various studies have also shown its

involvement in the pathophysiology of different diseases like autoimmune, GI and

endocrine disorders. Due to its multiple effects on the metabolic and immune

functions, any alteration in the gut microbiota leads to various inflammatory and non-

inflammatory diseases of GIT (Fukui et al., 2018; Ghoshal et al., 2012).

The “faecal mircrobiotal transplantation” procedure is effective in severe drug resistant

cases of Clostridium difficile infection (Valdes et al., 2018). Treatment of disorders

related with GI motility by modifying the gut microbioata through antibiotics, diet etc

are presently under investigations (Raja et al., 2018).

Page 41: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

38

Figure 1.10: Intrinsic and Extrinsic Innervation of Gut motility (Uesaka et al.,

2016)

Page 42: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

39

Figure 1.11: Bacterial distribution in the GI tract (Knight, et al., 2019)

Page 43: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

40

1.5 Animal models for in vitro preparations:

1.5.1 History of experimental animals:

Humans have been using animals in research and education since they started to look

for ways to prevent and cure ailments. Animals have played an essential role in

scientific progression throughout history. The Greek scientist, Aristotle used animals

in his research, primarily to get a deeper understanding of these living beings (Anderson

et al., 2017). Animal oriented models have been crucial instruments since the early ages

of scientific discovery. Presently they are essential in any medical and biological

studies, helping us perceive and understand the functions of genes, the causes and

pathophysiology behind different diseases as well as drugs, their efficacies and adverse

effects. The entire genome of the organism have been sequenced, authenticating the

presence of different genetic similarities between these organisms and humans (Badyal

et al., 2014).

Most of present day's drug discoveries were possible because of the use of animals in

research as are introduced in therapeutics after experimental evaluation (Badyal et al.,

2014). The use of animals made the greatest discoveries in the 19th and 20th centuries.

Over the last century, every Nobel Prize for medical research has been dependent on

animal research. Research involving use of horses won the first Nobel Prize in 1901 in

medicine which was for serum therapy. The most recent 2012 Nobel laureates also used

animals. Experiments on animals helped in rapid progress in clinical medicine.

Discovery of Penicillin by Alexander Fleming was a wonderful example of using rats

as experimental animals in 1941, which transform the treatment of bacterial infections.

Insulin was also first isolated from dogs in 1922, Domagk and Selman Waksman used

chickens in 1939 and 1952 respectively to discover antibacterial activity of prontosil

and streptomycin (Badyal et al., 2014; Sinoussi et al., 2015).

1.5.2 Purpose of animal models:

A variety of diseases affect both humans and animals, thus animals can be used for the

study of human diseases. Combining the scientific knowledge with basic biological

studies guide us towards the better perception of function of living beings and we use

this information for the betterment of humans as well as animals. Animal models are

also preferred because of their easy availability, large reserves and feasible handling.

Page 44: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

41

It must be predicted within a reasonable limit that studies involving animal models

will contribute to the information which will result in protection and enhancement of

welfare and health of humans or animals. It is important to recognize the

experimental protocols of animal model in order to delineate a research study.

Majority of the cellular processes in animals are similar to those in humans in regard

that they carry out and execute many vital functions like breathing, digestion,

movement, sight, hearing and reproduction in the same as humans (Esteves et al.,

2018).

The primary goal of developing animal models for research is to create an

experimental system in which the conditions occurring in humans are phenocopied

as accurately as possible in the laboratory animal (Esteves et al., 2018). Due to vast

anatomical and physiological parallelism present between animals and humans,

especially mammals, the researchers were instigated to explore the properties and

characteristics of therapeutic regimens in animal oriented models prior to their

application in the human population. (Sinoussi et al., 2015).

During the studies of the animal species a lot of vital information was obtained which

could not have been used without the use of these models. Moreover the society

considers the usage of a newly developed drug or surgical procedure on humans as

immoral due to its probability that it may lead to more harmful effects as compared to

beneficial effects. This is one of the main reasons that these effects are analyzed on

animals, prior to its administration in humans. These animal trials have led to the

discovery of life saving medications for humans as well as animals (Badyal et al.,

2014).

1.5.3 Animal models for GI disorders:

The use of animal models for GI diseases is of great significance because of the

limitations of evaluating human GI diseases in clinical setting. They are used very

frequently. They have been used to investigate the pathogenesis of GI related diseases,

to find the mechanism, actions and effectiveness of different drugs in various GI

disorders. For studying human diseases, rodents are most frequently used animal

models because they are economical, easily available and reproduces rapidly (Ziegler

et al., 2016).

Page 45: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

42

1.5.3.1 In vitro animal models for GI motility:

Newly developed drugs need trials due to the fact that the developers must compute

and assess the favourable and unfavourable effects of a compound on an organism.

Primarily, the drug is tested in vitro using isolated organs and tissues, but it must also

be assessed using a suitable animal model, prior to performing clinical based trials in

humans (Jodat et al., 2018). In vitro studies help enable us to develop new technique

and treatment rapidly (Dias et al., 2018). In vitro assays for toxicological studies is the

refinement and replacement of animal testing in order to minimize the use of traditional

animal based toxicology studies. In the recent decades, in vitro methods have been

routinely used as these can be correlated with the in vivo studies and can help in

understanding a specific in vivo response in any given species (Srivastava et al., 2018).

Many in vitro preparations like (i) Stomach of mice, rat and hamster (ii) Guinea pig and

rat’s ileum, (iii) Rabbits’ ileum, duodenum and jejunum are commonly used to study

motility patterns and responses of intestinal smooth muscle. Rabbit and rat intestines

are routinely used in experiments relating motility. Pieces of animals intestine continues

to give responses for many hours when kept in a suitable solution. Usually they are

placed in Tyrode's solution through which air is passed. Rabbit’s intestine shows

regular pendular movements i.e., continuous relaxation and contraction (Pediready,

2011).

1.5.3.2 Rabbits:

Rabbits are small sized mammals belonging to Leporidae family, which is classified in

order Lagomorpha, based in many parts of the world. (Nowland et al., 2015). They

mainly populate the forests, woods, meadows and grass lands. They are most frequently

utilized in animal oriented studies. The reason that they are used so frequently in animal

trials is because they are smaller in size and easier to handle (Mapara et al., 2012). They

are parallel in physiology to humans. The frequency of their use in research studies is

gaining a popularity not only because of their docile nature but also because of the

financial ease to populate and conserve them as compare to larger animals (Esteves et

al., 2018).

Amongst various strains of rabbits, New Zealand white (NZW) strains of rabbits are

most frequently utilized in research trials because of their more docile nature and fewer

Page 46: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

43

health related issues as compare to other species. They also have much more genetic

similarities to humans. They are utilized as models for ocular ailments as well as

disorders of skin, immune, cardiovascular and respiratory system and in vivo and in

vitro models of GI disorders (Esteves et al., 2018).

For the evaluation of effects (preferably spasmolytic) of drug/chemical/plant on

pendular movements, jejunum is usually preferred because of its strong spontaneous

contractions. Usually there is no need to wash rabbit’s jejunum most of the times as it

is relatively wider and its spontaneous contractions causes it to clear itself (Peddiready,

2011).

1.5.3.3 Rats:

Rats were the first animal species to be specifically raised for the experimental studies.

They contribute a major bulk to the percentage of the animal models used for different

biomedical researches. Around 80-90% of the animals used for biomedical researches

are mice and rats. Therefore, they are well characterized in a number of ways. The use

of laboratory mouse for experimental purposes is significantly increased over the last

few years (Hedrich, 2012).

Laboratory rats are similar to humans in terms of physiology, anatomy and genetics,

therefore, they are frequently used animal models for various studies. Humans, mice

and rats have almost 30,000 genes, about 95% are shared by these three species. Their

smaller size, shorter gestational period and rapid growth are other advantages which

make them ideal to be used as animal models (Bryda, 2013).

The Wistar, an Albino outbred rat developed in the Wistar institute in 1906 for use in

biomedical studies, is the first species of the rat (Rattus norvegicus) to be domesticated

to serve as model organism at a time when laboratories use the common house mouse

(Mus musculus) (Sengupta, 2013).

Rats are studied as experimental models for cancer (Szpirer, 2010), cardiovascular

disease (including hypertension and cardiac failure) (Riehle et al., 2019), obesity and

diabetes (Kanasaki et al., 2010), neurodegenerative diseases (Parkinson’s disease)

(Welchko et al., 2012), GI disorders (Werawatganon, 2014), inflammatory

(Ghattamaneni et al., 2019) and autoimmune diseases (Castañeda-Lopez et al., 2017).

Page 47: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

44

On the other hand rats are also preferred in certain experiments as they breed rapidly

and are easy to handle because of their small size. Rat is one of the most used animal

model for GI diseases because in comparison to humans, rats have similar anatomic

structure of body organs (Vdoviakova et al., 2015). As ileum has very weak

spontaneous contractions, therefore, usually it is used for evaluation of spasmogenic

activity (Peddiready, 2011).

1.5.3.4 Advantages of in vitro techniques:

In vitro techniques include isolation of one or multiple pieces of required tissue from a

freshly sacrificed animal and keeping the tissue(s) alive in an appropriate solution at

required temperature and pH. The purpose of in vitro technique is to evaluate the

biological response(s) of the experimental drug.

In vitro techniques have certain advantages over in vivo techniques like:

i. The biological responses of the new experimental drugs can be easily verified

on the isolated tissue.

ii. Usually less dose of the experimental drug is required to evaluate the desired

response.

iii. Multiple tissues can be obtained from single animal.

iv. The response of the experimental drug can be immediately and directly tested

as the drug does not undergo the processes of absorption, distribution,

metabolism and excretion as it goes through in vivo techniques. (Peddiready,

2011).

1.6 Current gastro-prokinetic agents:

Prokinetic are drugs which increases gastrointestinal motility by augmenting the

number of contractions in the small intestine or making them powerful, but without

disturbing their rhythm (Litou et al., 2019). Prokinetics also rectify gastric

dysrhythmias, and encourage the flow of luminal contents in the forward direction.

Usually these agents are given at least 30 min prior to meals to have more beneficial

effects (Waseem et al., 2009).

Various prokinetic agents are already in use for these conditions but they are associated

with a lot of adverse effects e.g.

Page 48: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

45

1.6.1 Cholinergic agonists:

Cholinergic stimulants, the original motility inducing agents, stimulates muscarinic

receptors on smooth muscle cells. Currently anti-cholinesterase have been used as well

to a limited extent. Evidence for their efficacy is inconsistent in motility disorder.

However, Bethanecol had to be used for treatment of gastroperesis, reflex and

postoperative paralytic ileus but its effectiveness in these diseases are now diminishing

with the discovery of newer agent. Neostigmine is clinically used for Ogilvie’s

syndrome and Pyridostigmine clinically used for pseudo obstruction, but its use for

these indications are now disappearing with the introduction of newer agents.

Neostigmine and pyridostigmine continue to be employed in clinical practice for

Ogilvie’s syndrome and pseudo-obstruction respectively (Quengley, 2015).

These drugs give good prokinetic effect but they also have multiple muscarinic as well

as nicotinic side effects, decreased heart rate, hypotension or even cardiac arrest,

flushing and precipitation of bronchial asthma attack may also occur. They also cause

an increase in glandular secretions like saliva, sweat, tears, gastric and tracheobronchial

gland secretions. It may also cause stimulation of ganglia by anticholinesterases

through the muscarinic receptors after administration of anticholinesterase, ACh may

result in muscle twitching and fasciculation. Due to repetitive firing, higher doses of

these anticholinesterases drug may completely block the transmission of impulses in

neuromuscular junction causing weakness and paralysis of muscle. (Quengley, 2015).

1.6.2 Serotonergic agonists:

Cisapride and tegaserod are the two serotonin agonists which were used as prokinetic

agents but later on withdrawn from the market because of serious adverse effects.

Cisapride was observed to increase the pressure in lower esophageal sphincter and also

enhance gastric emptying. Cisapride was discontinued from market due to its serious

cardiac related side effects, which included arrhythmias involving prolongation of QT

interval (Quengley, 2015).

Intestinal contractility is increased by serotonin agonists. Prucalopride, another

serotonin agonist, with high affinity and selectivity for 5-HT4 receptors. It promotes

intestinal motility. Its high affinity for the 5-HT4 receptor and low affinity for the

hERG-K+ channel clarifies why it is not arrhythmogenic. As a prokinetic agent

Page 49: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

46

prucalopride promotes intestinal motility and transit. It is effective in improving bowel

function and reducing constipation-related symptoms.

Headache, abdominal cramps, nausea, diarrhea, bloating, fatigue and dizziness are the

common adverse effects associated with its use. 5-HT4 agonist improves GIT motility,

but ventricular arrhythmias and life threatening cardiovascular conditions can be

induced by these drugs as they effect potassium ion transport in cardiac muscle (Kamel

et al., 2015).

1.6.3 Dopamine antagonists:

Metoclopramide and Domperidone, dopamine antagonists, are primarily effective in

the enhancement of gut motility. They have shown desirable effects in gastroesophageal

reflux disease (GERD), gastroparesis and dyspepsia. Previously metoclopramide was

widely used as prokinetic agent with both central and peripheral effects while

domperidone did not have CNS effects as it does not cross the blood-brain barrier and

mainly acts through peripheral (DA2) receptors (Quengley, 2015).

Metoclopramide, being a prokinetic agent, has been used to treat GERD by improving

muscle action in the GIT. It shows variable effects on motility disorders.

Metoclopramide elevates the contractile ability of esophagus and fundus of stomach. It

also increases the pressure in lower esophageal sphincter and enhances the coordination

between antrum, pylorus and duodenum. Its use for longer period of time has been

convoluted by its adverse effects on CNS as well as its tolerance, in patient towards it.

Around 25% of patients taking metoclopramide have been recorded to have

experienced its side effects, most frequently among these are extra pyramidal

manifestations such as neuroleptic syndrome and tardive dyskinesia. Due to these

adverse effects, its use is mainly limited to gastrointestinal disorders and only for a

short duration of time (Kamel et al., 2015). They also frequently cause sedation and

diarrhea (Lau et al., 2016). Prolongation of the corrected QT interval is the most

important adverse effect of domperidone (Morris et al., 2016).

Some of the mentioned adverse effects are irreversible and resulted in fixation of “black

box” to its package in the US. Metoclopramide and domperidone both may increase

the serum prolactin levels leading to galactorrhea and gynecomastia (Quengley, 2015).

Page 50: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

47

1.6.4 Erythromycin:

Erythromycin (ERY), a macrolide antibiotic, is found to be motilin agonist (Deloose et

al., 2016). Motilin, a polypeptide hormone found mainly in the stomach distally and

duodenum, enhances the pressure in lower esophageal sphincter and is responsible for

stimulating the migrating motor complex (MMC) in the antral part of the stomach. The

intravenous form of motilin is the most powerful stimulator of solid and liquid gastric

emptying. ERY increases the contractile ability of antrum, stimulates premature MMC

phase III activity and prompt gastric emptying by binding to Motilin receptors. ERY

also enhances gastric motility in patients who have undergone vagotomy and

antrectomy, possibly this may be because of its stimulatory effects on the fundus of the

stomach (Waseem, et al., 2009).

In a review by Camilleri showed that ERY is most functional in acute gastroparesis

(Quengley, 2015). It is used as a prokinetic agent in post-operative ileus. It is also used

in the treatment of various clinical condition such as skin infections ,respiratory

infections (i.e community acquired pneumonia, Legionnaires disease), rheumatic

fever, intestinal amebiasis prophylaxis, prophylaxis of neonatal conjunctivitis and

chlamydia, syphilis and pelvic inflammatory disease (PID) (Kato et al., 2019; Marchant

et al., 2018). It is given to pregnant women for the prevention of Group B streptococcal

infection in the newborn (Gerber et al., 2017).

ERY have been noted to lead to certain adverse effects which include rash, allergic

reaction reversible deafness and decrease in appetite. It also increases the QT interval

and causes torsades de pointes. It is an enzyme inhibitor mainly inhibiting cytochrome

P450 and carries the risk of interaction with wide variety of medications. This leads to

an increase in the concentration of those drugs that are metabolized via the cytochrome

P450 system leading to the toxicity of those drugs (Akiyoshi et al., 2013). ERY may

also lead to cholestatic liver injury. In Sweden, ERY was the second most commonly

reported antibiotic to have caused drug induced injury. Cholestatic injury was reported

in 69% of cases among a collection of case reports. (Padda et al., 2011). In addition,

tachyphylaxis can also develop in patients on long term ERY therapy. This is due to the

down regulation of motilin receptors to which erythromycin binds. This can occur

within the first few days of starting therapy (Richards et al., 2009).

Page 51: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

48

1.6.5 Miscellaneous drugs:

Other drugs are Mitemcinal (Tilman et al., 2016), Atilmotin (Korimilli,et al., 2010),

Ghrelin (Sallam et al., 2010), Cholecystokinin receptor antagonists, loxiglumide

(Romero et al., 2014), Camicinal (Barshop et al., 2015).

There are a number of prokinetic agents available, among which most are associated

with a lot of adverse effects. So, there is always a search for new agent which is much

safer and effective. Azithromycin is one of the antibiotics which is prescribed

frequently and is relatively free from various side effects.

1.7 Azithromycin:

Azithromycin (AZM) is a broad spectrum, semi synthetic antibiotic which belongs to a

group of drugs known as macrolides (Rao et al., 2014; Lu et al., 2006; Martinez et al.,

2015). It was synthesized in early 1980s, it is still under investigation for the treatment

of various diseases (Jelic et al., 2016; Kagkelaris et al., 2018).

1.7.1 Source:

AZM is produced by Streptomyces species (gram-positive aerobic bacteria) (Jelic et al.,

2016).

1.7.2 Structure:

AZM is a part of the Azalide subclass of macrolides. It is obtained from ERY, a methyl-

substituted nitrogen atom is incorporated instead of carbonyl group into the lactone

ring, which makes the lactone ring 15-membered (Figure 1.12 & 1.13) (Bakheit et al.,

2014). In this way AZM not only improves its spectrum of activity than ERY but also

increases its tolerability (Davidson, 2019).

AZM has the chemical name (2R,3S,4R,5R,8R,10R,11R,12S,13S,14R)-13-[(2,6-

dideoxy-3-C-methyl-3-O-methyl-α-L-ribo-hexopyranosyl)oxy]-2-ethyl-3,4,10-trihydr

oxy-,5,6,8,10,12,14-heptamethyl-11-[[3,4,6-trideoxy-3-dimethyl amino) -β-D-xylo-

hexopyranosyl]oxy]-1-oxa-6-azacyclopentadecan -15-one. Its molecular formula is

C38H72N2O12, and its molecular weight is 749.0g/mol. Azithromycin, as the dihydrate,

is a white crystalline powder with a molecular formula of C38H72N2O12•2H2O and a

molecular weight of 785.0g/mol (Adeli, 2016; Timoumi et al., 2014).

Page 52: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

49

Figure 1.12: Chemical structure of Azithromycin (Assi et al., 2017)

Page 53: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

50

Figure 1.13: 3D structure of Azithromycin (John, 2017)

Page 54: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

51

1.7.3 History:

Azithromycin is obtained from Erythromycin, the first 14-membered macrolide,

isolated from the actinomycete Streptomyces erythreus (Saccharopolyspora

erythraea). It has been in human use since 1952. Its antimicrobial spectrum is similar

to that of penicillin. It is widely used for penicillin allergic patients. Depending on the

antibiotic concentration used and on the type of microorganism, it exerts bacteriostatic

and bactericidal properties (Jelic et al., 2016).

AZM (9a-methyl-9-deoxo-9-dihydro-9a-aza-9a-homoerythromycin), the first 15-

membered macrolide antibiotic in the market, consists of nitrogen atom inserted into

the macrocyclic ring. AZM was discovered in 1980 by a team of researchers at PLIVA

Laboratories in Croatia, Yugoslavia. This team comprised of Gorjana Radobolja-

Lazarevski, Gabrijela Kobrehel, and Zrinka Tamburasev, led by Dr. Slobodan Dokic.

After twenty years, in 2000, they were given the medal of highest honor, “Heroes of

Chemistry”, awarded by the American Chemical Society for their remarkable

contribution to chemistry (Jelic et al., 2016).

In 1986, a licensing agreement was signed by the PLIVA and Pfizer (pharmaceutical

company), which gave Pfizer exclusive rights to sale AZM in the United States and

Western Europe. In 1988, PLIVA marketed AZM in Central and Eastern Europe. In

1991, Pfizer also launched AZM in other markets under Pliva's license (Tomisic, 2011).

1.7.4 Administration:

AZM is commercially available for oral as well as parenteral (IV) administration.

Orally it is available in the form of tablets and suspension. It can be taken on empty

stomach or 2 hours after food. Intravenous AZM must be infused slowly over the time

of 60 min at least.

1.7.5 Mechanism of action:

AZM is characteristically bacteriostatic drug that inhibits bacterial growth by

interfering with bacterial protein synthesis. It inhibits translation of mRNA by binding

to bacterial ribosomes at 50S (Figure 1.14) (Bakhiet et al., 2014). As bacterial growth

is halted, replication is stopped thus bacteria cannot increase in number and eventually

these bacteria die or are cleared by the immune system.

Page 55: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

52

Figure 1.14: Mechanism of action of Azithromycin

Page 56: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

53

1.7.6 Half-life and tissue penetration:

AZM is stable to acids in the stomach therefore, can be taken via oral route without

affecting its absorption. Absorption of AZM is enhanced on an empty stomach. Oral

administration of a single dose of AZM (500mg) results in 37% bioavailability with a

maximum concentration in serum of 0.4 mg/L. In adults, when given orally, time to

peak concentration (Tmax) is 2.1 to 3.2 hours. AZM’s high concentration in

phagocytes, facilitates its transportation to the target. Large amount of drug is released

during active phagocytosis. AZM is highly lipid soluble and also due to ion trapping,

its concentration in tissues is 50 times much higher than in the plasma. (James, 2017).

Concentration of drug in the tissue exceed the minimum inhibitory concentration (MIC)

that inhibits 90% of probable pathogens (MIC90) after a single oral dose of 500mg

(Jelic et al., 2016).

Even 12 to 24 hours after a single dose of 500mg high intracellular levels of AZM are

also present in the tissues (mean >2 mg/l). It has a prolong half-life of 30-40 hours

(Kong et al., 2017; McMullan et al., 2015). AZM is generally well-tolerated and has

been proved to be effective even with a single dose (Tribble, 2017).

1.7.7 Clinical uses:

AZM is used to treat infections like community-acquired pneumonia, respiratory

infections, like pertussis and legionellosis, pelvic inflammatory disease, sexually

transmitted infections chancroid and granuloma inguinale, bacterial enteritis, cholera,

traveler’s diarrhea, enteric fever, skin infections, middle ear infections, tonsillitis and

sinusitis (McMullan et al., 2015; Bakheit et al., 2014).

AZM distribution has been a core component of the World Health Organization’s

(WHO) trachoma control program, with over 700 million doses of AZM distributed to

adults and children aged 6 months and older. AZM distribution dramatically reduces

the prevalence of the ocular strains of Chlamydia trachomatis that leads to trachoma

(Oldenburg et al., 2018).

AZM is a pregnancy category B drug and is usually considered safe to treat various

bacterial infections in pregnant and breastfeeding ladies (Ayse, 2013).

Page 57: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

54

1.7.8 Advantages of AZM over erythromycin:

Once daily dosing frequency, better efficacy and relatively less adverse effects of AZM

makes it superior in some clinical conditions (Kong et al., 2017). In contrast with ERY,

AZM is more acid stable, which simplifies administration around food (McMullan et

al., 2015). AZM is one of the most commonly used antibiotics by physicians. More

than 50 million prescriptions are produced in the United States per annum, generally

for respiratory tract and sexually transmitted infections (Jurinnk, 2014). AZM is slowly

metabolized and generate inactive metabolites. Unlike ERY it has no effect on the

cytochrome P450 (cyt P450), enzymes responsible for the metabolism of many drugs

and in the synthesis of cholesterol and steroids, so it does not interrelate with other

medicines being metabolized by cyt P450 (De Oleveria et al., 2016; Kagkelaris et al.,

2018).

1.7.9 Advantages of AZM over other antibiotics:

Macrolides are regarded as one of the safest antibiotics available. Different researches

proved the safety and efficacy of Azithromycin. They confirmed its enhanced efficacy

as compare to other antibiotics in bacterial infections even against resistant strains of

bacteria. (Davidson, 2019).

Prolonged use of other antibiotic treatment affects patient compliance and also

increases overall cost. The recommended duration of therapy for antibiotics such as

amoxicillin is 7–10 days. It is preferred to advise an antibiotic like AZM, for 3 days, to

make certain better compliance of patient and lessen the chances of development of

bacterial resistance. There is a strong evidence that short term courses may be much

appropriate treatment for acute otitis media and acute bacterial sinusitis. Short-term

treatment has been advised even for acute pharyngitis (Donde et al., 2014).

1.8 Pharmacological actions of AZM:

1.8.1 Antibacterial action:

AZM is generally considered to be bacteriostatic, but may become bactericidal at higher

doses and exerts a post antibiotic effect (Jelic et al., 2016; Matzneller et al., 2013).

AZM has good activity against many gram-negative and gram-positive bacteria

including Haemophilus influenzae, Neisseria gonorrhoeae, Mycoplasma pneumoniae

Page 58: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

55

Branhamella catarrhalis, Campylobacter, Moraxella catarrhalis and Legionella sp.,

Treponema pallidum and Mycobacterium avium complex. It also shows activity against

Proteus, Escherichia, Shigella, Enterobacter, Salmonella, Yersinia etc of

Enterobacteriaceae family (McMullan et al., 2015; Jelic et al., 2016).

1.8.2 Immunomodulatory action:

In addition to their antimicrobial properties, macrolides are also found to exert

immunomodulatory effects in vitro and animal data on it is also available. Macrolides

(AZM) also possess immunomodulatory properties, which were first noticed and

described in the 1950s just after their introduction. (Zimmermann et al., 2018; Li et al.,

2017; Bulska et al., 2014). The idea of using macrolides for immunomodulatory actions

was introduced in the 1970s (Zimmermman et al., 2018). Their significant effectiveness

in treating diffuse panbronchiolitis (DPB) has helped to extend their use as

immunomodulator to a number of clinical conditions (Kanoh et al., 2010).

Treatment with AZM in acute phase of inflammation resulted in reduction in pro-

inflammatory cytokines and stimulates resolution of chronic inflammation in the late

phases. Specifically, AZM is found to exert direct action on airway epithelial cells to

keep their function and decrease mucus secretion. These characteristics of AZM have

favored its use in the treatment of a number of chronic lung diseases including cystic

fibrosis (CF), non-CF bronchiectasis, diffuse panbronchiolitis (DPB), bronchiolitis

obliterans syndrome, chronic obstructive pulmonary disease (COPD) and asthma

(Cramer et al., 2017).

In order to explore the immunomodulatory action of AZM in vivo, the effect of AZM

on survival and production of cytokine was assessed in the (lipopolysaccharide) LPS

tolerance model which was categorized by a macrophage-M2 skewed response.

Tolerance induction by LPS priming was related with diminished serum concentrations

of pro-inflammatory cytokines, interleukin-12 (IL-12p40), tumour necrosis factor-

alpha (TNF-α) and chemokine ligand 5 (CCL5). Treatment with AZM resulted in

reduced serum TNFα and CCL5. The immunomodulatory action of AZM was

confirmed by increasing survival and dropping pro-inflammatory cytokine production

(Bosnar et al., 2013). In another study AZM was also found to decrease matrix

Page 59: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

56

metalloproteinase-9 (MMP-9), an enzyme which is involved in the degradation of

extracellular matrix and interleukin-8 (IL-8) (Zimmermann et al., 2018).

1.8.3 Anti-inflammatory action:

AZM has anti-inflammatory properties (Zimmermann et al., 2018; Li et al., 2017;

Bulska et al., 2014). Cytokines and chemokines, both have proinflammatory effects.

e.g., tumour necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6),

interleukin-8 (IL-8), interlekin-10 (IL-10) granulocyte-macrophage colony-stimulating

factor (GM-CSF), and interferon-gamma (IFN-γ) and anti-inflammatory e.g., (Kanoh

et al., 2010). AZM in a study inhibited Resiquimod (R848) induced TNF, interleukin-

1beta (IL-1β), IL-6, IL-8 and IL-10, and lipopolysaccharide (LPS) induced IL-1β and

IL-10 (Speer et al, 2018). Cigana and coworkers used three CF (IB3-1, 16HBE14o-

AS3, and 2CFSMEo−) and two isogenic non-CF (C38 and 16HBE14o-S1) airway

epithelial cell lines to check whether AZM decreased TNF-α mRNA and protein levels

or not. AZM decreased TNF-α mRNA levels and TNF-α secretion, to almost the levels

in the isogenic non-CF cells. NF-κB and specificity protein 1 (Sp1) DNA-binding

activities were also significantly reduced after treatment with AZM (Kanoh et al.,

2010).

1.9 Research studies of AZM related with smooth muscles

contractility:

AZM in addition to antibacterial, immunomodulatory and anti-inflammatory activities,

is also involved in the contractility of smooth muscles in various systems of the body

(El-Baki et al., 2015).

1.9.1 Effect of AZM on aortic strips:

A study was performed on AZM to find out its effect on the contractility of isolated

aortic spiral strips of rabbits. Aortic strips were treated with 0.3μg/ml norepinephrine

(NE) in order to produce contractions. At low doses AZM did not produce any

significant change in the contractility of aortic strips but when the dose was increased

(16μg/ml), it was able to significantly reduce the NE induced contractions. Amplitude

of the contractions was also reduced at higher doses (El-Baki et al., 2015).

Page 60: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

57

1.9.2 Effect of AZM on uterine smooth muscles:

In order to find the effects of AZM on the duration of pregnancy in women at risk of

labor, a study was conducted at Sainte Justine hospital, Canada. It was a retrospective

study in which those women who were admitted in obstetrics ward from January, 1st

2006 to August, 1st 2010 and received AZM were included in the study. Total 127

women were exposed to AZM during the study period and these were matched with

127 controls through pharmacy software and medical records. A time-to-event analysis

was done to compare gestational age at the time of recorded events (rupture of

membranes, delivery, or second intervention to prolong pregnancy). Results of the

study showed that there was no significant difference found between the two groups in

terms of prolongation of pregnancy and gestational age at event (Goyer et al., 2016).

1.9.3 Effect of AZM on respiratory smooth muscles:

Macrolide, in recent years there is increasing evidence about their favourable effects on

a number of chronic respiratory conditions. Historically, ERY and clarithromycin were

found to alleviate pulmonary worsening in diffuse panbronchiolitis. Long term

treatment with AZM prevented exacerbations and improved lung function in patients

colonized with pseudomonas aeruginosa. In noncystic fibrosis bronchiectasis, same

effects on prevention of exacerbations has been established. Similarly, long term use of

AZM also prevents bronchiolitis obliterans syndrome in patients undergoing lung

transplantation. Acute exacerbations are also prevented by AZM in chronic obstructive

pulmonary disease (COPD). Besides producing anti-inflammatory actions, AZM also

affects the contractility of airway smooth muscles (ASM) (Shteinberg et al., 2016).

Daenas et al., performed a study to find out the effect of AZM on ASMs. Isolated

tracheal strips precontracted with carbachol and KCl were suspended in an organ bath

and treated with different doses of AZM. AZM showed relaxant effect in a

concentration dependent manner. AZM was able to produce relaxant effect both in

calcium free solution and in the presence of verapamil, Ca2+ antagonist. The result

proposes that AZM has a concentration-dependent and direct relaxant effect on

precontracted tracheal strips that is not associated with inhibition of Ca2+ influx or

Ca2+ release from intracellular stores (Daenas et al., 2006).

Page 61: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

58

Wang et al conducted a study on AZM to find out its effect on ASMs. Mouse tracheal

rings (TRs) were used for this purpose. Mouse TRs were precontracted using

100μmol/L ACh, and the response to different doses of AZM was observed.

Precontractions induced by ACh were completely inhibited by AZM. The same

procedure was repeated with same protocols on human ASMs as well. AZM again was

found to relax the ASMs precontracted with ACh. The underlying mechanisms for

producing ASMs relaxation were also investigated in this study. AZM blocked L-type

voltage dependent calcium channels (LVDCC), Ca²⁺ permeant ion channels,

Muscarinic-2 (M2) receptors, and Transient receptor potential channel-3 (TRPC3)

and/or stromal interaction molecule channels (STIM/Orai), which reduced cytosolic

Ca²⁺ concentrations and led to relaxation of muscles (Wang et al., 2019).

Another study was performed by El-Baki and his colleagues to evaluate the actions of

AZM on the isolated tracheal strips. Contraction of tracheal strips were produced by

treating them with ACh followed by increasing cumulative doses of ACh without

washing the tissues. It was noted that AZM at lower dose (1μg/ml) did not produce any

changes while at higher dose (16μg/ml) it produced a dose dependent reductions on

ACh induced tracheal contractions (El-Baki et al., 2015).

Paicentini et al, evaluated the effects of AZM on the bronchial contractility

(hyperresponsiveness), lung function, and airway inflammation in asthmatic children.

Sixteen known asthmatic children were included in the study. They were divided in

two group. One group received AZM while the other group was given placebo. Lung

function and contractility were expressed as the dose response curve (DRC) of forced

expiratory volume in 1 second (FEV1) fall after hypertonic saline inhalation while for

inflammation neutrophils and lymphocytes level were monitored. No significant

change was found in lung function. In some of the AZM treated asthmatic children

there was reduction in bronchial contractility and airway neutrophilic infiltration

(Paicentini et al., 2007).

AZM is found to increase the functions of lung in lung transplant recipients (LTx). It

has been reported that gastroesophageal reflux (GER) is involved with the pathogenesis

of chronic rejection after LTx. AZM by increasing esophageal and gastric motility

improves glomerular filtration rate (GFR). Mertens et al determine the effect of AZM

on GER and gastric aspiration after LTx. Both strong and weakly acidic GER was

Page 62: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

59

measured with 24-h pH-impedance monitoring in 47 recipients of lung transplant in

which 12 patients were using AZM. In a separate group of 30 LTx patients gastric

aspiration was evaluated before and after taking AZM by measuring pepsin and bile

acid in bronchoalveolar lavage fluid (BALF). Results showed that recipients who were

using AZM had less number of GER events. Analysis of BALF showed less

concentration of bile acids with no significant effect on the level of pepsin. Overall

patients treated with AZM had less GER events and bile acids aspiration. These effects

might be because of improved esophageal motility and enhanced gastric emptying

(Mertens et al., 2009).

GERD is common in COPD and is usually associated with the development of acute

exacerbations. Different studies have proved the prokinetic effect of AZM, so may be

helpful in GERD. Therefore it was hypothesized by the authors that AZM may decrease

acute exacerbations in COPD by decreasing the episodes of GERD. Therefore, they

performed a retrospective review of the data collected in a prospective, randomized

controlled trial (RCT) of AZM for preventing COPD exacerbations. Participants were

broadly divided into two groups (i) those having a history of GERD (ii) those having

no history of GERD. Participants with GERD developed more frequent and rapid

exacerbations as compare to without GERD participants. AZM decreased exacerbations

both in GERD and without GERD participants, but more significantly in those without

GERD (Ramos et al., 2014).

AZM decreases risk of aspiration in recipients of lung transplant and improves

bronchiolitis obliterans syndrome (BOS). The authors assumed that AZM can improve

graft and overall survival much effectively in LTx patients with BOS who suffered from

aspiration of bile acids (BA) by inhibiting the progression of BOS associated with

aspiration. The objective was to compare FEV1 (% baseline), BOS progression and

overall survival in the recipients of LTx treated with AZM for BOS, both with and

without aspiration of BA. Those LTx recipients were included in the study who were

treated with AZM for BOS. Before starting the AZM treatment, samples of

bronchoalveolar lavage (BAL) were checked for the presence of BA and neutrophilia.

Short term effect of AZM on FEV1 and BAL neutrophilia was evaluated, 3 years follow

up was done regarding progression and survival of BOS in patients with and without

BA aspiration. BA in BAL were found in 19 out of 37 participants. Aspiration of BA

Page 63: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

60

resulted in deterioration of outcome, in terms of FEV1, BOS progression and survival.

Resulted showed that AZM is unlikely to protect against the allograft dysfunction for

longer period caused by GER and BA aspiration. Therefore, supplementary treatment

may be required targeting aspiration in the recipients of lung transplant (Merten et al.,

2011).

1.9.4 Effect of AZM on gastrointestinal smooth muscles:

1.9.4.1 AZM and esophageal smooth muscle contractility:

Jafari et al conducted a study to determine the effects of AZM on esophageal

hypomotility. Twenty six participants were included in the study who were suffering

from hypomotility disorders of esophagus. Half of them received AZM and half were

kept as control by just giving them placebo. At the end of the study AZM improved the

contraction of esophagus while the esophageal contractions of the placebo treated group

remained unchanged (Jafari et al., 2015).

1.9.4.2 Effect of AZM on gall bladder smooth muscles:

Gallstone formation is directly related with impaired gallbladder motility. Cisapride

and ERY are effective cholecystokinetic agents but they also exist arrhythmogenic

properties, therefore not used in routine for this effect. AZM is similar to ERY in

structure but does not possess significant drug-drug interaction as seen with ERY

(Ugwu et al., 2013).

Ugwu et al studied the effects of AZM on the contractility of gall bladder and compared

its effects with those of ERY. Twenty four males, looking apparently healthy were

included in the study. They were studied in pre-prandial and post prandial states. After

an overnight fast, one of the groups received 500mg AZM and the other group 500mg

ERY, 30 minutes before the study. Length, width and height of the gall bladder were

noted in each participant just before the ingestion of a standardized liquid meal in order

to get the ellipsoid volume with real time sonography. After taking the liquid meal, the

gall bladder measurements were again` taken after every 5 min for 40 min and

gallbladder contraction index (GBCI) was calculated. Most of the times ERY proved

significantly higher GBCI values than AZM while AZM only at few points showed

Page 64: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

61

higher GBCI. Results are suggestive of gall bladder smooth muscle contractility

induced by AZM (Ugwu et al., 2013).

GBCIs were assessed in 24 healthy volunteers after an overnight fast by using Real-

time ultrasonography. AZM and placebo were given to participants of the two groups

respectively and GBCIs were obtained in supine position every 5 min for 40 min. AZM

significantly improved the motility of gallbladder in the first 5 to 10 min. This increase

was not sustained till the 35th min when maximum GBC1s for the placebo was noted

(Ugwu et al., 2013).

1.9.4.3 AZM and gastroparesis:

The treatment of gastroparesis (disease in which the stomach cannot empty food in a

normal way by itself) is directly related with the severity of the symptoms, but it usually

comprises dietary modifications, antiemetics and prokinetic medications. The

preliminary treatment for gastroparesis is a prokinetic agent. ERY showed very

significant effect on gastric emptying, therefore it is often used for this purpose. There

are certain limitations to ERY which include adverse reactions like QT interval

prolongation, GI disturbances, tachyphylaxis and CYP3A-associated drug interactions.

AZM, another macrolide, on the other hand has been shown less GI disturbances, less

incidence of QT interval prolongation, fewer drug interactions and a longer half-life.

Use of AZM may be useful in patients with gastric and small bowel dysmotility (Potter

et al., 2013).

Shakir et al., evaluated the effects of AZM on the antroduodenal motility and compared

it with the effects of ERY. A retrospective analysis of the manometric data of gastric

and small bowel activity of the patients who were given intravenous AZM was done.

The pressure profiles acquired during motility studies were compared to those of

patients who were treated with intravenous ERY during motility studies. Total duration

of effect, number of cycles per min, mean amplitude of contractions, and duration of

highest antral and duodenal contractions were compared for both the drugs. Intravenous

AZM produced migrating motor contractions (MMCs) in the stomach followed by

small intestinal contractions. The mean amplitude of stomach contractions were slightly

higher in patients treated with AZM. The mean amplitude of small intestinal MMCs,

frequency and duration of gastric and small intestinal contractions were almost same in

2 groups (Shakir et al., 2018).

Page 65: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

62

Larson et al., done a study to determine the effects of AZM on gastroparesis. They also

compared the effects of AZM with erythromycin (ERY). Patients already diagnosed

with gastroparesis were included in the study. They were divided into two groups. All

the medications affecting the gastric emptying were stopped before starting the study.

Patients were given a scrambled egg meal which was labelled with 18.5-37 MBq of

technetium-99m sulfur colloid followed by continuous imaging for 120 minutes, at 1

minute per frame. Gastric emptying half-time (t½) was noted (normal = 45-90 minutes).

If the stomach had not properly emptied at 75-80 minutes. Half of the patients were

given AZM while the other half was given ERY and a new post-treatment gastric

emptying t½ was noted. Both the drugs enhanced gastric emptying t½ with no

significant difference (p-value 0.30). The results of this study are suggestive of

prokinetic effect of AZM (Larson et al., 2010).

Sutera et al., studied the effects of AZM on gastric emptying time in a diabetic patient.

The patient was an 83-year-old woman with a fourty year history of type II diabetes.

She was admitted with malnutrition, persistent vomiting and severe constipation.

Different treatment regimens were given but were unsuccessful. She was given i/v

AZM (500mg/day). After getting treatment for 3 days, vomiting was settled and the

patient was able to pass normal stool, with obvious progress in her general conditions.

This study supports the prokinetic activity of the AZM (Sutera et al., 2008).

Goshal et al., did the comparison of AZM with ERY in terms of gastric emptying

acceleration in patients with gastroparesis. The authors established that AZM equally

accelerated the gastric emptying as ERY did. They further suggested that AZM may be

favored over ERY because of its less involvement in drug interactions (Cyt P450),

relatively less adverse effects and longer duration of action (Goshal et al., 2010).

Another study was performed by Reddy et al at Veterinary College and Research

Institute, Namakkal, to compare the effect of AZM with Neostigmine on GI motility in

cows with functional ileus. Fourty cows with ileus, during the study period were

presented to the institute, out of which 20 were selected. Cows were investigated for

the levels of hemoglobin (Hb), packed cell volume (PCV), total erythrocyte count

(TEC) and total leucocyte count (TLC). After collecting serum it was checked for

chloride, potassium, sodium, albumin, gamma glutamyl transferase (GGT), creatinine

kinase (CK), inorganic phosphorus and calcium. Trans abdominal and rectal ultrasound

Page 66: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

63

was also done by using 2 - 3.5 MHz transducer of MyLab Vet 40 ultrasound scanner.

After investigation the cows were divided into two groups i.e group 1 and group 2. Each

group included 10 cows. Group 1 was treated with neostigmine (0.02 mg/kg) while

animals of group 2 received AZM (1 mg/kg). All the cows were also given ringers

lactate (10 ml/kg), dextrose normal saline (10 ml/kg) and procaine penicillin (20,000

IU/kg) for three days. Efficacy of the drugs was evaluated on the basis of clinical

improvement in terms of passing dung, and resumption of water and food. Results

showed that cows treated with AZM passed dung earlier as compare to the cows who

received neostigmine (Reddy et al., 2018).

1.9.4.4 Gastric motility disorders:

Moshiree et al., performed a study to find the effects of AZM on the activity of gastric

antrum. Thirty patients were included in the study who were diagnosed to have prolong

gastric emptying t½. Before starting the study all the patients were advised not to take

any drug affecting the gastric motility at least 3 days earlier. Patients with a history of

psychiatric illness, diabetes, intestinal obstruction, eating disorders, malignancy and

allergic to macrolide antibiotics were excluded from the study. A six channel

manometric microtransducer catheter was inserted intranasally and advanced through

the pylorus into the duodenum to analyze the gastroduodenal activity. AZM was

found to stimulate gastric antral activity. AZM also increased the duration and mean

amplitude of contractions (Moshiree et al., 2010).

A similar study was conducted to evaluate the pharmacological actions of AZM on

isolated gastric fundal strips of experimental animals. Each gastric fundal strip after

mounting in an organ bath was treated with ACh to produce contractions. Then

different doses of AZM were added in a cumulative way and response to each dose

was recorded. AZM was found to increase the ACh induced contractions in a dose

dependent manner (El-Baki et al., 2015).

Blondeau et al., performed a study to check the effects of AZM on GERD. AZM was

given to one group of patients while the other group was kept devoid of it. When the

reflux parameters were compared between the patients with and without AZM, patients

with AZM were found to have significantly lowered esophageal reflux exposure.

Page 67: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

64

However, the frequency of reflux was not affected but the proximal extent of the acid

was observed less in AZM treated patients (Blondeau et al., 2007).

Broad et al., in 2013 designed a study to confirm the involvement of motilin receptors

in affecting the GI motility by AZM. It was performed on human isolated stomach.

Human stomach segments were obtained from patients going through surgery for

cancer or obesity. The stomach segments were shifted to the research laboratories

within 2 hours after resection in Krebs' solution aerated with 5% CO2 and 95% O2. The

strips were then mounted in tissue bath. Human recombinant motilin receptors in CHO

cells were used for conducting motilin binding and calcium flux experiments. AZM

was found to displace motilin binding to motilin receptors in a concentration dependent

manner. AZM and motilin concentration-dependently initiated short-term increase in

intracellular [Ca2+] in cells expressing the motilin receptor. In human stomach, AZM

induced cholinergically mediated contractions. The results of this study justifies the

gastric prokinetic action of AZM (Broad et al., 2013).

1.9.4.5 AZM and intestinal contractility:

Certain studies have been conducted to evaluate the effects of AZM on the contractility

of intestinal smooth muscles. (El-Baki et al., 2015; Ugwu et al., 2013). Some of them

have proved that AZM significantly increases the contractility of intestinal smooth

muscles while the others showed controversy regarding its effect (Broad et al., 2013;

Nguyen, 2014). So still there is a confusion whether AZM increases or decreases the

intestinal contractility.

A study was carried out by El-Baki and his co-workers to determine the

pharmacological effects of AZM on different isolated smooth muscle preparations of

experimental animals. After recording the normal contractions of rabbit’s jejunum,

AZM was given in different doses ranging from 1-16µg/ml. It was found out that AZM

increased the amplitude of jejunal contractions in all doses in a dose-dependent manner

(El-Baki et al., 2015).

Another study was conducted by Chini and his colleagues to determine the effect of

AZM on the small intestinal motility and to compare it with the effects of ERY. Twenty

one patients were included in the study. The frequency, amplitude and duration of small

intestinal contractions were recorded. Result of the study confirmed that AZM was

Page 68: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

65

found to stimulate gastro-duodenal motility. When the effects of AZM were compared

with those of ERY, AZM was found to produce gastro-duodenal contractions more

frequently (Chini et al., 2012). The results are in favour of prokinetic activity of AZM.

In another study, effects of AZM was assessed on GI motility in men. Eleven healthy

volunteers were included in a single blinded, placebo-controlled study. A 500mg dose

of AZM was given to them. Treatment with AZM resulted in a significant increase in

the GI motility as compared to placebo (Sifrim et al., 1994). Results were suggestive

of prokinetic activity of AZM.

In contrast to all the above mentioned studies which showed the prokinetic effects of

AZM, Chiragh et al., performed a study in vitro on rabbits duodenum. The results

showed that the prokinetic effect of AZM are not well-sustained. Thus, showing AZM

not to be a beneficial therapeutic agent to produce prokinetic actions (Chiragh et al.,

2006).

Not too much work is done to investigate the mechanisms through which AZM is going

to affect the intestinal contractility. In this regard El-Baki et al performed a study to

explore the mechanisms of AZM affecting intestinal contractility but they missed many

important regulators of intestinal motility which perform a key role in its regulation.

In a study El-Baki et al evaluated the effects of AZM on isolated colon of rats. AZM in

different doses (1-16μg/ml) produced an increase in the amplitude of contractions.

Doses were increased in a cumulative way and increase in response was noted in dose

dependent manner. They further investigated the involvement of muscarinic,

serotonergic and histaminergic receptors in the colonic contractions induced by AZM.

Isolated colon was first given histamine antagonist, then without washing the tissue

different doses of AZM were given. Same procedure was repeated for serotonin and

histamine receptor blocker. It was noted that blockade of muscarinic receptors was

associated with the prevention of the stimulant action of AZM on colonic contractions.

On the other hand, AZM in the presence of serotonergic and histaminergic receptors

blockade was still able to produce jejunal stimulation. The result of this study confirmed

that AZM exerts its effect on the intestinal motility via muscarinic receptors. Various

other receptors which are also involved in the intestinal motility are not explored in this

study, thus making it very limited (El-Baki et al., 2015).

Page 69: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

66

Rationale:

As far as there are controversial reports regarding the effect of AZM on the contractility

of intestinal smooth muscles and the mechanisms involved in it are still unrevealed.

Therefore, the present study is designed to find out the effect of AZM on the contraction

of intestinal smooth muscles and to know about its possible mechanisms through which

it affects the contractility. Knowing the exact mechanism(s) of action of AZM will help

the researchers in many ways for instance i) By finding the effect of AZM on intestinal

smooth muscles, new indications, contraindication and adverse effects can be identified

ii) It will help to decrease the use of multiple drugs in the same patient where antibiotics

for bacterial infections and other drugs to produce change in GI motility is required iii)

It will help to predict the possible drug interactions of azithromycin with other drugs.

Objectives:

1 To determine the effects of Azithromycin on the contractility of intestinal smooth

muscles in vitro model of rabbits and rats using Power Lab system.

2 To investigate the mechanisms of Azithromycin affecting the intestinal contractility

through receptors and voltage gated ion channels by using Power Lab system.

Page 70: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

67

2 MATERIAL AND METHODS

2.1 Animals:

Adult healthy New Zealand rabbits were purchased from the local market. Body weight

of the rabbits varied from 2-2.5kg. Wistar rats were purchased from the National

Institute of Health Laboratory, Islamabad. Body weight of the rats varied from 180-

230g. Both male and female rabbits and rats were included in the study. All the rabbits

and rats were carefully evaluated to make sure that none of them are pregnant or

apparently in a diseased state.

Animals were kept in the animal house of Institute of Basic Medical Sciences (IBMS),

Khyber Medical University, Peshawar under hygienic conditions. The animals were

transported from National Institute of Health Laboratory, to the animal house of IBMS

in laboratory iron cages, where the temperature was maintained at 20-24°C. The animals

were exposed to 12 hours light and 12 hours dark cycle (Guo et al., 2016). They were

given free access to drink water and to take food; rabbits were mostly given fresh grass

and carrots while rats were given rat chow. They received humane care under standard

protocols outlined in the "Animals Bye-Laws 2008 of the University of Malakand

(Scientific Procedures Issue- 1) (Ali et al., 2009).

The animals were acclimatized for one week, before performing the experiment. They

were kept in fasting condition for 24 hours prior to experiment. However, they were

allowed to drink water freely (Naseri et al., 2008). The protocols were approved for this

study by the Ethical Committee of Khyber Medical University, Peshawar via No.

Dir/KMU-EB/MU/000745.

2.2 Chemicals and drugs:

All the chemicals and drugs used in the experiments were of analytical grade. The

chemicals and drugs used in the experiment were azithromycin dihydrate,

acetylcholine, atropine sulphate, loratadine, ondansetron, metoclopramide, verapamil,

propranolol and amiodarone (Sigma-Aldrich chemicals, St. Louis, U.S or E. Merck

grade). Fresh stock solutions for all the chemicals were prepared on the day of

experiments.

Page 71: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

2-Material and Methods

68

2.3 Preparation of isolated tissues

2.3.1 Rabbits’ jejunal and rats’ ileal preparations

The abdomen of each rabbit and rat after scarifying was opened through a midline

incision. The abdominal viscera on the ventral abdominal wall was exposed, 1-1.5 cm

pieces of jejunum from each rabbit and the same size pieces of ileum from each rat were

isolated. All the tissues were handled very gently in order to keep the morphology and

physiology of the tissues intact. The isolated tissues were then quickly placed in a petri

dish (Nath et al., 2016; Ali et al., 2011). The petri dish contained Tyrode’s solution

which had the following composition; NaCl 8, KCl 0.2, MgCl2 0.1, CaCl2 0.2, NaHCO3

1, NaH2PO4 0.05, Glucose 1 g/L. All the chemicals were dissolved in distilled water

(Chiragh et al, 2006).

The petri dish was persistently aerated with carbogen gas (a mixture of 95% O2 and 5%

CO2) in order to keep the tissues viable (Ghayur et al., 2012). Mesentery was detached

from each tissue and each tissue was also properly cleaned from fecal content where

found. Jejunal and ileal preparations were suspended individually in a 15 milliliter (ml)

tissue bath containing Tyrode’s solution. The temperature of the tissue bath was kept at

37±1°C and aerated with carbogen gas persistently. One end of each preparation was

tied to the hook and the other was attached by a thread to the transducer, it was made

sure that lumen of the jejunum and ileum was not closed by the threads (Janbaz et al.,

2015; Chiragh et al., 2006). The transducers were linked to an amplifier to augment the

magnitude of contractions; these in turn were interpreted by a data acquisition system

to get the final results on computer system (Yasin et al., 2012).

Each tissue was kept undisturbed and allowed to equilibrate for a period of 30 min

before adding any drug. The bath fluid was replaced subsequently with normal Tyrode’s

solution before starting the experiment. (Chda et al., 2016; Janbaz et al., 2015). Those

jejunal preparations in which the spontaneous contractions were found were included

in the study while the preparations in which the spontaneous contractions were either

absent or improper were removed from the study protocols. The excluded tissues were

then replaced by the other healthy tissues having proper spontaneous contractions. To

prevent desensitization, single tissue was used once only for one concentration-

response curve (Figure 2.1).

Page 72: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

2-Material and Methods

69

Figure 2.1: Small and Large intestines of a rabbit

Page 73: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

2-Material and Methods

70

Figure 2.2: Isolated rabbit’s jejunal preparation

Page 74: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

2-Material and Methods

71

2.4 Effects of drugs on rabbit’s jejunal and rat’s ileal preparations

2.4.1 Effect of AZM on rabbit’s jejunal and rat’s ileal preparations

Each rabbit’s jejunal and rat’s ileal preparation was suspended in an organ bath and

following stabilization for 30min, AZM was added cumulatively in doses of 0.01, 0.03,

0.1, 0.3, 1, 3, 5, 10, 15µM. The response to each dose was recorded for 2 min. The same

doses of AZM were considered as standard for the next series of experiments. The

contraction of isolated tissue preparations was expressed as percent of the control

response produced by ACh (3µM). Each rabbit’s jejunal and rat’s ileal preparation was

pre-treated with a validated dose of respective receptor and channel blockers. The dose

at which the receptor and channel blockers showed maximum decrease in the amplitude

of contractions was consider standard for pre-treating the relevant tissues, after which

the above mentioned doses of AZM were added. Contraction and relaxation of the

mounted tissues were detected by Force transducer (Model No: MLT 0210/A Pan Lab

S.I.) attached to Power Lab. (Model No: 4/25 T) ADInstruments, Australia. Data were

recorded at range 20 mv, Low pass 5Hz X 10 gain using input 1, rate 40/S. The

cumulative dose response was recorded in three rabbits and rats individually. Results

were described as percent response of control on y-axis while dose (µM) on x-axis.

2.4.2 Validation of curves:

Validation of curves was performed after following the procedure described in section

2.4.1, for different drugs used in the experiments in order to verify the desired response.

2.4.2.1 Acetylcholine:

Each tissue after stabilization was treated with 3 µM ACh which served as control. This

procedure was repeated on three different rabbits and rats. From each rabbit and rat, a

single tissue was taken and reading of each tissue was recorded. Results were described

as percent response on y-axis while dose (µM) on x-axis (Ali et al., 2011).

2.4.2.2 Atropine:

After stabilization, each rabbit’s jejunal and rat’s ileal preparation was given 0.01 µM

Atropine and response was noted. The concentration of atropine was gradually

increased without washing the tissue. The contractile response of tissues for each dose

Page 75: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

2-Material and Methods

72

was recorded. At a certain point no further decrease in tissue’s response was found

depite increasing the dose of atropine. It was considered as maximum response (Emax)

of Atropine. The same method was repeated on three different rabbits and rats. Results

were described as percent response on y-axis while dose (µM) on x-axis (Ali et al.,

2011).

2.4.2.3 Loratadine:

Each rabbit’s jejunal and rat’s ileal preparation after stabilization was treated with 0.01

µM Loratadine. After using Loratadine, the response of individual tissue was recorded.

The concentration of Loratadine was kept on increasing cumulatively and the response

for each dose was noted till there was no further decrease in tissue’s contractile response

despite adding more dose, It was considered as maximum response (Emax) of

Loratadine. The same procedure with same protocols was repeated on three different

rabbits and rats. Results were described as percent response on y-axis while dose (µM)

on x-axis (Ali et al., 2011; Faisal et al., 2018).

Loratadine belongs to the second generation anti-histamines. It was selected for this

study because it lacks various adverse effects including anti-muscarinic actions which

are usually associated with first generation anti-histamines. Therefore, using loratadine

will give the actual results as compared to first generation anti-histamines where the

result may be biased because of their anti-muscarinic effects (Church et al., 2013; Liu

et al., 2005).

2.4.2.4 Ondansetron:

After stabilization, each rabbit’s jejunal and rat’s ileal preparation was given 0.01 µM

Ondansetron and response of each tissue was noted for 2min. The concentration of

Ondansetron was gradually increased without washing the tissue. The contractile

response of tissues for each dose was recorded. At a certain stage no further decrease

in tissue’s response was found despite increasing the dose of Ondansetron, it was

considered as maximum response (Emax) of Ondansetron. The same method was

repeated on three different rabbits and rats and its response was recorded. Results were

described as percent response on y-axis while dose (µM) on x-axis (Ali et al., 2011;

Afzal et al., 2016).

Page 76: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

2-Material and Methods

73

2.4.2.5 Metoclopramide:

Each tissue after stabilization was treated with 0.01 µM Metoclopramide. After using

Metoclopramide, the response of individual tissue was recorded. The concentration of

Metoclopramide was kept on increasing cumulatively and the response for each dose

was noted. At a certain point no further increase in tissue’s contractile response was

found despite adding more dose, it was considered as maximum response (Emax) of

Metoclopramide. The same procedure with same protocols was repeated on three

different rabbits and rats. Results were described as percent response on y-axis while

dose (µM) on x-axis (Ali et al., 2011; Kamel et al., 2015).

2.4.2.6 Verapamil:

After stabilization, tissues were given 0.01 µM Verapamil and response of each tissue

was noted. The concentration of Verapamil was gradually increased without washing

the tissue. The contractile of tissues for each dose was recorded. At a certain stage no

further decrease in tissue’s response was found despite of increasing the dose of

Verapamil, it was considered as maximum response (Emax) of Verapamil. The same

method was repeated on three different rabbits and rats. Results were described as

percent response on y-axis while dose (µM) on x-axis (Ali et al., 2011; Ali et al., 2017).

2.4.2.7 Propranolol:

Each tissue after stabilization was treated with 0.01 µM Propranolol. After using

Propranolol, the response of individual tissue was recorded. The concentration of

Propranolol was kept on increasing cumulatively and the response for each dose was

noted till there was no further decrease in tissue’s contractile response despite of adding

more dose, it was considered as maximum response (Emax) of Propranolol. The same

procedure with same protocols was repeated on three different rabbits and rats. Results

were described as percent response on y-axis while dose (µM) on x-axis (Ali et al.,

2011; Ali et al., 2017).

2.4.2.8 Amiodarone:

After stabilization, each tissue was given 0.01 µM Amiodarone and response of each

tissue was noted. The concentration of Amiodarone was gradually increased without

Page 77: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

2-Material and Methods

74

washing the tissue. The contractile response of tissues for each dose was recorded. At

a certain stage no further decrease in tissue’s response was found despite increasing the

dose of amiodarone, it was considered as maximum response (Emax) of Amiodarone.

The same method was repeated on three different rabbits and rats. Results were

described as percent response on y-axis while dose (µM) on x-axis (Ali et al., 2011).

2.4.3 Effect of AZM in the presence of receptor and channel blockers:

2.4.3.1 Effect of AZM in the presence of Atropine:

To determine the effects of AZM for possible actions on muscarinic receptors, each

rabbit’s jejunal and rat’s ileal preparation was first allowed to get stabilized for 30 min.

Tyrode’s solution of the bath in which tissues were suspended was replaced by fresh

Tyrode’s solution before adding any drug. Each jejunal and ileal preparation was pre-

treated with atropine (0.3µM), a muscarinic receptor antagonist, for 30 minutes (Faisal

et al., 2018; Ehlert et al., 2012). Different cumulative doses of AZM were applied in

the same concentration as mentioned in the section 2.4.1 and responses were recorded

(each for 2 min). This procedure was repeated on three rabbits and rats. Results were

described as percent response of control on y-axis while dose (µM) on x-axis.

2.4.3.2 Effect of AZM in the presence of Loratadine:

For evaluation of possible actions of AZM on histamine receptors, each rabbit’s jejunal

and rat’s ileal tissue after stabilization was incubated with 0.3µM Loratadine, a

histamine receptor antagonist for 30 min. Different cumulative doses of AZM were

applied in the same concentration as mentioned in the section 2.4.1 and responses were

recorded (each for 2 min). This procedure was repeated three times on three rabbits and

rats. Results were described as percent response of control on y-axis while dose (µM)

on x-axis (Faisal et al., 2018).

2.4.3.3 Effect of AZM in the presence of Ondansetron:

In order to investigate the effects of AZM for possible actions on serotonin receptors,

each rabbit’s jejunal and rat’s ileal preparation was first stabilized. Tyrode’s solution

of the bath in which tissues were suspended was replaced by fresh Tyrode’s solution

before adding any drug. Each tissue was incubated with 0.3µM Ondansatron, a

serotonin receptor antagonist, for 30 min. Different cumulative doses of AZM were

Page 78: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

75

applied in the same concentration as mentioned in the section 2.4.1 and responses were

recorded (each for 2 min). This procedure was repeated three times on three rabbits and

rats. Results were described as percent response of control on y-axis while dose (µM)

on x-axis (Faisal et al., 2018).

2.4.3.4 Effect of AZM in the presence of Metoclopramide:

To explore the effects of AZM for possible actions on dopamine receptors, each rabbit’s

jejunal and rat’s ileal preparation was first allowed to get stabilized. Before adding any

drug, Tyrode’s solution of the bath was replaced by fresh Tyrode’s solution. Each tissue

was incubated with 10µM Metoclopramide, a dopamine receptor antagonist, for 30 min.

Different cumulative doses of AZM were applied in the same concentration as

mentioned in the section 2.4.1 and responses were recorded (each for 2 min). This

procedure was repeated three times on three rabbits and three rats. Results were

described as percent response of control on y-axis while dose (µM) on x-axis (Kamel

et al., 2015).

2.4.3.5 Effect of AZM in the presence of Verapamil:

For evaluation of possible actions of AZM on calcium channels, each rabbit’s jejunal

and rat’s ileal preparation after stabilization was treated with 0.3µM verapamil, a

calcium channel blocker, for 30 minutes. Different cumulative doses of AZM were

applied in the same concentration as mentioned in the section 2.4.1 and responses were

recorded (each for 2 min). This procedure was repeated on three rabbits and rats. Results

were described as percent response of control on y-axis while dose (µM) on x-axis

(Faisal et al., 2018).

2.4.3.6 Effect of AZM in the presence of Propranolol:

In order to investigate the effects of AZM for possible actions on sodium channels, each

tissue was first allowed to get stabilized. Tyrode’s solution of the bath in which tissues

were suspended was replaced by fresh Tyrode’s solution before adding any drug. Each

rabbit’s jejunal and rat’s ileal preparation was incubated with 0.3µM propranolol for 30

minutes to block the sodium channels. Different cumulative doses of AZM were applied

in the same concentration as mentioned in the section 2.4.1 and responses were recorded

(each for 2 min). This procedure was repeated on three rabbits and rats. Results were

Page 79: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

1-Introduction

76

described as percent response of control on y-axis while dose (µM) on x-axis (Faisal et

al., 2018).

2.4.3.7 Effect of AZM in the presence of Amiodarone:

To determine the effects of AZM for possible actions on potassium channels, each

rabbit’s jejunal and rat’s ileal preparation after stabilization was incubated with 3µM

Amiodarone, a potassium channel blocker, for 30 min. Different cumulative doses of

AZM were applied in the same concentration as mentioned in the section 2.4.1 and

responses were recorded (each for 2 min). This procedure was repeated on three rabbits

and rats. Results were described as percent response of control on y-axis while dose

(µM) on x-axis (Faisal et al., 2018).

2.4.3.8 Effect of AZM in the presence of Atropine, Ondansetron, Verapamil &

Propranolol:

Each rabbit’s jejunal and rat’s ileal preparation was pretreated with Atropine (0.3µM),

Ondansetron (0.3µM), Verapamil (0.3µM) and Propranolol (0.3µM) for 30 min.

Tissues were then treated with various increasing doses of propranolol until Emax was

achieved. Different cumulative doses of AZM were applied in the same concentration

as mentioned in the section 2.4.1 and responses were recorded (each for 2 min). This

procedure was repeated on three rabbits and rats. Results were described as percent

response of control on y-axis while dose (µM) on x-axis (Ali et al., 2017; Islami et al.,

2009).

2.5 Statistical Analysis:

Intestinal responses (%) of both rabbits’ jejunal and rats’ ileal preparations were plotted

as percent of ACh maximum concentration response using Graph Pad Prism version 6.

Similarly, in the presence and absence of each antagonists for possible involvement in

intestinal tissues, intestinal responses were expressed in tables. Their Emax in presence

of antagonist was compared with Emax of the tissues without respective antagonists by

using t-test. P-value ≤ 0.05 was considered significant.

Page 80: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

77

3 RESULTS

This study contained 80 animals in which 48 were rabbits and 32 were rats. AZM

produced the spasmogenic response in all the animals. The current study was an attempt

to find out the effects of AZM on intestinal contractility and to investigate the possible

mechanisms by which AZM is affecting it. Percent spasmogenic effects of AZM was

expressed as % of ACh maximum (3uM), which served as control.

3.1 Validation of curves:

Validation of curve was performed for various drugs to verify the desired response and

to know the dose at which the maximum response (Emax) was produced.

3.1.1 Atropine:

Atropine decreased the contractile response both in rabbits’ jejunal and rats’ ileal

preparations. Emax for atropine was 3.6±1.3 and 1.2±0.8 for rabbits’ jejunal and rats’

ileal preparations, respectively (Table 3.1, Figure 3.1a & 3.1b).

Table 3.1: Effect of Atropine (µM) in % of ACh on rabbits’ jejunal preparations and

rats’ ileal preparations (Mean±SD, n=3)

Concentrations

(µM)

Effect of atropine on rabbits’

jejunal preparation

Effect of atropine on rats’ ileal

preparation

0.01 16.8±1.8 4.9±1.1

0.03 12.9±2.1 4.6±1.3

0.1 8.7±1.4 2.8±1.1

0.3 3.6±1.3 1.2±0.8

1 3.6±1.3 1.2±0.8

3 3.6±1.3 1.2±0.8

5 3.6±1.3 1.2±0.8

10 3.6±1.3 1.2±0.8

15 3.6±1.3 1.2s±0.8

Page 81: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

78

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

20

Atropine (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.1a: Effect of Atropine on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 82: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

79

0.01 0.03 0.1 0.3 1 3 5 10 15

0

2

4

6

Atropine (M)

Resp

on

se i

n %

0f

Acety

lch

oli

ne (

3u

M)

Figure 3.1b: Effect of Atropine on spontaneous rats’ ileal preparations

(n=3; Mean±SD)

Page 83: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

80

3.1.2 Loratadine:

Loratadine decreased the contractile response both in rabbits’ jejunal and rats’ ileal

preparations. Emax for loratadine was 9.0±1.0 and 5.2±0.3 for rabbits’ jejunal and rats’

ileal preparations, respectively (Table 3.2, Figure 3.2a & 3.2b).

Table 3.2: Effect of Loratadine (µM) in % of ACh on rabbits’ jejunal preparations and

rats’ ileal preparations (Mean±SD, n=3)

Concentrations

(µM)

Effect of loratadine on rabbits’

jejunal preparation

Effect of loratadine on rats’ ileal

preparation

0.01 20.1±1.6 14.3±0.8

0.03 18.4±1.4 12.6±0.4

0.1 12.9±1.6 8.8±0.4

0.3 10.4±1.3 6.2±0.3

1 9.2±1.3 5.8±0.3

3 9.0±1.0 5.2±0.3

5 9.0±1.0 5.2±0.3

10 9.0±1.0 5.2±0.3

15 9.0±1.0 5.2±0.3

Page 84: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

81

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

20

25

Loratadine (M)

Resp

on

se i

n %

0f

Acety

lch

oli

ne (

3u

M)

Figure 3.2a: Effect of Loratadine on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 85: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

82

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

Loratadine (M)

Resp

on

se i

n %

0f

Acety

lch

oli

ne (

3u

M)

Figure 3.2b: Effect of Loratadine on spontaneous rats’ ileal preparations

(n=3; Mean±SD)

Page 86: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

83

3.1.3 Ondansetron:

Ondansetron decreased the contractile response both in rabbits’ jejunal and rats’ ileal

preparations. Emax for ondansetron was 8.6±1.3 and 4.4±0.3 for rabbits’ jejunal and

rats’ ileal preparations, respectively (Table 3.3, Figure 3.3a & 3.3b).

Table 3.3: Effect of Ondansetron (µM) in % of ACh on rabbits’ jejunal preparations and

rats’ ileal preparations (Mean±SD, n=3)

Concentrations

(µM)

Effect of ondansetron on rabbits’

jejunal preparation

Effect of ondansetron on rats’ ileal

preparation

0.01 18.2±1.6 10.3±0.8

0.03 16.4±1.4 7.3±1.2

0.1 12.7±1.6 5.8±1.4

0.3 8.6±1.3 4.4±0.3

1 8.6±1.3 4.4±0.3

3 8.6±1.3 4.4±0.3

5 8.6±1.3 4.4±0.3

10 8.6±1.3 4.4±0.3

15 8.6±1.3 4.4±0.3

Page 87: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

84

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

20

Ondansetron (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.3a: Effect of Ondansetron on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 88: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

85

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

Ondansetron (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.3b: Effect of Ondansetron on spontaneous rats’ ileal preparations

(n=3; Mean±SD)

Page 89: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

86

3.1.4 Metoclopramide:

Metoclopramide increased the contractile response both in rabbits’ jejunal and rats’

ileal preparations. Emax for metoclopramide was 66.4±1.8 and 58.4±1.6 for rabbits’

jejunal and rats’ ileal preparations, respectively (Table 3.4, Figure 3.4a & 3.4b).

Table 3.4: Effect of Metoclopramide (µM) in % of ACh on rabbits’ jejunal preparations and

rats’ ileal preparations (Mean±SD, n=3)

Concentrations (µM)

Effect of metoclopramide on rabbits’ jejunal preparation

Effect of metoclopramide on rats’ ileal preparation

0.01 18.9±1.2 2.9±0.3

0.03 26.9±0.6 8.6±1.4

0.1 32.5±0.9 16.8±0.8

0.3 45.5±1.1 28.2±1.8

1 48.6±1.3 38.4±1.3

3 56.4±0.8 44.6±1.4

5 58.4±1.6 50.2±1.4

10 66.4±1.8 58.4±1.6

15 66.4±1.8 58.4±1.6

Page 90: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

87

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

80

Metoclopramide (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.4a: Effect of Metoclopramide on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 91: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

88

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

80

Metoclopramide (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.4b: Effect of Metoclopramide on spontaneous rats’ ileal

preparations (n=3; Mean±SD)

Page 92: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

89

3.1.5 Verapamil:

Verapamil decreased the contractile response both in rabbits’ jejunal and rats’ ileal

preparations. Emax for verapamil was 6.2±0.8 and 3.8±0.2 for rabbits’ jejunal and rats’

ileal preparations, respectively (Table 3.5, Figure 3.5a & 3.5b).

Table 3.5: Effect of Verapamil (µM) in % of ACh on rabbits’ jejunal preparations and rats’

ileal preparations (Mean±SD, n=3)

Concentrations (µM)

Effect of verapamil on rabbits’ jejunal preparation

Effect of verapamil on rats’ ileal preparation

0.01 16.4±1.4 9.8±1.2

0.03 14.2±1.2 8.2±0.8

0.1 12.7±1.0 5.7±0.4

0.3 6.2±0.8 3.8±0.2

1 6.2±0.8 3.8±0.2

3 6.2±0.8 3.8±0.2

5 6.2±0.8 3.8±0.2

10 6.2±0.8 3.8±0.2

15 6.2±0.8 3.8±0.2

Page 93: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

90

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

20

Verapamil (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.5a: Effect of Verapamil on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 94: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

91

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

Verapamil (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.5b: Effect of Verapamil on spontaneous rats’ ileal preparations

(n=3; Mean±SD)

Page 95: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

92

3.1.6 Propranolol:

Propranolol decreased the contractile response both in rabbits’ jejunal and rats’ ileal

preparations. Emax for propranolol was 5.6±0.8 and 3.2±0.3 for rabbits’ jejunal and

rats’ ileal preparations, respectively (Table 3.6, Figure 3.6a & 3.6b).

Table 3.6: Effect of Propranolol (µM) in % of ACh on rabbits’ jejunal preparations and rats’

ileal preparations (Mean±SD, n=3)

Concentrations

(µM)

Effect of propranolol on rabbits’

jejunal preparation

Effect of propranolol on rats’ ileal

preparation

0.01 18.6±0.6 9.7±0.8

0.03 15.4±1.3 6.8±0.2

0.1 10.8±1.1 4.8±0.4

0.3 5.6±0.8 3.2±0.3

1 5.6±0.8 3.2±0.3

3 5.6±0.8 3.2±0.3

5 5.6±0.8 3.2±0.3

10 5.6±0.8 3.2±0.3

15 5.6±0.8 3.2±0.3

Page 96: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

93

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

20

Propranolol (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.6a: Effect of Propranolol on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 97: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

94

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

Propranolol (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.6b: Effect of Propranolol on spontaneous rats’ ileal preparations

(n=3; Mean±SD)

Page 98: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

95

3.1.7 Amiodarone:

Amiodarone decreased the contractile response both in rabbits’ jejunal and rats’ ileal

preparations. Emax for amiodarone was 10.0±0.2 and 7.1±0.4 for rabbits’ jejunal and

rats’ ileal preparations, respectively (Table 3.7, Figure 3.7a & 3.7b).

Table 3.7: Effect of Amiodarone (µM) in % of ACh on rabbits’ jejunal preparations and

rats’ ileal preparations (Mean±SD, n=3)

Concentrations

(µM)

Effect of Amiodarone on rabbits’

jejunal preparation

Effect of Amiodarone on rats’ ileal

preparation

0.01 18.1±1.2 14.3±0.8

0.03 17.8±1.4 12.6±0.6

0.1 15.9±0.8 10.8±1.2

0.3 14.4±0.6 8.6±0.5

1 13.2±0.6 8.0±0.5

3 10.0±0.2 7.1±0.4

5 10.0±0.2 7.1±0.4

10 10.0±0.2 7.1±0.4

15 10.0±0.2 7.1±0.4

Page 99: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

96

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

20

Amiodarone (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.7a: Effect of Amiodarone on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 100: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

97

0.01 0.03 0.1 0.3 1 3 5 10 15

0

5

10

15

Amiodarone (M)

Res

pon

se i

n %

0f

Ace

tylc

holi

ne

(3u

M)

Figure 3.7b: Effect of Amiodarone on spontaneous rats’ ileal preparations

(n=3; Mean±SD)

Page 101: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

98

3.2 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations:

AZM produced significant spasmogenic response both in rabbits’ jejunal and rats’ ileal

preparations. Mean (%) Emax for AZM was 67.6±1.6 and 54.0±2.1 for rabbits’ jejunal

and rats’ ileal preparations, respectively (Table 3.8, Figure 3.8a & 3.8b).

Table 3.8: Effect of Azithromycin (in % of ACh) on rabbits’ jejunal preparations and

rats’ ileal preparations (Mean±SD, n=3)

Concentrations (µM) Effect of AZM on rabbits’ jejunal

preparations

Effect of AZM on rats’ ileal

preparations

0.01 33.9±2.8 4.0±2.8

0.03 38.6±1.9 9.7±4.2

0.1 45.3±1.6 11.4±3.7

0.3 47.7±3.8 15.5±3.7

1 50.5±3.7 26.9±0.1

3 52.5±4.9 35.1±1.4

5 65.9±1.3 44.9±2.8

10 67.7±1.6 54±2.1

15 67.7±1.6 54±2.1

Page 102: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

99

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of A

cetylc

holi

ne (

3u

M)

Figure 3.8a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations (n=3; Mean±SD)

Page 103: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

100

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Figure 3.8b: Effect of Azithromycin on rats’ ileal preparations (n=3;

Mean±SD)

Page 104: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

101

3.3 Effect of AZM in the absence and presence of antagonists:

3.3.1 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Atropine:

The spasmogenic response produced by AZM in the presence of Atropine was

significantly reduced both in rabbits’ jejunal and rats’ ileal preparations. Mean (%)

Emax for AZM in the absence and presence of Atropine for rabbits’ jejunal preparations

was 68.3±1.3 and 2.4±0.1, respectively, while for rats’ ileal preparations it was

57.8±1.8 and 11.4±1.3, respectively (Table 3.9, Figure 3.9a & 3.9b). When the mean

(%) Emax was compared in the absence and presence of Atropine, it was found

significant with p˂0.0001 for rabbits’ jejunal preparations and p˂0.0001 for rats’ ileal

preparations (Figure 3.9c & 3.9d).

Table 3.9: Effect of Azithromycin (µM) in % of ACh in the absence and presence of

Atropine (0.3µM) on rabbits’ jejunal preparations and rats’ ileal preparations

(Mean±SD, n=3)

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Concentrations

(µM)

Rabbits’ Jejunum Rats’ Ileum

Effect of Azithromycin in

the absence of

Atropine

Effect of Azithromycin in

the presence of

Atropine (0.3µM)

Effect of Azithromycin in

the absence of

Atropine

Effect of Azithromycin

in the presence

of Atropine (0.3µM)

0.01 25.8±0.9 3.1±1.5*** 2.4±0 0±0**

0.03 29.5±1.1 3.1±1.5*** 5.6±2.8 2.4±4.2

0.1 40.5±0.8 2.3±0.1*** 9.7±2.4 4±3.7

0.3 48.1±1.4 2.3±0.1*** 13.8±1.4 4±3.7**

1 57.0±1.2 2.4±0*** 22.1±4.9 6.5±3.7**

3 63.3±1.1 2.4±0.1*** 35.9±1.4 10.6±1.3***

5 67.8±1.0 2.4±0.1*** 45.8±1.3 11.4±1.3***

10 68.3±1.3 2.4±0.1*** 57.8±1.8 11.4±1.3***

15 68.3±1.3 2.4±0.1*** 57.8±1.8 11.4±1.3***

Page 105: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

102

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Atropine

With Atropine (0.3µM)

Figure 3.9a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Atropine (n=3; Mean±SD)

Page 106: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

103

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Atropine

With Atropine (0.3µM)

Figure 3.9b: Effect of Azithromycin on rats’ ileal preparations in the

absence and presence of Atropine (n=3; Mean±SD)

Page 107: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

104

Without Atropine With Atropine

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.9c: Comparison of Azithromycin’s effect on spontaneous rabbits’

jejunal preparations in the absence and presence of Atropine (n=3;

Mean±SD)

Page 108: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

105

Without Atropine With Atropine

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.9d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Atropine (n=3; Mean±SD)

Page 109: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

106

3.3.2 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Loratadine:

The response of AZM in the presence of Loratadine was unaffected. Mean (%) Emax

for AZM was 68.8±0.6 for rabbits’ jejunal preparations in the absence of Loratadine

while it was 67.9 ± 2.0 in the presence of Loratadine. Similarly, mean (%) Emax for

rats’ ileal preparations was 52.4±2.8 and 50.7±1.9 in the absence and presence of

Loratadine respectively (Table 3.10, Figure 3.10a & 3.10b). Insignificant difference

(p=0.4 each) was found in rabbits’ jejunal and rats’ ileal preparation when the mean

(%) Emax for AZM was compared in the presence and absence of Loratadine (Figure

3.10c and 3.10d).

Table 3.10: Effect of Azithromycin (µM) in % of ACh in the absence and presence of

Loratadine (3µM) on rabbits’ jejunal preparations and rats’ ileal preparations (Mean±SD,

n=3)

Concentrations

(µM)

Rabbits’ Jejunum Rats’ Ileum

Effect of

Azithromycin in

the absence of Loratadine

Effect of

Azithromycin

in the presence of Loratadine

(3µM)

Effect of

Azithromycin in

the absence of Loratadine

Effect of

Azithromycin

in the presence of

Loratadine

(3µM)

0.01 31.3±3.6 22.9±1.0** 3.2±1.4 4.8±4.2

0.03 37.6±2.6 26.1±1.2*** 7.3±0 8.1±3.7

0.1 43.2±5.3 36.0±1.4 10.6±1.3 12.2±2.4

0.3 50.7±2.7 44.1±3.9 14.7±2.2 11.9±0.4

1 56.5±1.6 65.6±3.3** 26.4±2.1 19.5±4.2

3 59±1.7 66.1±3.0** 36.9±0.1 31.9±4.2

5 65.5±1.3 67.5±1.9 45.7±3.1 44.3±2.1

10 68.8±0.6 67.9±2 52.4±2.8 48.1±1.3

15 68.8±0.6 67.9±2 52.4±2.8 48.1±1.3

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Page 110: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

107

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Loratadine

With Loratadine (3µM)

Figure 3.10a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Loratadine (n=3; Mean±SD)

Page 111: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

108

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Loratadine

With Loratadine (3µM)

Figure 3.10b: Effect of Azithromycin on rats’ ileal preparations in the

absence and presence of Loratadine (n=3; Mean±SD)

Page 112: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

109

Without Loratadine With Loratadine

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p = 0.4

Figure 3.10c: Comparison of Azithromycin’s effect on spontaneous

rabbits’ jejunal preparations in the absence and presence of Loratadine

(n=3; Mean±SD)

Page 113: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

110

Without Loratadine With Loratadine

0

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p = 0.4

Figure 3.10d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Loratadine (n=3; Mean±SD)

Page 114: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

111

3.3.3 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Ondansetron:

AZM was unable to produce its full spasmogenic response in the presence of

Ondansetron. Mean (%) Emax for AZM in the absence and presence of Ondansetron

was 68.3±1.0 and 27.5±0.5 for rabbits’ jejunal preparations, and 55.6±1.5 and 34.0±2.9

for rats’ ileal preparations, respectively, (Table 3.11, Figure 3.11a & 3.11b). When the

mean (%) Emax for AZM in the absence and presence of Ondansetron was compared,

a significant difference of p˂0.0001 and p=0.0003 was found for rabbits’ jejunal and

rats’ ileal preparation respectively (Figure 3.11c & 3.11d).

Table 3.11: Effect of Azithromycin (µM) in % of ACh in the absence and presence of

Ondansetron (0.3µM) on rabbits’ jejunal preparations and rats’ ileal preparations

(Mean±SD, n=3)

Concentrations

(µM)

Rabbits’ Jejunum Rats’ Ileum

Effect of

Azithromycin in the absence

of Ondansetron

Effect of

Azithromycin in the presence of

Ondansetron

(0.3µM)

Effect of

Azithromycin in the absence

of Ondansetron

Effect of

Azithromycin in the presence

of Ondansetron

(0.3µM)

0.01 27.8±1.7 12.4±0*** 3.2±1.4 5.6±2.8

0.03 35.8±1.5 14±1.4*** 5.6±2.8 6.5±1.3

0.1 43.2±1.3 14±1.4*** 9.7±2.4 7.3±2.4

0.3 49.4±0.6 15.7±1.3*** 13.8±1.4 9.4±2.4*

1 53.8±1.3 17.3±0*** 27.2±0.4 12.1±4.1*

3 58.7±0.4 17.9±1.1*** 35.2±1.3 20.6±4.2**

5 64.3±1.5 21.6±1.1*** 45.9±1.0 31.2±1.4***

10 68.3±1.0 27.5±0.5*** 55.6±1.0 34±2.9***

15 68.3±1.0 27.5±0.5*** 55.6±1.0 34±2.9***

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Page 115: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

112

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

With Ondansetron (0.3µM)

Without Ondansetron

Figure 3.11a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Ondansetron (n=3; Mean±SD)

Page 116: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

113

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Ondansetron

With Ondansetron (0.3µM)

Figure 3.11b: Effect of Azithromycin on rats’ ileal preparations in the

absence and presence of Ondansetron (n=3; Mean±SD)

Page 117: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

114

Without Ondansetron With Ondansetron

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.11c: Comparison of Azithromycin’s effect on spontaneous

rabbits’ jejunal preparations in the absence and presence of Ondansetron

(n=3; Mean±SD)

Page 118: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

115

Without Ondansetron With Ondansetron

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p = 0.0003

Figure 3.11d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Ondansetron (n=3; Mean±SD)

Page 119: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

116

3.3.4 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Metoclopramide:

The response of AZM in the presence of Metoclopramide was unaffected. Mean (%)

Emax for AZM was 68.7±2.2 for rabbits’ jejunal preparations in the absence of

Metoclopramide while it was 70.3 ± 1.9 in the presence of Metoclopramide. Similarly,

mean (%) Emax for rats’ ileal preparations was 64.4±1.9 and 68.8±2.9 in the absence

and presence of Metoclopramide respectively (Table 3.12, Figure 3.12a & 3.12b).

Insignificant difference was found in rabbits’ jejunal (p=0.4) and rats’ ileal preparation

(p=0.09) when the mean (%) Emax for AZM was compared in the presence and

absence of Metoclopramide (Figure 3.12c and 3.12d).

Table 3.12: Effect of Azithromycin (µM) in % of ACh in the absence and presence of

Metoclopramide (10µM) on rabbits’ jejunal preparations and rats’ ileal (Mean±SD, n=3)

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Concentrations (µM)

Rabbit’s Jejunum Rat’s Ileum

Effect of

Azithromycin in

the absence of

metoclopramide

Effect of

Azithromycin in

the presence of

metoclopramide (10µM)

Effect of

Azithromycin

in the absence of

metoclopramide

Effect of

Azithromycin

in the presence

of metoclopramide

(10µM)

0.01 37±1.1 38.4±1.9 29.2±0.7 28.6±1.3

0.03 42.5±2 43.7±1.2 33.7±1.1 35.2±1.5|

0.1 43.7±1.1 47.6±0.8 35.5±1 36.8±0.2

0.3 50.5±1.9 51.3±3.7 41.3±0.9 44.2±3.8

1 54.4±2.1 56.2±2.9 45.1±0.8 48.9±0.4

3 61.7±1.2 64.1±2.1 52.4±1.8 54.2±0.2

5 62.1±1.7 65.1±1.4 60.4±2.2 67.8±5.1

10 68.7±2.2 70.3±1.9 64.4±1.9 68.8±2.9

15 68.7±2.2 70.3±1.9 64.4±1.9 68.8±2.9

Page 120: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

117

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Metoclopramide

With Metoclopramide (10µM)

Figure 3.12a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Metoclopramide (n=3;

Mean±SD)

Page 121: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

118

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Metoclopramide (10µM)

With Metoclopramide (10µM)

Figure 3.12b: Effect of Azithromycin on rats’ ileal preparations` in the

absence and presence of Metoclopramide (n=3; Mean±SD)

Page 122: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

119

Without Metoclopramide With Metoclopramide

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p=0.4

Figure 3.12c: Comparison of Azithromycin’s effect on spontaneous

rabbits’ jejunal preparations in the absence and presence of

Metoclopramide (n=3; Mean±SD)

Page 123: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

120

Without Metoclopramide With Metoclopramide

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p=0.09

Figure 3.12d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Metoclopramide (n=3;

Mean±SD)

Page 124: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

121

3.3.5 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Verapamil:

The spasmogenic response produced by AZM in the presence of Verapamil was

significantly reduced both in rabbits’ jejunal and rats’ ileal preparations. Mean (%)

Emax for AZM in the presence of Verapamil for rabbits’ jejunal preparations was

13.6±1.2 and for rats’ ileal preparations it was 22.3±2.5 (Table 3.13, Figure 3.13a &

3.13b). When the mean (%) Emax for AZM in the absence and presence of Verapamil

was compared, a significant difference of p˂0.0001 and p=0.0003 was found for

rabbits’ jejunal and rats’ ileal preparations respectively (Figure 3.13c & 3.13d).

Table 3.13: Effect of Azithromycin (µM) in % of ACh in the absence and presence of Verapamil

(0.3µM) on rabbits’ jejunal preparations and rats’ ileal preparations (Mean±SD, n=3)

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Concentrations

(µM)

Rabbit’s Jejunum Rat’s Ileum

Effect of

Azithromycin in the absence

of verapamil

Effect of

Azithromycin in the presence

of verapamil

(0.3µM)

Effect of

Azithromycin in the absence of

verapamil

Effect of

Azithromycin in the presence of

verapamil

(0.3µM)

0.01 43.4±1 2.4±0*** 5.5±2.7 2.4±0

0.03 45.7±1.2 2.4±0*** 6.5±3.7 2.4±0

0.1 48.9±0.5 2.4±0*** 9.9±2.7 2.4±0**

0.3 52.2±0.8 4.3±0.5*** 17.2±4.3 3.2±1.4**

1 56±0.7 8.1±0.5*** 20.7±5.3 5.7±1.4***

3 59±0.4 8.4±0.2*** 34.4±0.2 12.3±2.5***

5 63.3±0.8 9.7±0.7*** 43.3±2.4 18.1±2.8***

10 68.9±0.4 13.6±1.2*** 53.2±3.7 22.3±2.5***

15 68.9±0.4 13.6±1.2*** 53.2±3.7 22.3±2.5***

Page 125: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

122

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M) With Verapamil (0.3uM)

Without Verapamil

Figure 3.13a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Verapamil (n=3; Mean±SD)

Page 126: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

123

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Verapamil

With Verapamil (0.3µM)

Figure 3.13b: Effect of Azithromycin on rats’ ileal preparations in the

absence and presence of Verapamil (n=3; Mean±SD)

Page 127: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

124

Without Verapamil With Verapamil

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.13c: Comparison of Azithromycin’s effect on spontaneous

rabbits’ jejunal preparations in the absence and presence of Verapamil

(n=3; Mean±SD)

Page 128: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

125

Without Verapamil With Verapamil

0

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p = 0.0003

Figure 3.13d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Verapamil (n=3; Mean±SD)

Page 129: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

126

3.3.6 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Propranolol:

The amplitude of contraction in rabbit’s jejunum and rat’s ileum was markedly reduced

when AZM was given in the presence of Propranolol. Mean (%) Emax in the presence

of Propranolol was found to be 10.2±2.1 for rabbits’ jejunal preparations and 15.6±1.4

for rat’s ileal preparations (Table 3.14, Figure 3.14a & 3.14b). A significant difference

of p˂0.0001 for rabbit’s jejunal preparations and p˂0.0001 for rat’s ileal preparations

was found when the mean (%) Emax of AZM in the absence and presence of

Propranolol was compared (Figure 3.14c & 3.14d).

Table 3.14: Effect of Azithromycin (µM) in % of ACh in the absence and presence of

Propranolol (0.3µM) on rabbits’ jejunal preparations and rats’ ileal preparations

(Mean±SD, n=3)

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Concentrations

(µM)

Rabbit’s Jejunum Rat’s Ileum

Effect of Azithromycin

in the absence

of propranolol

Effect of Azithromycin

in the presence

of propranolol

(0.3µM)

Effect of Azithromycin

in the absence

of propranolol

Effect of Azithromycin

in the presence

of propranolol

(0.3µM)

0.01 32.8±0.5 9.0±2.8*** 3.2±1.4 3.2±1.4

0.03 37.9±1.5 9.0±2.8*** 4.8±2.4 3.2±1.4

0.1 47±1.6 9.8±2.5*** 9.7±2.4 5.7±2.8

0.3 51.6±1.3 9.8±2.5*** 12.2±2.4 6.5±1.4**

1 53.9±1.3 9.9±2.5*** 24±2.4 5.7±1.4***

3 57.7±1.4 9.9±2.5*** 35.2±1.3 8.2±1.3***

5 66.6±2.1 10.2±2.1*** 45.1±2.5 9.8±2.4***

10 68.2±1.1 10.2±2.1*** 54.8±1.1 15.6±1.4***

15 68.2±1.1 10.2±2.1*** 54.8±1.1 15.6±1.4***

Page 130: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

127

Figure 3.14a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Propranolol (n=3; Mean±SD)

Page 131: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

128

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Propranolol

With Propranolol (0.3µM)

Figure 3.14b: Effect of Azithromycin on rats’ ileal preparations in the

absence and presence of Propranolol (n=3; Mean±SD)

Page 132: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

129

Without Propranolol With Propranolol

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.14c: Comparison of Azithromycin’s effect on spontaneous

rabbits’ jejunal preparations in the absence and presence of Propranolol

(n=3; Mean±SD)

Page 133: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

130

Without Propranolol With Propranolol

0

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.14d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Propranolol (n=3; Mean±SD)

Page 134: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

131

3.3.7 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Amiodarone:

The response of AZM on rabbits’ jejunal and rats’ ileal contraction remained

undisturbed in the presence of Amiodarone. Mean (%) Emax for AZM in the absence

of Amiodarone was 69.8±0.5 and 62.9±3.0 for rabbits’ jejunal and rats’ ileal

preparations, respectively, while in the presence of Amiodarone, mean (%) Emax was

68.4±1.3 for rabbits’ jejunal preparations and 58.0±3.4 for rats’ ileal preparations

(Table 3.15, Figure 3.15a & 3.15b). Mean (%) Emax for AZM was compared in the

presence of Amiodarone, it showed an insignificant difference with p=0.1 for rabbits’

jejunal preparations and p=0.1 for rats’ ileal preparations (Figure 3.15c & 3.15d).

Table 3.15: Effect of Azithromycin (µM) in % of ACh in the absence and presence of

Amiodarone (3µM) on rabbits’ jejunal preparations and rats’ ileal preparations (Mean±SD,

n=3)

Concentrations (µM)

Rabbit’s Jejunum Rat’s Ileum

Effect of

Azithromycin in

the absence of Amiodarone

Effect of

Azithromycin in

the presence of Amiodarone

(3µM)

Effect of

Azithromycin in

the absence of Amiodarone

Effect of

Azithromycin

in the presence of Amiodarone

(3µM)

0.01 35.3±2.8 30.9±1.3 4±2.8 4.1±1.3

0.03 44.5±4.9 37.5±1.4 5.6±2.8 4.1±1.3

0.1 55.6±7.6 48.8±1.7 11.3±3.8 6.5±1.3

0.3 60.9±5.5 58.5±2.2 15.5±3.7 10.5±1.3

1 64.6±2.3 60.8±1.2 23.6±5.6 22.2±2.2

3 67±2.5 62.5±3.9 35.1±1.4 38.7±7.4

5 68.4±1 65.9±1.8 44.9±2.8 49.3±2.0

10 69.8±0.5 68.4±1.3 62.9±3 58.0±3.4

15 69.8±0.5 68.4±1.3 62.9±3 58.0±3.4

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Page 135: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

132

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Amiodarone

With Amiodarone (3µM)

Figure 3.15a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Amiodarone (n=3; Mean±SD)

Page 136: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

133

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without Amiodarone

With Amiodarone (3µM)

Figure 3.15b: Effect of Azithromycin on rats’ ileal preparations in the

absence and presence of Amiodarone (n=3; Mean±SD)

Page 137: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

134

Without Amiodarone With Amiodarone

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p = 0.17

Figure 3.15c: Comparison of Azithromycin’s effect on spontaneous

rabbits’ jejunal preparations in the absence and presence of Amiodarone

(n=3; Mean ± SD)

Page 138: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

135

Without Amiodarone With Amiodarone

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p = 0.16

Figure 3.15d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Amiodarone (n=3; Mean±SD)

Page 139: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

136

3.3.8 Effect of AZM on rabbits’ jejunal and rats’ ileal preparations in the

absence and presence of Atropine, Ondansetron, Verapamil &

Propranolol:

The spasmogenic response of AZM was completely lost in the presence of Atropine,

Ondansetron, Verapamil and Propranolol (AOVP), both in rabbits’ jejunal and rats’

ileal preparations (Table 3.16, Figure 3.16a & 3.16b). When the mean (%) Emax for

AZM in the absence and presence of AOVP was compared, a significant difference of

p˂0.0001 and p˂0.0001 was found for rabbits’ jejunal and rats’ ileal preparation,

respectively (Figure 3.16c & 3.16d).

Table 3.16: Effect of Azithromycin (µM) in % of ACh in the absence and presence of atropine,

ondansetron, verapamil & propranolol (AOVP) (0.3µM) on rabbits’ jejunal preparations

and rats’ ileal preparations (Mean±SD, n=3)

Concentrations (µM)

Rabbit’s Jejunum Rat’s Ileum

Effect of

Azithromycin

in the absence

of AOVP

Effect of

Azithromycin

in the presence

of AOVP (0.3µM)

Effect of

Azithromycin in

the absence

of AOVP

Effect of

Azithromycin

in the

presence

of AOVP

(0.3µM)

0.01 27.8±1.7 0.0±0.0*** 7.5±0.4 0.0±0.0***

0.03 35.8±1.5 0.0±0.0*** 11.5±3.8 0.0±0.0***

0.1 43.2±1.3 0.0±0.0*** 12.4±2.4 0.0±0.0***

0.3 49.4±0.6 0.0±0.0*** 16.8±2.4 0.0±0.0***

1 53.8±1.3 0.0±0.0*** 22.4±3.9 0.0±0.0***

3 58.7±0.4 0.0±0.0*** 30±3.7 0.0±0.0***

5 64.3±1.5 0.0±0.0*** 38.8±3.1 0.0±0.0***

10 68.3±1 0.0±0.0*** 45±3.2 0.0±0.0***

15 68.3±1 0.0±0.0*** 45±3.2 0.0±0.0***

p ≤ 0.05 = *, p ≤ 0.01 = **, p ≤ 0.001 = ***, Student ‘t’ test

Page 140: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

137

0.01 0.03 0.1 0.3 1 3 5 10 15

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

With AOVP (0.3µM)

Without AOVP

Figure 3.16a: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Atropine, Ondansetron,

Verapamil & Propranolol (n=3; Mean±SD)

Page 141: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

138

0.01 0.03 0.1 0.3 1 3 5 10 150

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3u

M)

Without AOVP

With AOVP (0.3µM)

Figure 3.16b: Effect of Azithromycin on spontaneous rabbits’ jejunal

preparations in the absence and presence of Atropine, Ondansetron,

Verapamil & Propranolol (n=3; Mean±SD)

Page 142: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

139

Without AOVP With AOVP

0

20

40

60

80

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.16c: Comparison of Azithromycin’s effect on spontaneous

rabbits’ jejunal preparations in the absence and presence of Atropine,

Ondansetron, Verapamil & Propranolol (n=3; Mean±SD)

Page 143: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

3-Results

140

Without AOVP With AOVP

0

20

40

60

Azithromycin (M)

Resp

on

se i

n %

of

Acety

lch

oli

ne (

3

M)

p < 0.0001

Figure 3.16d: Comparison of Azithromycin’s effect on rats’ ileal

preparations in the absence and presence of Atropine, Ondansetron,

Verapamil & Propranolol (n=3; Mean±SD)

Page 144: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

141

4 DISCUSSION

Azithromycin is one of the most widely prescribed antibiotics worldwide for various

clinical conditions. Therefore, Azithromycin remained the focus for researchers from

very start to further explore its pharmacological actions, adverse effects and drug

interactions. Very limited studies are conducted to investigate its effects on intestinal

smooth muscles and the possible mechanisms involved in it. The focus of this study

was to evaluate the effects of AZM on the contractility of intestinal smooth muscles

and to investigate the underlying mechanisms.

Results of the present study confirmed spasmogenic response by AZM, it increased

intestinal contractility by acting on muscarinic and serotonergic receptors, and voltage

gated calcium and sodium channels. Statistical comparison of Emax of AZM in the

absence and presence of respective antagonist is as follows; p˂0.0001 for rabbits’

jejunum and p˂0.0001 for rats’ ileum in the presence of Atropine; p˂0.0001 for rabbits’

jejunum and p=0.0003 for rats’ ileum in the presence of Ondansetron; p˂0.0001 for

rabbits’ jejunum and p=0.0003 for rats’ ileum in the presence of Verapamil; p˂0.0001

for rabbits’ jejunum and p˂0.0001 for rats’ ileum in the presence of Propranolol.

4.1 Effect of AZM on intestinal motility:

Spasmogenic response produced by AZM was a bit more in rabbit’s jejunum than rat’s

ileum. It may be because of more stronger and propagative contractions of jejunum as

compare to ileum (Seidl et al., 2012).

Rabbit’s jejunum was found more sensitive even to smaller doses of AZM as it has

responded well to very low doses of AZM i.e. 0.01 to 0.3µg/ml. Not like other smooth

muscles of the GIT, rabbit’s jejunum characteristically contains high electrically

excitable cells and hence even smaller doses of AZM have produced the significant

contractions (Kurle et al., 2017).

A study was performed Sifrim et al to find the effects of AZM on the contractions of

small intestine. AZM was found to increase the frequency and amplitude of small

intestinal contractions, confirming the spasmogenic action of AZM. Similar studies

Page 145: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

4-Discussion

142

were conducted by El-Baki et al., and Chini et al., (El-Baki et al., 2015; Chini et al.,

2012). They also found the spasmogenic activity of AZM. The results of our study are

also in accordance to the results of these studies that AZM produces spasmogenic

response.

Unlike the results of our and above mentioned studies, Chiragh et al conducted a study

and noted that the prokinetic effects of AZM in vitro on rabbit’s duodenum are not well-

sustained. Thus, showing AZM not to be a beneficial therapeutic agent to produce

prokinetic actions (Chiragh et al., 2006). Nevertheless, its spasmogenic effect was

confirmed.

4.2 AZM and atropine:

Smooth muscle of small intestine contracts at regular intervals in the absence of

neuronal or hormonal stimulation; such contractions are referred to as phasic

(Weisbrodt, 2007). A spike potential must be produced for contraction which are

assumed to be generated by inward Ca2+ currents (Montgamry, et al., 2016).

ACh is the major excitatory neurotransmitter of the ENS, has been shown to increase

the amplitude of spontaneous contractions in the rabbits small intestine, through

M3 subtype of muscarinic receptor (Montgommery et al., 2016; Grassa et al., 2014).

This subtype is coupled to Gq receptors and stimulation of M3/Gq‐coupled receptors

activates phospholipase C (PLC) which cleaves triggers Ca2+ release from sarcoplasmic

reticulum (SR) via inositoltriphosphate (IP3) receptors to initiate contraction (Semenov

et al., 2018). Atropine by blocking the binding of ACh with muscarinic receptors blocks

all these events. Hence, we tried AZM in the presence of atropine to confirm the

involvement of M3 receptors as these receptors are predominant in GIT.

According to Gordienko et al., IP3 Ca2+ release is even facilitated by Ca2+ influx

through voltage operated channels. Thus, it is possible to speculate that the AZM

might add to its spasmogenic effect through increase in extracellular influx of

Ca2+ (Gordienko et al., 2008) produced by voltage gated Ca2+ channels.

Therefore, voltage gated Ca2+ channels can facilitate in ACh induced

contractions (Chokri et al., 2010). The response of AZM in the presence of

voltage gated Ca2+ channels is discussed in the section 4.6.

Page 146: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

4-Discussion

143

However, our results are suggestive of cholinergic receptors mediated effects of AZM

because in the presence of cholinergic antagonist (atropine 0.3µM), the increase in the

intestinal contractility was significantly minimal. The spasmogenic effect of AZM is

mainly due to release of Ca2+ from sarcoplasmic reticulum which is triggered

through M3 receptors excitation. In relation to our findings, another study also

confirmed that AZM exerts its effect on the jejunal motility via involvement of

muscarinic receptors but in contrast to our results this study did not show involvement

of muscarinic receptors by AZM in the motility of ileum (El-Baki et al., 2015). Wang

et al also showed the involvement of muscarinic receptors by AZM in the contractions

of airway smooth muscles (ASMs). They pretreated the ASMs with ACh both in

animals (rabbits, guinea pigs and mice) and humans. AZM completely inhibited the

ACh-induced precontractions of ASMs (Wang et al., 2019). Thus it is suggested the

AZM predominantly produces spasmogenic response via involvement of M3 receptors.

4.3 AZM and loratadine:

H1 and H4 receptors are mainly found in the GIT (Mittal et al., 2017). H1 receptors are

a bit dominant and widely expressed in blood vessels, muscle layer, enterocytes,

immune cells and ganglion cells of the myenteric plexus in the GIT of humans. On the

other hand expression of H4 receptors is lower than H1 receptors in human stomach,

small intestine and colon (Kim et al., 2011; Dieteren et al., 2015).

Coupling of H1 receptors is dual in nature. They are coupled to Gq/11 proteins, activates

PLC and the phosphatidylinositol 4,5-bisphosphate (PIP2) pathway. H1 receptor

stimulation results in smooth muscle contraction mediated by IP3 induced mobilization

of intracellular Ca2+. They are also associated to Gi/o proteins, resulting in decrease

production of cAMP (Monczor et al., 2016). Decrease in cAMP levels result in increase

in the smooth muscle contraction (Bilington et al., 2003; Mizuta et al., 2013; Kuo et

al., 2015).

It can be speculated from the above mentioned points that AZM has no effect on the

levels or activity of adenylyl cyclase and cAMP, therefore it does not significantly

altered the intestinal motility in the presence of histamine receptor blocker. However,

there might be a possibility that it had increased intestinal contraction up to some extent

by increasing mobilization of intracellular Ca2+ mediated by IP3, as it was obvious in

Page 147: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

4-Discussion

144

case of muscarinic receptor blocker (section 4.2). The latter idea was confirmed when

the tissue preparations were tested in the presence of all antagonists.

In a study the effects of AZM on intestinal motility was evaluated through histamine

receptors. It was noted that AZM in the presence of histamine receptors blockade was

still able to produce jejunal stimulation (El-Baki et al., 2015). The results of this study

confirmed that AZM does not exerts its effect on the intestinal motility via histamine

receptors. Same results were found in our study that AZM does not significantly involve

the H1 receptors in the spasmogenic response.

4.4 AZM and ondansetron:

5-HT acts as an important neurotransmitter in the ENS. 5-HT has various subtypes but

5-HT3 & 5-HT4 are predominantly present in the intestinal smooth muscles (Mittal et

al, 2017). Various researches have proved significant role of 5-HT in regulating GI

functions (Bomstein, 2012).

Stimulation of 5-HT3 receptors leads to different effects, including depolarization of

membrane and rise in intracellular Ca2+, modulation of neurotransmitter release, central

and peripheral neuron’s excitation, and release of 5-HT from EC cells of small intestine.

5-HT3 receptors may enhance spontaneous release by producing depolarization locally

at presynaptic terminal, leading to augmented influx of Ca2+ through VGCCs.

Moreover, 5-HT3 receptors are closely related with the ACh receptor, present in the

central and peripheral nervous systems. (Altamirano et al, 2018; Terry et al., 2017). 5-

HT receptors increase the release of ACh from the motor nerve endings. All these

events increases the amplitude of intestinal muscles contractions Serotonin antagonist

are reported to decrease the intestinal contractility by blocking all these happenings

(Terry et al., 2017).

As in the present study we found that one of the mechanisms by which AZM affects

the intestinal contractility is through muscarinic receptors (ACh) which also act by

increasing the extracellular influx of Ca2+(Gordienko et al., 2008) . 5-HT also acts

through the same mechanism. Therefore, it can be predicted that AZM in the

presence of 5-HT antagonists increases the intestinal contractility by increasing

the extracellular influx of Ca2+.

Page 148: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

4-Discussion

145

In contrast to the results of our study where the involvement of 5-HT receptors were

found in the spasmogenic response produced by AZM, another study to investigate the

role of 5-HT receptors in the AZM induced spasmogenic response was conducted. The

effects of AZM on the contractions of jejunum were evaluated in the presence of 5-HT

antagonist. Results showed no involvement of 5-HT receptors in increasing the

contractility of small intestine as AZM was found to increase the jejunal contractions

even in the presence of 5-HT receptors blockade (El-Baki et al., 2015).

4.5 AZM and metoclopramide:

Various subtypes of DA receptors are found in the intestinal smooth muscles but D2 is

found abundantly (Ayano, 2016). Both inhibitory and excitatory effects of DA are

observed in GIT but usually inhibitors effects are more frequent. Release of DA from

enteric dopaminergic neurons and subsequent binding to DA receptors in GI smooth

muscles inhibits the GI motility mainly by activating D2 receptors (Giudicessi et al.,

2018). D2 receptors also inhibit the release of ACh from intrinsic cholinergic nerve

terminals (Tonini et al., 2004).

AZM is not found to act through dopamine receptors because there is no significant

difference in the amplitude of contractions between the two groups. In the presence of

Metoclopramide (10µM) the AZM increases the intestinal contractility though not

statistically significant. It might be because of the effects of AZM on Metoclopramide

induced release of ACh as discussed in section 4.2 (Tonini et al., 2004).

4.6 AZM and verapamil:

The Cav 1.2 L-type Ca2+ channel is the dominant voltage-activated Ca2+ channel in

heart and smooth muscles. A rise in intracellular calcium is the trigger for Gl smooth

muscles contractions. Under normal conditions, membrane depolarization triggers an

influx of calcium and this calcium serves as an activator source for contractions. A

number of studies have found that GI smooth muscles do not show spontaneous

contractile activity during exposure to calcium-free solution (Evans et al., 2011). In the

cytoplasm of SMC, Ca2+ binds to calmodulin and the calcium-calmodulin complex

combines with the catalytic subunit of myosin light chain (MLC) kinase to form a

receptor leading to the phosphorylation of serine (Ser) at position 19 in the MLC.

Phosphorylation of Ser-19 of MLC then allows myosin ATPase to be activated by actin

Page 149: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

4-Discussion

146

and muscle contraction takes place. When cytoplasmic (Ca2+) is reduced, MLC

phosphatase dephosphorylates MLC20, thereby deactivating actomyosin ATPase and

causing relaxation. Ca2+ is provided by both extracellular and intracellular sources (Zhu

et al., 2001).

Thus it can be predicted that the spasmogenic response of AZM is mostly dependent on

Ca2+. Verapamil, a calcium channel blocker, decreases the influx of Ca2+ by blocking

calcium channels. AZM in the presence of verapamil is unable to increase the inward

flux of Ca2+ which is basically responsible for increase in intestinal contraction.

A study performed by Wang et al., has reported the inhibition of VGCCs by AZM

(Wang et al., 2019) in smooth muscles. Zhaug and his co-workers also found the

involvement of AZM in the inhibition of VGCCs (Zhaug et al., 2014). In another study

performed by Silu and his colleagues, AZM was found to decrease the mucus secretion

in ASMs by inhibiting the Ca2+ entry into submucus gland cells (SMGCs) (Silu et al,

2010). Our results are also indicative of the fact that AZM inhibits VGCCs, as when

the effect of AZM was evaluated in the presence of calcium channel blocker, a very

minimal increase in the amplitude of intestinal smooth muscle contraction was found.

El-Baki et al., performed a study to evaluate the effects of AZM on rabbit’s jejunum.

They also investigated the role of calcium channels in the spasmogenic response of

AZM. They found that calcium channels play a significant role in the actions of AZM

as stimulant to contractions of jejunum (El-Baki et al., 2015). The results of this study

is in accordance with the results of our study as we also found the involvement of

VGCCs in jejunal and ileal contractions.

The effects of AZM were also assessed in vitro on isolated colon of rats. AZM was

found to increase colonic contractions. For further investigation of the underlying

mechanisms by which AZM affects the colonic contractility, first tissue was treated

with calcium channel blockers and then the response of AZM was noted. Colonic

contractions induced by AZM were declined in the presence of calcium channel blocker

(El-Baki et al., 2015). This showed the involvement of calcium channels in AZM

induced contractions. The association of VGCCs with intestinal contractility is

common both in this as well as in our study.

Page 150: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

4-Discussion

147

In contrast to the results of our study, Daenas et al., conducted a study to evaluate the

effect of AZM on precontracted ASMs. AZM was found to possess dose-dependent,

relaxant effect on precontracted ASMs which is not mediated via inhibition of

Ca2+ release or Ca2+ influx. These results are dissimilar from the results of our study

which showed partially Ca2+ mediated contraction of intestinal smooth muscles by

AZM (Daenas et al., 2006).

4.7 AZM and propranolol:

The involvement of voltage-gated sodium channels (NaV) in the regulation of resting

membrane potential are evident from the Intracellular recordings of human small

intestine smooth muscle strips (Nashatian et al., 2015). NaV channels play a key role

in initiation of action potentials. During membrane depolarization, Na+ appears to

gather in areas under the plasma membrane, producing local concentration gradients

that effect cell Ca2+ handling mechanisms (Soomer et al., 2017).

AZM failed to increase the intestinal contractions in the presence of NaV channel

blocker, confirming to affect the process of depolarization. During depolarization, the

membrane potential quickly moves from negative to positive. The NaV channels open

in reaction to an early change in voltage. As the Na+ rush back into the cell, the interior

of the cell become positive, ultimately changing the membrane potential from negative

to positive. When the interior of the cell becomes more positively charged,

depolarization of the cell is complete, and the channels close again (Molnar et al.,

2015). Thus involvement of Na+ channels cannot be ruled out in the AZM triggered

tissue contractions.

4.8 AZM and amiodarone:

Potassium channels comprise half of the ion channel superfamily, and voltage-sensitive

potassium channels (KV) are the largest group. Wide diversity of potassium channels is

detected in ICCs and SMCs of the GIT. The intracellular K+ concentration is 10 times

higher than extracellular (Beyder et al., 2012).

Following depolarization, the NaV channels close again that had been open while the

cell was undergoing depolarization. The potassium channels now open due to increased

positive charge within the cell. K+ begins to move out of the cell and the potential within

Page 151: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

4-Discussion

148

the cell declines and approaches its resting potential (Molnar et al., 2015). The effect

of AZM in the presence AZM was not found to act through KV channels, thus not

interfering in the process of repolarization.

AZM effect was completely blocked in the presence of all these antagonists suggesting

the involvement of mixed pathways in the spasmogenic response produced by AZM.

Though it is evident that the maximum spasmogenic response is mediated through

muscarinic receptors of the intestine.

Page 152: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

149

5 CONCLUSION

Maximum spasmogenic response of Azithromycin is through involvement of triggering

of muscarinic receptors as Azithromycin effect was primarily blocked in the presence

of atropine, a standard anticholinergic drug. Moreover, our results suggest that

azithromycin increases the contractility of rabbits’ jejunal and rats’ ileal preparations

by involving multiple receptors and voltage gated channels. Decreased contractility of

rabbits’ jejunal and rats’ ileal preparations in the presence of atropine, ondansetron,

verapamil and propranolol confirmed the core involvement of muscarinic receptors

following mixed pathways involving serotonergic receptors, voltage gated calcium and

sodium channels in the production of spasmogenic response by azithromycin.

Recommendations:

1). Azithromycin is frequently prescribed by clinicians to pregnant ladies for treating

various infections. Uterus is also made up of smooth muscles and more likely it will

also respond to Azithromycin in the same manner (increase in contractility) as other

smooth muscles of the body respond, Azithromycin may lead to abortion or premature

labour if used in pregnancy. Therefore, results of our study strongly suggest to evaluate

the effects of Azithromycin on uterine smooth muscles as well for safe clinical practice.

2). Azithromycin by increasing the intestinal contractility may significantly affect the

absorption of drugs which requires more time for absorption. Hence, our studies open

a new window of research to basic scientists and clinicians to study further the possible

drug-drug interactions of Azithromycin and other primary drug therapies that requires

Azithromycin in combination.

Limitations:

The limitations of the study are that no in vivo activity was performed, all the activities

performed through Power Lab. were in vitro. Power Lab. has very sensitive transducers,

which needs specific temperature and constant pressure during the procedure. A small

change either in the temperature or pressure may affect the results.

Page 153: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

150

REFERENCES

ADELI, E. 2016. Preparation and evaluation of azithromycin binary solid dispersions

using various polyethylene glycols for the improvement of the drug solubility and

dissolution rate. Braz. J. Pharm. Sci. 52, 1-13.

AFZAL, A., KHAN, B. T., BAKHTIAR, S. 2011. Ondansetron: A newer aspect of

dose response relationship on ileal smooth muscles of rabbit. Pak J Pharm Sci, 29, 119-

24.

ALAHAMD, Y., SWEHLI, H. A. I., RAHHAL, A., SARDAR, S., ELHASSAN, M. A.

M., ALSAMEL, S. & IBRAHIM, O. A. 2020. Calcium Channel Blockers. New Insight

into Cerebrovascular Diseases: An Updated Comprehensive Review, 217.

AKIYOSHI, T., ITO, M., MURASE, S., MIYAZAKI, M., GUENGERICH, F. P.,

NAKAMURA, K., YAMAMOTO, K., OHTANI, H. 2013. Mechanism-based

inhibition profiles of erythromycin and clarithromycin with cytochrome p450 3a4

genetic variants. Drug Metab Pharmacokinet, 28(5):411-15.

ALI, N., AHMAD, B., SHAH, S. W. 2011. Spasmogenic and spasmolytic activities of

Onosma griffithii Vatke. Pak J Pharm Sci, 24, 553-8.

ALI, N., BASHIR, A., SHUMAILA, B., JEHANDAR, S., SADIQ, A. & MANZOOR,

A. 2009. Calcium channel blocking activities of Withania coagulans. Afr. J. Pharm.

Pharmacol, 3, 439-442.

ALI, N., JAMIL, A., SHAH, S. W. A., SHAH, I., AHMED, G. 2017. Spasmogenic and

spasmolytic activity of rind of Punica granatum Linn. BMC Complement Altern Med,

17, 2-7.

ALTAMIRANO, J. L. C., HERNANDEZ, A. O., JAIME, H. B., MORA, P.

C., BANDALA, C., LONG, S. R., RODRÍGUEZ, A. A. 2018. Review: 5-HT1, 5-HT2,

5-HT3 and 5-HT7 receptors and their role in the modulation of pain response in the

central nervous system. Curr Neuropharmacol, 16, 210–21.

Page 154: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

151

ANDERSON, M. L., WINTER, M.L.F. 2017. Animal models in biological and

biomedical research - experimental and ethical concerns. An Braz Acad Sci, 91, 461-

67.

ASSI, R. A., DARWIS, Y., ABDULBAQI, I. M., ASIF, S. M. 2017. Development and

validation of a stability-indicating RP-HPLC method for the detection and

quantification of azithromycin in bulk, and self-emulsifying drug delivery system

(SEDDs) formulation. J Appl Pharm Sci, 7, 20–29.

AYANO, G. 2016. Dopamine: receptors, functions, synthesis, pathways, locations and

mental disorders: review of literatures. J Ment Disord Treat, 2, 2-4.

AYERBE, J. I. G., HAUSER, B., SALVATORE, S., VANDENPLAS, Y. 2019.

Diagnosis and management of gastroesophageal reflux disease in infants and children:

from guidelines to clinical practice. Pediatr Gastroenterol Hepatol Nutr, 22, 107–21.

AYSE, E. R. 2013. Azithromycin prevents pregnancy loss: Reducing the level of tumor

necrosis factor-alpha and raising the level of interleukin-10 in rats. Mediators

Inflamm, 928137.

BADYAL, D. K., DESAI, C. 2014. Animal use in pharmacology education and

research: The changing scenario. Indian J Pharmacol, 46, 257–65.

BAKHEIT, A. H., AL-HADIYA, B. M., ABD-ELGALIL, A. A. Azithromycin.

Profiles of Drug Substances Excipients Related Methodology (Third Edition). 2014.

Elsevier; Academic Press.

BARSHOP, K., KUO, B. 2015. The investigational drug camicinal for the treatment of

gastroparesis. Expert Opin Inv Drug, 24, 133-40.

BEYDER, A., FARRUGIA, G. 2012. Targeting ion channels for the treatment of

gastrointestinal motility disorders. Therap Adv Gastroenterol, 5, 5-21.

Bhagavan N. V., Chung-Eun H. In Essentials of Medical Biochemistry,

Gastrointestinal Digestion and Absorption. 2011. Science direct.

BIELEFELDT, K., TUTEJA, A., NUSRAT, S. 2016. Disorders of gastrointestinal

hypomotility. Faculty Rev, 1, 1897-1905.

Page 155: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

152

BILLINGTON, C. K., PENN, R. B. 2003. Signaling and regulation of G protein-

coupled receptors in airway smooth muscle. Respir Res, 4, 2.

BITAR, K. N. 2003. Function of gastrointestinal smooth muscle: from signaling to

contractile proteins. Am J Med. 18;115 Suppl 3A:15S-23S.

BLONDEAU, K., MERTENS, V., VANAUDENAERDE, B. M., RAEMDONCK, D.

E., VERLEDEN, G. M., SIFRIM, D., DUPONT, L. J. 2007. Effect of azithromycin on

gastroesophageal reflux (acid-non-acid) and pepsin in BALF of lung transplant

patients. J Heart Lung Transpl, 26, 139-40.

BOMSTEIN, J. C. 2012. Serotonin in the gut: What does it do? Front Neurosci, 6, 1612-

17.

BOSNAR, M., KRAMARIC, D. M., NUJIC, K., POLANCEC, S. D., MARJANOVIC,

N., GLOJNARIC, I., HABER, E. V. 2013. Immunomodulatory effects of azithromycin

on the establishment of lipopolysaccharide tolerance in mice. Int

Immunopharmacol, 15, 498-504.

BRANDLER, J., MILLER, L. J., WANG, X. J., BURTON, D., BUSCIGLIO, I.,

ARNDT, K., HARMSEN, W. S. & CAMILLERI, M. 2020. Secretin effects on gastric

functions, hormones and symptoms in functional dyspepsia and health: randomized

crossover trial. Am J Physiol Gastrointest Liver Physiol, 318, G635-G645.

BROAD, J., SANGER, G. J. 2013. The antibiotic azithromycin is a motilin receptor

agonist in human stomach: comparison with erythromycin. Br J Pharmacol, 168, 1859–

67.

BROWNING, K. N. & TRAVAGLI, R. A. 2011. Central nervous system control of

gastrointestinal motility and secretion and modulation of gastrointestinal functions.

Compr physiol, 4, 1339-1368.

BRYDA, E. C. 2013. The Mighty Mouse: the impact of rodents on advances in

biomedical research. Mo Med, 110, 207.

BULSKA, M., MICHALAK, D. O. 2014. Immunomodulatory and anti-inflammatory

properties of macrolides. Curr. Issues Pharm. Med. Sci. 27, 61-64.

Page 156: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

153

CAMPBELL, L. 2015. Gut motility and its control. Anesth Intens Care Med, 16, 40-

42.

CASTANEDA-LOPEZ, M. E., GARZA-VELOZ, I., ORTIZ-RODRIGUEZ, J. M.,

CASTANEDA-MIRANDA, R., SOLIS-SANCHEZ, L. O., VEGA-CARRILLO, H. R.,

et al. 2017. Animal Models of Rheumatoid Arthritis. Experimental Animal Models of

Human Diseases-An Effective Therapeutic Strategy. IntechOpen.

CHAMBERS, H. F., DECK, D. H. Tetracyclines, macrolides, clindamycin,

chloramphenicol, streptogramins & Oxazolidinones. In: Basic and clinical

Pharmacology. editors KATZUNG, B. G., MASTERS, S. B., TREVOR, A. J. 11th Ed.

McGrawHill; 201. P. 797.

CHDA, A., EL-KABBAOUI, M., CHOKRI, A., EL-ABIDA, K., TAZM, A., CHEIKH,

R. B. 2016. Spasmolytic action of Centaurium erythraea on rabbit jejunum is through

calcium channel blockade and NO release. European J Med Plants, 11, 1-13.

CHINI, P., TOSKES, P. P., WASEEM, S., HOU, W., MCDONALD, R., MOSHIREE,

B. 2012. Effect of azithromycin on small bowel motility in patients with gastrointestinal

dysmotility. Scand J Gastroenterol, 47, 422–27.

CHIRAGH, S., BEGUM, A., KARIM, S. 2006. Prokinetic effect of clarithromycin and

azithromycin in vitro study on rabbit duodenum. Biomedica, 22, 130-31.

CHOKRI, A., DOUKALI, R., EL-ABIDA, K., CHEIKH, R. B. 2010. Myorelaxant and

spasmolytic effects of Globularia alypum L. extract on rabbit jejunum. Int J Pharm, 6,

608-15.

CHURCH, M. K., CHURCH, D.S. Pharmacology of antihistamines. Indian journal of

dermatology, 58, 219.

CRAMER, C, L., PATTERSON, A., ALCHAKAKI, A., SOUBANI, A. O. 2017.

Immunomodulatory indications of azithromycin in respiratory disease: a concise

review for the clinician. Postgrad Med, 129, 493-99.

CURRO, D. The Modulation of Potassium Channels in the Smooth Muscle as a

Therapeutic Strategy for Disorders of the Gastrointestinal Tract. In: DONEV, R.

Page 157: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

154

editors. Advances in Protein Chemistry and Structural Biology. Elsevier: Vol.

104, 2016. P. 263-305.

DAENAS, C., HATZIEFTHIMIOU, A., GOURGOULIANIS, K., MOLYVDAS, P. A.

2006. Azithromycin has a direct relaxant effect on precontracted airway smooth muscle.

Eur J Pharm, 553, 280-7.

DAVIDSON, R. J. 2019. In vitro activity and pharmacodynamic/pharmacokinetic

parameters of clarithromycin and azithromycin: why they matter in the treatment of

respiratory tract infections. Infect Drug Resist, 12, 585–96.

DE OLIVEIRA, I. S., CROSARA, P. F. T. B., CASSALI, G. D., REIS, D. C., DE

RESENDE, C. B., NUNES, F. B., GUI MARAES, R. E. S. 2016. Evaluation of the

improvement of quality of life with azithromycin in the treatment of eosinophilic nasal

polyposis. Braz J Otorhinolaryngol, 82, 198-202.

DEITEREN, A., DE-MAN, J. G., PELCKMANS, P. A., DE WINTER, B. Y. 2015.

Histamine H4 receptors in the gastrointestinal tract. Br J Pharmacol, 172, 1165–78.

DELOOSE, E., VOS, R., JANSSEN, P., BERGH, O. V. D., OUDENHOVE, L. V.,

DEPOORTERE, I., TACK, J. 2016 The motilin receptor agonist erythromycin

stimulates hunger and food intake through a cholinergic pathway. Am J Clin Nutr, 103,

730-37.

DIAS, D. M., COSTA, N. M. B., NUTTI, M. R., TAKO, E. & MARTINO, H. S. D.

2018. Advantages and limitations of in vitro and in vivo methods of iron and zinc

bioavailability evaluation in the assessment of biofortification program effectiveness.

Crit Rev Food Sci Nutr, 58, 2136-2146.

DIETERICH, W., SCHINK, M., ZOPF, Y. 2018. Microbiota in the Gastrointestinal

Tract. Med Sci, 6(4), 116.

DONDE, S., MISHRA, A., KOCHHAR, P. 2014. Azithromycin in acute bacterial

upper respiratory tract infections: An Indian non-interventional study. Indian J

Otolaryngol Head Neck Surg, 66, 225–30.

Page 158: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

155

EHLERT, F. J., PAK, K. J., GRIFFIN, M. T. 2012. Muscarinic agonists and

antagonists: Effects on gastrointestinal function. Handb Exp Pharmacol, 208, 343-74.

EL-BAKI, Z. A. A., YOUSSEF, F. A., OUDA, E. A., EL-HAKAM, A. F. Z. 2015.

Pharmacological effects of azithromycin on different isolated smooth muscle

preparations of experimental animals. Al-Azhar Med. J, 44, 114-23.

ELKATTAN, A. E., GHEITH, M., FAYED, M. S., EL-YAZEED, M. B., FARRAG,

A. R. H., KHALI, W. K. B. 2019. Effects of different parameters of diode laser on

acceleration of orthodontic tooth movement and its effect on relapse: An experimental

animal study. Maced J Med Sci,7, 412–20.

ESTEVES, P. J., ABRANTES, J., MAGE, R. 2018. The wide utility of rabbits as

models of human diseases. Exp. Mol. Med, 50, 1-10.

EVANS, E. D., MANGEL, A. W. 2011. Depolarization-stimulated contractility of

gastrointestinal smooth muscle in calcium-free solution: a review. Gastroenterol, 2011,

692528.

FABISIAK, A., WŁODARCZYK, J., FABISIAK, N., STORR, M., FICHNA1, J. 2017.

Targeting histamine receptors in irritable bowel syndrome: A critical appraisal. J

Neurogastroenterol Motil, 23, 341-48.

FAISAL, M. S., ALI, N., ALI, K. 2018. Citrullus colocynthis; fractionation of

methanolic extract of citrullus colocynthis for spasmogens. Professional Med J, 25, 96-

103.

FLEMING, M. A., EHSAN, L., MOORE, S. R. & LEVIN, D. E. 2020. The Enteric

Nervous System and Its Emerging Role as a Therapeutic Target. Gastroenterol Res

Pract, 8024171.

FREEMAN, W. H. Neuroscience. 6th ed. Elsevier; 2018.

FUENTES, I. M., CHRISTIANSON, J. A. 2016. Ion channels, ion channel receptors,

and visceral hypersensitivity in irritable bowel syndrome. Neurogastroenterol Motil.

28, 1613–18.

Page 159: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

156

FUKUI, H., XU, XIN., MIWA, H. 2018. Role of Gut Microbiota-Gut Hormone Axis

in the Pathophysiology of Functional Gastrointestinal Disorders. J Neurogastroenterol

Motil, 24(3): 367-86.

FURNESS, J. B., CALLAGHAN, B. P., RIVERA, L. R., CHO, H. J. 2014. The enteric

nervous system and gastrointestinal innervation: integrated local and central control.

Adv Exp Med Biol, 817:39-71.

GAO, X., ZHAO, Y., SU, Y., LIU, K. U. N., YU, X., CUI, C., YANG, Z., SHI, Y.,

JING, X., ZHU, B. 2016. β1/2 or M2/3 receptors are required for different

gastrointestinal motility responses induced by acupuncture at heterotopic or homotopic

acupoints. PLOS ONE, 11, e0168200.

GERBER, J. S. & ZAOUTIS, T. E. 2017. Azithromycin Prophylaxis for Laboring

Mothers. Pediatrics, 139, e20163643.

GHAYUR, M. N., GILLANI, A. H., JANSSEN, L. J. 2012. Intestinal, airway, and

cardiovascular relaxant activities of thymoquinone. J Evid Based Complementary

Altern Med, 1-13.

GHATTAMANENI, N. K., PANCHAL, S. K. & BROWN, L. 2019. An improved rat

model for chronic inflammatory bowel disease. Pharmacol Rep, 71, 149-155.

GHOSHAL, U. C., SHUKLA, R., GHOSHAL, U., GWEE, K. A., NG, S. C.,

QUIGLEY, E., M. 2012. The Gut Microbiota and Irritable Bowel Syndrome: Friend or

Foe? International Journal of Inflammation, 2012, 151085.

GIUDICESSI, J. R., ACKERMAN, M. J., CAMILLERI M. 2018. Cardiovascular

safety of prokinetic agents: A focus on drug-induced arrhythmias. Neurogastroenterol

Motil, 30: e13302.

GIULIETTI, M., VIVENZIO, V., PIVA, F., PRINCIPATO, G., BELLANTUONO,

C., NARDI, B. 2014. How much do we know about the coupling of G-proteins to

serotonin receptors? Mol Brain, 7, 143-48.

GORDIENKO, D.V., HARHUN, M. I., KUSTOV, M. V., PUCOVSKY, V., Bolton, T.

B. 2008. Sub-plasmalemmal [Ca2+]i upstroke in myocytes of the guinea-pig small

Page 160: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

157

intestine evoked by muscarinic stimulation: IP3R-mediated Ca2+ release induced by

voltage-gated Ca2+ entry. Cell Calcium, 43, 122-41.

GOSHAL, U. C. 2010. Pharmacotherapy for Gastroparesis: An attempt to evaluate a

safer alternative. J Neurogastroenterol Motil, 16, 350–52.

GOYER, I., FERLAND, G., RUO, N., MORIN, C., BROCHET, M. S., MORIN,

L., FERREIRA, E. 2016. Impact of azithromycin on pregnancy prolongation in women

at risk of preterm labor: A time-to-event analysis. J Popul Ther Clin Pharmacol, 23,

e183-e92.

GRASA, L., REBOLLAR, E., ARRUEBO, M. P., PLAZA, M. A., MURILLO, M. D.

2004. The role of Ca2 in the contractility of rabbit small intestine in vitro. J Physiol

Pharmacol, 55, 639–50.

GUO, Y., ZHANG, Y., HUANG, W., SELWYN, F. P., KLAASSEN, C. D. 2016.

Dose-response effect of berberine on bile acid profile and gut microbiota in mice. BMC

Complement Altern Med, 16, 1-12.

HAFEN, B. B., BURNS, B. Physiology, smooth muscles. Statpearls.

HEDRICH, H. J. The use of laboratory mouse. Editor(s): HOITINGA, M. R.,

MIKKELSON, L. F. 2nd edition. Science direct: 2012.

ISLAMI, H., BEXHETI, S., AHMETAJ, H., ŠUKALO, A., MANXHUKA, S.,

NURAJ, B., KAMBERI, X., QORRAJ, H., KASHTRATI, B., DESHA, M. 2009.

Action of propranolol in the reaction of smooth musculature of tracheal rings induced

with acetyl- choline, histamine, serotonin (5-ht) and prostaglandin (pgf2_alfa) in vitro

and in vivo. Bosn J Basic Med Sci, 9, 142–47.

ISOLA, S., ADAMS. N. 2020. Metoclopramide. Stat Pearls, 15-20.

ISRAELYAN, N., COLLE, A. D., LI, Z., PARK, Y., XING, A., JACOB, P. R. et al.

2019. Effects of serotonin and slow release 5-Hydroxytryptophan on gastrointestinal

motility in a mouse model of depression. Gastroenterol, 157, 507-21.

JAFARI, J., YAZAKI, E., WOODLAND, P., WONG, T., ANGGIANSAH, A.,

SIFRIM, D. 2015. Effect of azithromycin on esophageal hypomotility (EH) and

Page 161: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

158

prediction of response by esophageal stimulations tests during high resolution

manometry. Gastroenterol. 148, 70-75.

JANBAZ, K. H., HASSAN, W., MEHMOOD, M. H., GILANI, A. H. 2015.

Antidiarrheal, antispasmodic and bronchodilator activities of Pistacia integerrima are

mediated through dual inhibition of muscarinic receptors and Ca++ influx. Sci.

Technol. Dev, 34, 52-59.

JELIC, D., ANTOLOVIC, R. 2016. From erythromycin to azythromycin and new

potential ribosome-binding antimicrobials. Antibiotics, 5, 1-13.

JIMSHELEISHVILI, S. MARWAHA, K. 2019. Physiology, Neuromuscular

Transmission. StatPearls.

JODAT, Y. A., KANG, M. G., KIAEE, K., KIM, G. J., MARTINEZ, A. F.,

ROSENKRANZ, A., BAE, H. & SHIN, S. R. 2018. Human-derived organ-on-a-chip

for personalized drug development. Curr. Pharm. Des, 24, 5471-5486.

JOHN. Azithromycin. [Online]. Available:

https://www.dreamstime.com/azithromycin-molecular-structure-isolated-black-azithro

omycin-antibiotic-useful-treatment-number-bacterial-image121265297 [Accessed 10-

8-2019].

JONGE, W. J. 2013. The Gut's Little Brain in Control of Intestinal Immunity. ISRN

Gastroenterol. 2013: 630159.

JUURINK, D. N. 2014. The cardiovascular safety of azithromycin. CMAJ. 186: 1127–

28.

KAGKELARIS, K. A., MAKRI, O. E., GEORGAKOPOULOS, C. D.,

PANAYIOTAKOPOULOS, G. D. An eye for Azithromycin: review of the literature.

Ther Adv Ophthalmol. 2018; 10: 2515841418783622.

KAPALKA, G. M. 2009. Nutritional and herbal therapies for children and adolescents:

A handbook for mental health clinicians, Academic Press.

Page 162: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

159

KAMEL, E. M., SHABAYEK, H. A., ALY, F. A., ELGARHI, R. I. 2015. The

prokinetic effect of itopride, a comparative study with metoclopramide. Az. J. Pharm

Sci, 52, 18-30.

KANASAKI, K. & KOYA, D. 2011. Biology of obesity: lessons from animal models

of obesity. BioMed Research International, 2011.

KANOH, S., RUBIN, B. K. 2010. Mechanisms of action and clinical application of

macrolides as immunomodulatory medications. Clin Microbiol Rev, 23, 590-615.

KATO, S., TAKAHASHI, A., SHINDO, M., YOSHIDA, A., KAWAMURA, T.,

MATSUMOTO, K., MUTSUURA, B. 2019. Characterization of the gastric motility

response to human motilin and erythromycin in human motilin receptor-expressing

transgenic mice. PloS ONE, 14, e0205939.

KIM, H., DWYER, L., SONG, J. H., MARTIN-CANO, F. E., BAHNE, J., PERI, L.,

BRITTON, F. C., SANDERS, K. M., KOH, S. D. 2012. Identification of histamine

receptors and effects of histamine on murine and simian colonic excitability.

Neurogastroenterol Motil. 23, 949–e409.

KIM, J. G., LEEM, Y.-E., KWON, I., KANG, J.-S., BAE, Y. M. & CHO, H. 2018.

Estrogen modulates serotonin effects on vasoconstriction through Src inhibition. Exp

Mol Med, 50, 1-9.

KINDIG, D. M., GRIDER, J. R. 2015. Serotonin and colonic motility.

Neurogastroenterol Motil. 27, 899–905.

KNIGHT, J., BAYRAM-WESTON, Z., NIGAM, Y. 2019. Gastrointestinal tract 6: the

effects of gut microbiota on human health. Nurs Times, 115, 46-50.

KONG, F. Y. S., RUPASINGHE, T. W., SIMPSON, J. A., VODSTRCIL, L. A.,

FAIRLEY, C. K., MCCONVILLE, M. J., MCCONVELLI M. J., HOCKING, J. S.

2017. Pharmacokinetics of a single 1g dose of azithromycin in rectal tissue in men.

PLoS ONE, 12, e0174372.

Page 163: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

160

KORIMILLI, A., PARKMAN, H. P. 2010. Effect of Atilmotin, a Motilin Receptor

Agonist, on Esophageal, Lower Esophageal Sphincter, and Gastric Pressures. Dig Dis

Sci, 55, 300–06.

KUO, I. Y., EHRLICH, B. E. 2015. Signaling in Muscle Contraction. Cold Spring Harb

Perspect Biol. 7, a006023.

KURLE, D. G., BARGAJE, R. S., DESHMUKH, Y. A. 2017. Study of effect of

erythromycin base on gastrointestinal smooth muscles in laboratory animals in-vitro.

Int J Pharm Sci Res, 3, 331-45.

LARSON, J. M., TAVAKKOLI, A., DRANE, W. E., TOSKES, P. P., MOSHIREE, B.

2010. Advantages of azithromycin over erythromycin in improving the gastric

emptying half-time in adult patients with gastroparesis. J Neurogastroenterol Motil, 16,

407-13.

LAU, M. L. M., ROBINSON, P. D., FLANK, J., SUNG, L., DUPUIS, L. L. 2016. The

Safety of metoclopramide in children: a systematic review and meta-analysis. Drug

Saf, 39, 675–87.

LE, H. T., LIE, K. K., GIROUD-ARGOUD, J., RØNNESTAD, I. & SÆLE, Ø. 2019.

Effects of cholecystokinin (CCK) on gut motility in the stomachless fish ballan wrasse

(Labrus bergylta). Front Neurosci, 13, 553.

LI, D. H., HUANG, X., GUO, X., MENG, X. M., WU, Y. S., LU, H. L, ZHANG C.

M., KIM, Y. C., XU, W. X. 2014. Voltage dependent potassium channel remodeling in

murine intestinal smooth muscle hypertrophy induced by partial obstruction. Voltage

dependent potassium channel remodeling in murine intestinal smooth muscle

hypertrophy induced by partial obstruction. PLoS ONE, 9, e86109.

LI, F., HUANG, J., JI, D., MENG, Q., WANG, C., CHEN, S. et al. 2017. Azithromycin

effectively inhibits tumor angiogenesis by suppressing vascular endothelial growth

factor receptor 2-mediated signaling pathways in lung cancer. Oncol Lett., 14, 89-96.

LI, Z. S., SCHMAUSS, C., CUENCA, A., RATCLIFFE, E., GERSHON, M. D. 2006.

Physiological modulation of intestinal motility by enteric dopaminergic neurons and

Page 164: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

161

the D2 receptor: analysis of dopamine receptor expression, location, development, and

function in wild-type and knock-out mice. J Neurosci, 8, 2798-807.

LIPSCOMBE, D., TORO, C. P. Biophysics of Voltage-Gated Ion Channels. In:

BYRNE, J. H., HEIDELBERGER, R., WAXHAM, M. N. editors. From molecules to

networks. Academic Press; 2014 p. 377-407.

LITOU, C., EFFINGER, A., KOSTEWICZ, E. S., BOX, K. J., FOTAKI, N. &

DRESSMAN, J. B. 2019. Effects of medicines used to treat gastrointestinal diseases on

the pharmacokinetics of coadministered drugs: a PEARRL Review. J Pharm

Pharmacol, 71, 643-673.

Liu H, Farley JM. 2005. Effects of first and second generation antihistamines on

muscarinic induced mucus gland cell ion transport. BMC Pharmacol, 24;5:1079883.

Lordfred. Conformations of Voltage-Gated Potassium Channels. 2015. Memorang.

MAPARA, M., THOMAS, B. S. & BHAT, K. 2012. Rabbit as an animal model for

experimental research. J Dent Res, 9, 111.

MARCHANT, J. M., PETSKY, H. L., MORRIS, P. S., CHANG, A. B. 2018.

Antibiotics for prolonged wet cough in children. Cochrane Database Syst Rev, 31,

CD004822.

MARTIN, E., HINE, R. A dictionary of biology. 7th ed. Oxford: UK, 2015.

MATZNELLER, P., KRASNIQI, S., ZEITLINGER, M. 2013. Blood, tissue, and

intracellular concentrations of azithromycin during and after end of therapy.

Antimicrob Agents Chemother, 57, 1736-42.

MCCORVY, J. D., ROTH, B. L. 2015. Structure and Function of Serotonin G protein

Coupled Receptors. Pharmacol Ther, 150, 129–42.

MCMULLAN, B. J., MOSTAGHIM, M. 2015. Prescribing azithromycin. Aust Prescr,

38, 87–89.

MERTENS, V., BLONDEAU, K., PAUWELS, A., FARRE,

R., VANAUDENAERDE, B., VOS, R., VERLEDEN, G., RAEMDONCK, D. E. V.,

Page 165: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

162

DUPONT, L. J., SIFRIM, D. 2009. Azithromycin Reduces Gastroesophageal Reflux

and Aspiration in Lung Transplant Recipients. Dig Dis Sci, 54, 972-9.

MERTENS, V., BLONDEAU, K., VAN, O. L., VANAUDENAERDE, B,, VOS,

R., FARRE, R., PAUWELS, A., VERLEDEN, G., RAEMDONCK, D. E. V., SIFRIM,

D., DUPONT, L. J. 2011. Bile acids aspiration reduces survival in lung transplant

recipients with BOS despite azithromycin. Am J Transplant, 11, 329-35.

MIKAMI, H., ISHIMURA, N., FUKAZAWA, K., OKADA, M., IZUMI,

D., SHIMURA, S., OKIMOTO, E., AIMI, M., ISHIHARA, S., KINOSHITA, Y. 2016.

Effects of Metoclopramide on Esophageal Motor Activity and Esophagogastric

Junction Compliance in Healthy Volunteers. J Neurogastroenterol Motil. 22, 112–17.

MISHRA, A., SINGH, S., SHUKLA, S. 2018. Physiological and functional basis of

dopamine receptors and their role in neurogenesis: Possible implication for Parkinson’s

disease. J Exp Neurosci, 31, 1179069518779829.

MITTAL, R., DEBS, L. H., PATEL, A. P, NGUYEN, D., PATEL, P., CONNOR, G.,

et al. 2017. Neurotransmitters: The critical modulators regulating gut-brain axis. J Cell

Physiol, 232, 2359–72.

MIZUTA, K., ZHANG, Y., XU, D., MIZUTA, F., OVIDIO, F. D., MASAKI,

E., EMALA, C. W. 2013. The dopamine D1 receptor is expressed and facilitates

relaxation in airway smooth muscle. Respir Res. 14, 89-94.

MOLNAR, C., GAIR, J. Concepts of Biology-First Canadian edition. Press Books.

MONCZOR, F., FERNANDEZ, N. 2016. Current knowledge and perspectives on

histamine H1 and H2 receptor Pharmacology: functional selectivity, receptor crosstalk,

and repositioning of classic histaminergic ligands. Mol Pharmacol, 90, 640-48.

MONTGOMERY, L. E., TANSEY, E. A., JOHNSON, C. D., ROE, S. M., QUINN, J.

G. 2016. Autonomic modification of intestinal smooth muscle contractility. Adv

Physiol Educ. 40:104–09.

Page 166: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

163

MORAN, Y., BARZILAI, M. G., LIEBESKIND, B. J., ZAKON, H. H. 2015.

Evolution of voltage-gated ion channels at the emergence of Metazoa. J. Exp.

Biol. 218, 515-25.

MORRIS, A. D., CHEN, J., LAU, E., POH, J. 2016. Domperidone-associated QT

interval prolongation in non-oncologic pediatric patients: a review of the literature. Can

J Hosp Pharm, 69, 224–30.

MORSE, K. L., BEHAN, J., LAZ, T. M., WEST, R. E. J., GREENFEDER SA,

ANTHES, J. C. et al. 2001. Cloning and characterization of a novel human histamine

receptor. J Pharmacol Exp Ther, 296, 1058–66.

MOSHIREE, B., MCDONALD, R., HOU, W., TOSKES, P. P. 2010. Comparison of

the effect of azithromycin versus erythromycin on antroduodenal pressure profiles of

patients with chronic functional gastrointestinal pain and gastroparesis. Dig Dis Sci, 55,

675-83.

NASERI, M. K. G., YAHYAVI, H., ARABIAN, M. 2008. Antispasmodic activity of

onion (Allium cepa L.) peel extract on rat ileum. Iran J Pharm Res, 7, 155-59.

NATH, S. K., DAS, S., KAR, J., AFRIN, K., DASH, A. K., AKTER, S. 2016.

Topographical and biometrical anatomy of the digestive tract of white Newzealand

rabbit (Oryctolagus cuniculus). J. Adv Vet. Ani. Res. 3, 145-51.

NESHATIAN, L., STREGE, P. R., RHEE, P. L., KRAICHEL, Y. R. E, MAZZONE,

A., BERNARD, C. E. 2015. Ranolazine inhibits voltage-gated mechanosensitive

sodium channels in human colon circular smooth muscle cells. Am J Physiol

Gastrointest Liver Physiol, 309, 372-87.

NEZAMI, B. G., SRINIVASAN, S. 2010. Enteric nervous system in the small intestine:

pathophysiology and clinical implications. Curr. Gastroenterol Rep, 12(5), 358–365.

NGUYEN, N. 2014. Controversies in using IV azithromycin to treat gastroparesis.

Gastroenterol, 39, 13-17.

Page 167: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

164

NOWLAND, M. H., BRAMMER, D. W., GARCIA, A., RUSH, H. G. In: Fox JG,

Anderson LC, Whary MT editors. Biology and Diseases of Rabbits. Laboratory Animal

Medicine. Academic Press; 2015, p. 411-61.

OHNO, T., MOCHIKI, E. & KUWANO, H. 2010. The roles of motilin and ghrelin in

gastrointestinal motility. Int J. Pep, 820794.

OLDENBURG, C. E., ARZIKA, A. M., MALIKI, R., KANE, M. S., LEBAS, E., RAY,

K. J. et al. 2018. Safety of azithromycin in infants under six months of age in Niger: A

community randomized trial. PloS Negl Trop Dis, 12, e0006950.

PACHECO, M. A. Receptor types and sub-types. In: X-Pharm: The comprehensive

Pharmacology reference. editors. ENNA, S. J., BYLUND, D. B. Elsevier, 2008.

PADDA, M. S., SANCHEZ, M., AKHTAR, A. J., BOYER, J. L. 2011. Drug-induced

cholestasis. Hepatol, 53(4), 1377–87.

PARSONS, M. E., GANELLIN, C. R. 2006. Histamine and its receptors. Br J

Pharmacol, 147(1), S127–S135.

PARTHASARATHY, G., RAVI, K., CAMILLERI, M., ANDREWS, C., SZARKA, L.

A., LOW, P. A. et al. 2015. Effect of neostigmine on gastroduodenal motility in patients

with suspected gastrointestinal motility disorders. Neurogastroenterol Motil, 27, 1736–

46.

PEDIREADDY, M. K. R. 2011. In vitro evaluation techniques for gastrointestinal

motility. Indian J Pharm Educ Res, 45, 184-91.

PIACENTINI, G. L., PERONI, D. G., BODINI, A., PIGOZZI, R., COSTELLA,

S., LOIACONO, A., BONER, A. L. 2007. Azithromycin reduces bronchial

hyperresponsiveness and neutrophilic airway inflammation in asthmatic children: a

preliminary report. Allergy Asthma Proc, 28, 194-8.

POTTER, T. G., SNIDER, K. R. 2013. Azithromycin for the treatment of gastroparesis.

Ann Pharmacother, 47, 411-5.

PROSAPIO, J. G., SANKAR, P. & JIALAL, I. 2020. Physiology, Gastrin. StatPearls.

Page 168: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

165

PURVES, D., AUGUSTINE, G. J., FITZPATRICK, D. Neuroscience. 3rd ed. Elsevier;

2014.

QUINGLEY, E. M. M. 2015. Prokinetics in the management of functional

gastrointestinal disorders. J Neurogastroenterol Motil, 21, 330-36.

RADU, B. M., OSCULATI, A. M. M., SUKU, E., BANCIU, A., TSENOV, G.,

MERIGO, F. 2017. All muscarinic acetylcholine receptors (M1-M5) are expressed in

murine brain microvascular endothelium. Sci Rep, 7, 5083-90.

RADULOVIC, M., ANAND P., KORSTEN M. A., GONG, B. Targeting Ion

Channels: An important therapeutic implication in gastrointestinal dysmotility in

patients with spinal cord injury. J Neurogastroenterol Motil. 2015; 21(4): 494–502.

RAMOS, F. L., LINDBERG, S. M., KRAHNKE, J. S., CONNETT, J., ALBERT, R.

K., CRINER, G. J. 2014. Azithromycin and COPD exacerbations in the presence or

absence of symptoms or active treatment for gastroesophageal reflux. J COPD F, 1,

221-28.

RAO, G. A., MANN, J. R., SHOAIBI, A., BENNETT, C. L., NAHHAS, G., SUTTON,

S. S. et al. 2014. Azithromycin and Levofloxacin use and increased risk of cardiac

arrhythmia and death. Ann Fam Med, 12, 121–27.

RAO, M., GERSHON, M. D. 2016. The bowel and beyond: the enteric nervous system

in neurological disorders. Nat Rev Gastroenterol Hepatol. 13, 517–28.

Raja S., Batra V., Srinivasan S. The Influence of Microbiota on Gastrointestinal

Motility. In: Sun J., Dudeja P. editors. Mechanisms Underlying Host-Microbiome

Interactions in Pathophysiology of Human Diseases. Physiology in Health and Disease.

Springer, Boston, 2018.

REDDY, B. S., VIJAYAKUMAR, G., BALASUBRAMANIAM, G. A.,

SIVARAMAN, S., KATHIRVEL, S. 2018. Evaluation of efficacy of neostigmine and

azithromycin as motility modifiers in the medical management of functional ileus in

twenty cows - A pilot study. Indian Vet. J, 95, 47-50.

Page 169: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

166

REYNOLDS, J. 2016. Small‐Intestinal Hormones and Neurotransmitters. Practical

Gastroenterology and Hepatology Board Review Toolkit, 181-186.

RICHARDS, R. D., DAVENPORT, K., MCCALLUM, R. W. 2009. The treatment of

idiopathic and diabetic gastroparesis with acute intravenous and chronic oral

erythromycin. Am J Gastroenterol, 88, 203-07.

RIEHLE, C., BAUERSACHS, J. 2019. Small animal models of heart failure.

Cardiovasc Res, 115, 1838–49.

ROCHA, H. A. C., DANTAS, B. P. V., ROLIM, T. L., COSTA, B. A., DE

MEDEIROS, A. C. 2014. Main ion channels and receptors associated with visceral

hypersensitivity in irritable bowel syndrome. Ann Gastroenterol, 27, 200-06.

ROMERO, C. M., FASS, R. 2014. New and Future Drug Development for

Gastroesophageal Reflux Disease. J Neurogastroenterol Motil. 20, 6–16.

ROSENBAUM, D. M., RASMUSSEN, S. G., & KOBILKA, B. K. 2009. The structure

and function of G-protein-coupled receptors. Nature, 459(7245), 356–63.

SALLAM, H. S., CHEN, J. D. Z. 2010. The Prokinetic Face of Ghrelin. Int J Pept, 8,

493614.

SEIDL, H., GUNDLING, F., PFEIFFER, A., PEHL, C., SCHEPP, W., SCHMIDT, T.

2012. Comparison of small-bowel motility of the human jejunum and ileum.

Neurogastroenterol Motil, 24, 373-80.

SEMENOV, I., BRENNER, R. 2018. Voltage effects on muscarinic acetylcholine

receptor‐mediated contractions of airway smooth muscle. Physiol Rep. 6, 1-10.

SENGUPTA, P. 2013. The laboratory rat: relating its age with human's. Int J Prev Med,

4, 624.

SHAKIR, A. K., ALTAF, M. A. 2018. Azithromycin induces migrating motor

complexes in pediatric patients undergoing antroduodenal motility studies. J Pediatr

Pharmacol Ther. 5, 390-94.

Page 170: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

167

SHTEINBERG, M., SCHNEER, S., LAVON, O., ADIR, Y. 2016. Long term treatment

with macrolides in chronic lung diseases. Harefuah, 155, 567-71.

SIFRIM, D., MATSUO, H., JANSSENS, J., VANTRAPPEN, G. 1994. Comparison of

the effects of midecamycin acetate and azithromycin on gastrointestinal motility in

man. Drugs Exp Clin Res, 20, 121-6.

SINOUSSI, B. F., MONTAGUTELLI, X. 2015. Animal models are essential to

biological research: issues and perspectives. Fut Sci OA,1, FSO63.

SOOMER, B., SOTO, E. F., AVILA, G. G. 2017. Cellular Na+ handling mechanisms

involved in airway smooth muscle contraction (Review). Int J Mol Med, 40, 3–9.

SPEER, E. M., DOWLING, D. J., XU, J., OZOG, L. S., MATHEW, J. A., CHANDER,

A. et al. 2018. Pentoxifylline, dexamethasone and azithromycin demonstrate distinct

age-dependent and synergistic inhibition of TLR- and inflammasome-mediated

cytokine production in human newborn and adult blood in vitro. PLoS ONE, 13,

e0196352.

SRIVSTAVA, S., RATH, S. K. Principles for in vitro toxicology. In: DHAWAN, A.,

KWON, S. editors. In vitro toxicology. Academic Press; 2018, p. 21-43.

STEVENS, L., RODIN I. Introduction to drug treatments. Editor(s): LESLEY, S., IAN

R. Psychiatry, 2nd edition. Churchill Livingstone: 2011.

SUNDARAM, V., BJORNSSON, E. S. 2017. Drug induced cholestasis. Hepatol, 1,

726-35.

SURYANARAYANAN, A. Acetylcholine. In: WEXLER P. Editor. Encyclopedia of

Toxicology. Academic Press; 2014. P 49-50.

SUTERA, L., DOMINGUEZ, L. J., BELVEDERE, M., PUTIGNANO, E.,

VERNUCCIO, L., FERLISI, A., FAZIO, G., COSTANZA, G., BARBAGALLO, M.

2008. Azithromycin in an older women with diabetic gastroparesis. Am. J. Ther, 15,

85-88.

SZPIRER, C. 2010. Cancer research in rat models. Rat Genomics. Springer.

Page 171: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

168

TERRY, N., MARGOLIS, K. G. 2016. Serotonergic mechanisms regulating the GI

tract: Experimental evidence and therapeutic relevance. Handb Exp Pharmacol. 239,

319–42.

THANGAM, E. B., JEMIMA, E. A., SINGH, H., BAIG, M. S., KHAN,

M., MATHIAS, C. B., CHURCH, M. K., SALUJA, R. 2018. The role of histamine and

histamine receptors in mast cell-mediated allergy and inflammation: The hunt for new

therapeutic targets. Front Immunol, 9, 1873-76.

TILMAN, E. M., SMETANA, K. S., BANTU, L., BUCKLEY, M. G. 2016.

Pharmacologic treatment for pediatric gastroparesis: A review of the literature. J Pediatr

Pharmacol Ther. 21, 120–32.

TIMOUMI, S., MANGIN, D., PECZALSKI, R., ZAGROUBA, F. & ANDRIEU, J.

2014. Stability and thermophysical properties of azithromycin dihydrate. Arab. J.

Chem, 7, 189-195.

TONINI, M., CIPOLLINA, L., POLUZZI, E., CREMA, F., CORAZZA, G. R., PONTI,

F. D. 2004. Clinical implications of enteric and central D2 receptor blockade by

antidopaminergic gastrointestinal prokinetics. Alimen Pharm Ther, 19, 379-90.

TRIBBLE, D. R. 2017. Antibiotic therapy for acute watery diarrhea and dysentery. Mil

Med, 182, 17–25.

TOMISIC, Z. B. 2011. The Story of Azithromycin. J Chemists Chem Eng, 60(12):603-

617.

UESAKA, T., YOUNG, H. M., PACHNIS, V. & ENOMOTO, H. 2016. Development

of the intrinsic and extrinsic innervation of the gut. Dev. Biol, 417, 158-167.

UGWU, A. C., AGWU, K. K., UMEH, E. C. 2013. Comparison of the effect of

azithromycin versus erythromycin on gallbladder motility: A sonographic study. Afr J

Pharm. Pharmacol, 7, 2348-51.

VALDES, A. M., WALTER, J., SEGAL, E. 2018. Role of gut microbiota in nutrition

and health. BMJ, 361:k2179.

Page 172: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

169

VDOVIAKOVA, K., PETROVOVA, E., MALOVESKA, M., KRESAKOVA,

L., TELEKY, J., ELIAS, M. Z. J., PETRASOVA. 2016. Surgical anatomy of the

gastrointestinal tract and its vasculature in the laboratory rat. Gastroenterol Res Pract.

3, 1-11.

WANG, Q., YU, M. F., ZHANG, W. J., LIU, B. B., ZHAO, Q. Y., LUO X. et al. 2019.

Azithromycin inhibits muscarinic 2 receptor‐activated and voltage‐activated Ca2+

permeant ion channels and Ca2+ sensitization, relaxing airway smooth muscle

contraction. Clin Exp Pharmacol Physiol, 7, 13062.

WASEEM, S., MOSHIREE, B., DRAGANOV, P. V. 2009. Gastroparesis: Current

diagnostic challenges and management considerations. World J Gastroenterol, 15, 25-

37.

WEISBRODT, N. W. Regulation: nerves and smooth muscle. In: Gastrointestinal

Physiology, editor JOHNSON, L. R. Philadelphia, PA: Mosby Elsevier, 2007, p. 13–

21.

WELCHKO, R. M., LÉVÊQUE, X. T. & DUNBAR, G. L. 2012. Genetic rat models of

Parkinson's disease. Parkinson's Dis, Article ID 128356

WERAWATGANON, D. 2014. Simple animal model of Helicobacter pylori infection.

World J. Gastroenterol: WJG, 20, 6420.

WHITEHEAD, K. M., CORTES, Y., EIRMANN, L. 2016. Gastrointestinal dysmotility

disorders in critically ill dogs and cats. J Vet Emer Crit Care, 26, 234–53.

XUE, R., ZHANG, H., PAN, J., DU, Z., ZHOU, W., ZHANG, Z. et al. 2018. Peripheral

dopamine controlled by gut microbes inhibits invariant natural killer T cell-mediated

hepatitis. Front. Immunol, 9, 2398-2404.

YASIN, N. A. Z., EL-ROKH, S. M., EL-SHENAWY, S. M. A, IBRAHIM, B. M. M.

2012. The study of the antispasmodic effect of Ginger (Zingiber officinale) in vitro.

Der Pharmacia Lettre, 4, 263-74.

Page 173: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

170

ZABIROWICZ, E. S., GAN, T. J. Pharmacology of postoperative nausea and vomiting.

In: HEMMINGS, H. C., EGAN, T. D. editors. Pharmacology and physiology for

anesthesia. Elsevier; 2019. P 671-92.

ZHAG, LU., SHI, G. 2016. Gq-coupled receptors in autoimmunity. J. Immunol Res.

Article ID 3969023.

ZHANG, X.C., YANG, H., LIU, Z., SUN, F. 2018. Thermodynamics of voltage-gated

ion channels. Biophys Rep, 4, 300–19.

ZHU, Y., MICHALOVICH, D., WU, H., TAN, K. B., DYTKO, G. M., MANNAN, I.

J. et al. 2001. Cloning, expression, and pharmacological characterization of a novel

human histamine receptor. Mol Pharmacol. 59, 434–41.

ZIEGLER, A., GONZALEZ, L. & BLIKSLAGER, A. 2016. Large animal models: the

key to translational discovery in digestive disease research. Cell. Mol Gastroenterol

Hepatol, 2, 716-724.

ZIMMERMANN, P., ZIESENITZ, V. C., CURTIS, N., RITZ, N. 2018. The

immunomodulatory effects of macrolides-A systematic review of the underlying

mechanisms. Front Immunol, 9, 302-06.

Page 174: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

171

Published Paper

Page 175: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

172

BITAR, K. N. 2003. Function of gastrointestinal smooth muscle: from signaling to contractile proteins. The American journal of medicine, 115, 15-23.

BRANDLER, J., MILLER, L. J., WANG, X. J., BURTON, D., BUSCIGLIO, I., ARNDT, K., HARMSEN, W. S. & CAMILLERI, M. 2020. Secretin effects on gastric functions, hormones and

symptoms in functional dyspepsia and health: randomized crossover trial. American Journal of Physiology-Gastrointestinal and Liver Physiology, 318, G635-G645.

BRYDA, E. C. 2013. The Mighty Mouse: the impact of rodents on advances in biomedical research. Missouri medicine, 110, 207.

CASTAÑEDA-LOPEZ, M. E., GARZA-VELOZ, I., ORTIZ-RODRIGUEZ, J. M., CASTAÑEDA-MIRANDA, R., SOLIS-SANCHEZ, L. O., VEGA-CARRILLO, H. R., DEL ROSARIO

MARTINEZ-BLANCO, M., TREJO-VAZQUEZ, F., ORNELAS-VARGAS, G. & RODRIGUEZ-SANCHEZ, I. P. 2017. Animal Models of Rheumatoid Arthritis. Experimental Animal

Models of Human Diseases-An Effective Therapeutic Strategy. IntechOpen. DIAS, D. M., COSTA, N. M. B., NUTTI, M. R., TAKO, E. & MARTINO, H. S. D. 2018. Advantages

and limitations of in vitro and in vivo methods of iron and zinc bioavailability evaluation in the assessment of biofortification program effectiveness. Critical

reviews in food science and nutrition, 58, 2136-2146. ESTEVES, P. J., ABRANTES, J., BALDAUF, H.-M., BENMOHAMED, L., CHEN, Y., CHRISTENSEN,

N., GONZÁLEZ-GALLEGO, J., GIACANI, L., HU, J. & KAPLAN, G. 2018. The wide utility of rabbits as models of human diseases. Experimental & molecular medicine, 50, 1-

10. GHATTAMANENI, N. K., PANCHAL, S. K. & BROWN, L. 2019. An improved rat model for

chronic inflammatory bowel disease. Pharmacological Reports, 71, 149-155. JODAT, Y. A., KANG, M. G., KIAEE, K., KIM, G. J., MARTINEZ, A. F., ROSENKRANZ, A., BAE, H. &

SHIN, S. R. 2018. Human-derived organ-on-a-chip for personalized drug development. Current pharmaceutical design, 24, 5471-5486.

KIM, J. G., LEEM, Y.-E., KWON, I., KANG, J.-S., BAE, Y. M. & CHO, H. 2018. Estrogen modulates serotonin effects on vasoconstriction through Src inhibition. Experimental &

molecular medicine, 50, 1-9. LE, H. T., LIE, K. K., GIROUD-ARGOUD, J., RØNNESTAD, I. & SÆLE, Ø. 2019. Effects of

cholecystokinin (CCK) on gut motility in the stomachless fish ballan wrasse (Labrus bergylta). Frontiers in neuroscience, 13, 553.

LITOU, C., EFFINGER, A., KOSTEWICZ, E. S., BOX, K. J., FOTAKI, N. & DRESSMAN, J. B. 2019. Effects of medicines used to treat gastrointestinal diseases on the pharmacokinetics of coadministered drugs: a PEARRL Review. Journal of Pharmacy and Pharmacology,

71, 643-673. MAPARA, M., THOMAS, B. S. & BHAT, K. 2012. Rabbit as an animal model for experimental

research. Dental research journal, 9, 111. MITTAL, R., DEBS, L. H., PATEL, A. P., NGUYEN, D., PATEL, K., O'CONNOR, G., GRATI, M. H.,

MITTAL, J., YAN, D. & ESHRAGHI, A. A. 2017. Neurotransmitters: The critical modulators regulating gut–brain axis. Journal of cellular physiology, 232, 2359-2372.

OHNO, T., MOCHIKI, E. & KUWANO, H. 2010. The roles of motilin and ghrelin in gastrointestinal motility. International journal of peptides, 2010.

SENGUPTA, P. 2013. The laboratory rat: relating its age with human's. International journal of preventive medicine, 4, 624.

SZPIRER, C. 2010. Cancer research in rat models. Rat Genomics. Springer. THANGAM, E. B., JEMIMA, E. A., SINGH, H., BAIG, M. S., KHAN, M., MATHIAS, C. B., CHURCH,

M. K. & SALUJA, R. 2018. The role of histamine and histamine receptors in mast cell-mediated allergy and inflammation: the hunt for new therapeutic targets. Frontiers

in immunology, 9, 1873.

Page 176: MECHANISMS UNDERLYING THE CONTRACTILE RESPONSES OF

173

UESAKA, T., YOUNG, H. M., PACHNIS, V. & ENOMOTO, H. 2016. Development of the intrinsic and extrinsic innervation of the gut. Developmental biology, 417, 158-167.

WELCHKO, R. M., LÉVÊQUE, X. T. & DUNBAR, G. L. 2012. Genetic rat models of Parkinson's disease. Parkinson's Disease, 2012.

WERAWATGANON, D. 2014. Simple animal model of Helicobacter pylori infection. World Journal of Gastroenterology: WJG, 20, 6420.

ZIEGLER, A., GONZALEZ, L. & BLIKSLAGER, A. 2016. Large animal models: the key to translational discovery in digestive disease research. Cellular and molecular

gastroenterology and hepatology, 2, 716-724.