142
A Novel Mechanism for Nicotinic Acetylcholine Receptor Antagonist-Induced Cancer Cell Cytotoxicity By Mitch Clarke Master of Science Thesis University of Otago, Dunedin New Zealand 10 February 2016

A Novel Mechanism for Nicotinic Acetylcholine Receptor

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A Novel Mechanism for Nicotinic Acetylcholine Receptor

A Novel Mechanism for Nicotinic Acetylcholine Receptor

Antagonist-Induced Cancer Cell Cytotoxicity

By Mitch Clarke

Master of Science Thesis

University of Otago, Dunedin

New Zealand

10 February 2016

Page 2: A Novel Mechanism for Nicotinic Acetylcholine Receptor

ii

Abstract

Nicotinic acetylcholine receptors (nAChRs) are common ligand-activated neurotransmitter

receptors located throughout the body. The up-regulation of nAChRs subunits has been observed in

a number of cancer types; while current literature has identified various nAChR pathways that

contribute to the development of cancer. The present study identified a novel mechanism for the

observed cytotoxicity of the highly potent nAChR antagonist, pinnatoxin (PnTX), on human

epithelial carcinoma cell lines. Cell cycle and intracellular calcium studies suggest that PnTX exerts

its cytotoxic effects by inducing changes in the cell cycle via antagonism of the highly calcium

permeable α7-, and to a certain extent, the α4β2-nAChR receptors. This leads to an induction of cell

apoptosis via calcium-independent pathways. The use of nAChRs in combination with other known

cytotoxic agents, such as cyclosporin A, or apoptotic inducing chemotherapy agents, may provide a

sensitising tool that can reduce the dose required to elicit a cytotoxic response on developing

tumours.

Page 3: A Novel Mechanism for Nicotinic Acetylcholine Receptor

iii

Acknowledgements

I would first like to thank Dr Sarah Baird for her time, guidance and support over my

extended time in her lab. As my Masters and previously Honours supervisor, she has contributed a

great deal of knowledge and encouragement throughout my time in Dunedin. Her mentorship and

independent thinking have been invaluable while keeping me motivated throughout the many

repeated experiments. Her patience and willingness to let her students investigate any idea, no

matter how lateral, will continue to be her greatest strength as an academic mentor. It has been a

pleasure working alongside Sarah and I wish her all the best for the future.

I would like to thank the fellow students of the Baird Lab, Ben, Sean, Chloe and Irene, for

their contribution to this project. They have all provided a great deal of knowledge and willingness

to learn that have kept me motivated during my research. Their readiness to lend a hand or provide

helpful suggestions in times of experimental agony has made this experience a lot more enjoyable. I

can only hope I have contributed as much to their academic progress as they have to mine.

To the members of the various other labs within the department, in particular Floriane and

Sebastien of the Rosengren Lab, thank you for your time and effort. The materials, technical

knowledge and guidance they both provided were invaluable. Furthermore, their willingness to

dedicate time at the expense of their own research was truly humbling and a reflection of what

incredible individuals they are. To Shane of the Kerr Lab, thank you for your constant supply of

experimental materials, knowledge surrounding the concepts of this project and sense-of-humour in

times of insanity. I am grateful to you all.

Finally, to the University of Otago, and more importantly the Pharmacology and Toxicology

Department, I would like to express my gratitude for allowing me to use the facilities during my

time in Dunedin. In particular, I would like to thank all the staff and students of the Pharmacology

and Toxicology for being such genuine people, while never falling short of providing a laugh at the

most stressful of times.

Page 4: A Novel Mechanism for Nicotinic Acetylcholine Receptor

iv

Table of Contents

A Novel Mechanism for Nicotinic Acetylcholine Receptor Antagonist-Induced Cancer Cell

Cytotoxicity ........................................................................................................................................... i

Abstract ................................................................................................................................................ ii

Acknowledgements ............................................................................................................................. iii

Table of Contents ................................................................................................................................ iv

List of Tables .................................................................................................................................... viii

List of Figures ..................................................................................................................................... ix

List of Abbreviations .......................................................................................................................... xi

1 Introduction .................................................................................................................................. 1

1.1 Nicotinic Acetylcholine Receptors ........................................................................................ 1

1.1.1 Structure, Diversity and Localisation ............................................................................. 1

1.1.2 Neuronal nAChR Function ............................................................................................ 3

1.1.3 Non-Neuronal nAChR Function .................................................................................... 5

1.1.4 nAChRs in Disease ........................................................................................................ 7

1.2 nAChRs in Cancer Development .......................................................................................... 8

1.2.1 nAChR Expression in Cancer ........................................................................................ 9

1.2.2 nAChRs in Cancer Development and Proliferation ..................................................... 12

1.2.3 Role of nAChRs in Cancer Angiogenesis and Metastasis ........................................... 14

1.2.4 Anti-Apoptotic Effects of nAChRs in Cancer Survival ............................................... 16

1.2.5 nAChRs and Cancer Cell Death................................................................................... 18

1.3 nAChR-Modulators and nAChR-Based Therapies ............................................................. 19

Page 5: A Novel Mechanism for Nicotinic Acetylcholine Receptor

v

1.3.1 nAChR-Based Therapies for Common Diseases ......................................................... 20

1.3.2 nAChR-Modulators for Cancer Treatment .................................................................. 22

1.3.3 Marine Toxins .............................................................................................................. 25

1.4 Aims and Hypothesis........................................................................................................... 26

2 Methods ...................................................................................................................................... 29

2.1 Cell Culture ......................................................................................................................... 29

2.1.1 Materials....................................................................................................................... 29

2.1.2 Cell Lines and Cell Culture .......................................................................................... 29

2.1.3 MTT Assay .................................................................................................................. 30

2.1.4 Statistical Analysis ....................................................................................................... 31

2.2 Cell Viability Assay ............................................................................................................ 31

2.2.1 Materials....................................................................................................................... 31

2.2.2 Cell Viability Assay ..................................................................................................... 31

2.3 Western Blot ........................................................................................................................ 32

2.3.1 Materials....................................................................................................................... 32

2.3.2 Antibodies .................................................................................................................... 33

2.3.3 Protein Extraction ........................................................................................................ 33

2.3.4 Western Blotting .......................................................................................................... 34

2.4 Immunocytochemistry ......................................................................................................... 35

2.4.1 Materials....................................................................................................................... 35

2.4.2 Sample .......................................................................................................................... 35

2.4.3 Fluorescence Microscopy ............................................................................................ 35

Page 6: A Novel Mechanism for Nicotinic Acetylcholine Receptor

vi

2.5 Flow Cytometry ................................................................................................................... 36

2.5.1 Materials....................................................................................................................... 36

2.5.2 Cell Treatment and Staining ........................................................................................ 36

2.5.3 Fluorescence-Activated Cell Sorting (FACS).............................................................. 37

2.6 Intracellular Calcium ........................................................................................................... 38

2.6.1 Materials....................................................................................................................... 38

2.6.2 Fura-2 AM Fluorescence Measurement ....................................................................... 38

2.7 Cell Death Inhibitors ........................................................................................................... 39

2.7.1 Materials....................................................................................................................... 39

2.7.2 Cell Culture and Inhibitors ........................................................................................... 39

2.7.3 Isobologram ................................................................................................................. 39

2.8 Combination Therapy .......................................................................................................... 40

2.8.1 Materials....................................................................................................................... 40

2.8.2 Chemotherapy Combination Therapy .......................................................................... 40

3 Results ........................................................................................................................................ 41

3.1 Effects of nAChR Antagonists on Cell Viability ................................................................ 41

3.1.1 Cell Viability Assay ..................................................................................................... 41

3.1.2 Western Blot - Basal nAChR Levels ........................................................................... 47

3.1.3 Immunocytochemistry ................................................................................................. 48

3.1.4 Discussion .................................................................................................................... 52

3.2 Investigation of nAChR Antagonist Induced Cell Death .................................................... 57

3.2.1 Western Blot – α7 nAChR Levels following Treatment ............................................. 57

Page 7: A Novel Mechanism for Nicotinic Acetylcholine Receptor

vii

3.2.2 Flow Cytometry - Cell Cycle Analysis ........................................................................ 60

3.2.3 Flow Cytometry - TMRE ............................................................................................. 66

3.2.4 Flow Cytometry - Annexin V/PI .................................................................................. 68

3.2.5 Discussion .................................................................................................................... 75

3.3 Inhibition of nAChR Antagonist Induced Cell Death ......................................................... 78

3.3.1 Cell Death Inhibitors .................................................................................................... 79

3.3.2 Intracellular Calcium .................................................................................................... 83

3.3.3 Discussion .................................................................................................................... 87

3.4 Combination Treatment ....................................................................................................... 91

3.4.1 CsA + MECA ............................................................................................................... 91

3.4.2 Chemotherapy Agents + nAChR Antagonists ............................................................. 98

3.4.3 Discussion .................................................................................................................. 104

4 Discussion ................................................................................................................................ 107

4.1 Signalling through nAChRs is linked to Cancer Development ......................................... 107

4.1.1 nAChR Agonist Induced Cancer Progression ............................................................ 107

4.1.2 nAChR Antagonist Driven Cytotoxicity .................................................................... 107

4.2 The nAChR Antagonist PnTX Induces Cytotoxicity by Altering Intracellular Calcium

Levels ........................................................................................................................................... 108

4.3 Synergistic Cytotoxic Effects of Selective nAChR Antagonists with Common

Chemotherapeutic Agents ............................................................................................................ 109

4.4 Future Directions ............................................................................................................... 109

4.5 Final Conclusion................................................................................................................ 111

5 References ................................................................................................................................ 112

Page 8: A Novel Mechanism for Nicotinic Acetylcholine Receptor

viii

List of Tables

Table 1: Examples of Selective α7 nAChR-Modulators Investigated ............................................... 22

Table 2: Details of the Carcinoma Cell Lines .................................................................................... 30

Table 3: Cell Viability Assay Treatment Profile ............................................................................... 32

Table 4: Cell Death Inhibitor Treatment Profile ................................................................................ 39

Page 9: A Novel Mechanism for Nicotinic Acetylcholine Receptor

ix

List of Figures

Figure 1: Protein ribbon structure of a nicotinic acetylcholine. ........................................................... 2

Figure 2: The regulation of intracellular pathways .............................................................................. 9

Figure 3: The determination of MDA-MB-231, HT29 ...................................................................... 42

Figure 4: The evaluation of MDA-MB-231, HT29 ........................................................................... 46

Figure 5: Western blot analysis showing the basal level ................................................................... 48

Figure 6: Immunocytochemistry images of non-permeabilised ........................................................ 50

Figure 7: Immunocytochemistry images of non-permeabilised ........................................................ 51

Figure 8: Western blot analysis investigating the effects .................................................................. 59

Figure 9: Cell cycle analysis of antagonist treated MDA-MB-231 ................................................... 61

Figure 10: Cell cycle analysis of treated MDA-MB-231 ................................................................... 63

Figure 11: Cell cycle analysis of treated HT29 .................................................................................. 64

Figure 12: Cell cycle analysis of treated PC3 .................................................................................... 65

Figure 13: Evaluation of the mitochondrial transmembrane ............................................................. 67

Figure 14: Flow cytometric detection of apoptotic and necrotic cells ............................................... 70

Figure 15: Flow cytometric detection of apoptotic and necrotic cells. .............................................. 72

Figure 16: Flow cytometric detection of apoptotic and necrotic cells ............................................... 74

Figure 17: Examination of the effect of the cell death inhibitor Cyclosporin A ............................... 80

Figure 18: Examination of the effect of the cell death inhibitor ALLN ............................................ 81

Figure 19: Examination of the effect of the cell death inhibitor BAPTA .......................................... 82

Figure 20: Examination of the effect of the cell death inhibitor zVAD ............................................ 83

Figure 21: Ca2+ accumulation through Fura-2 AM detection ............................................................ 86

Figure 22: The cytotoxic effect of combined MECA and CsA treatment ......................................... 92

Figure 23: Isobologram example displaying the line of additivity .................................................... 94

Figure 24: Isobologram demonstrating the relationship between MECA ......................................... 95

Page 10: A Novel Mechanism for Nicotinic Acetylcholine Receptor

x

Figure 25: Flow cytometric detection of apoptotic and necrotic cells ............................................... 97

Figure 26: The cytotoxic effect of DOXO, DTX and ADI treatment ................................................ 99

Figure 27: The cytotoxic effect of DOXO, DTX and ADI treatment .............................................. 100

Figure 28: Determination of cell viability following treatment with DOXO .................................. 101

Figure 29: Evaluation of cell viability following treatment with DOXO ........................................ 103

Page 11: A Novel Mechanism for Nicotinic Acetylcholine Receptor

xi

List of Abbreviations

[Ca2+] Intracellular Calcium Concentration

5-HT Serotonin or 5-hydroxytryptamine

ACh Acetylcholine

AChE Acetylcholinesterase

ADI Adiphenine

AEBSF 4-(2-Aminoethyl)benzenesulfonyl fluoride hydrochloride

Akt Protein Kinase B

ALLN N-Acetyl-Leu-Leu-Nle-al

AM Acetoxymethyl Ester

ANOVA Analysis of Variance

API Activating Protein 1

APS Alkypyridinium Salt

APS Ammonium Persulfate

BAPTA 1,2-bis(o-Aminophenoxy)Ethane-N,N,N’,N’-Tetraacetic Acid

Bcl B-cell Lymphoma Protein

BSA Bovine Serum Albumin

CA1 Cornu Ammonis 1

Ca2+ Calcium Ion

CaCl2 Calcium Chloride

cAMP Cyclic Adenosine Monophosphate

CD4+ Cluster of Differentiation 4-Positive

CD8+ Cluster of Differentiation 8-Positive

cdc42 Cell Division Control Protein 42

CDDP Cisplatin

Page 12: A Novel Mechanism for Nicotinic Acetylcholine Receptor

xii

CDK4 Cyclin-Dependent Kinase 4

CDX Candoxin

ChAT Choline Acetyltransferase

CHT1 Choline Transporter 1

CNS Central Nervous System

CO2 Carbon Dioxide

COX Cyclooxygenase

CREB cAMP Response Element Binding Protein

CsA Cyclosporin A

DAPI 4,6-Diamidino-2-Phenylindole

DeN N-Nitrosodiethylamine or Diethylnitrosamine

DMEM Dulbecco’s Modified Eagle Medium

DMSO Dimethyl Sulfoxide

DNA Deoxyribonucleic Acid

DNAse Deoxyribonuclease

DOXO Doxorubicin

DTT Dithiothreitol

DTX Docetaxel

E2 Estradiol

ECL Electrochemiluminescence

EDTA Ethylenediaminetetraacetic Acid

EGCG (–)-Epigallocatechin-3-Gallate

EGF Epidermal Growth Factor

EGFR Epidermal Growth Factor Receptor

EMT Epithelial-Mesenchymal Transition

ER Estrogen Receptor

Page 13: A Novel Mechanism for Nicotinic Acetylcholine Receptor

xiii

ERK Extracellular Signal-Regulated Kinases

FACS Fluorescence-Activated Cell Sorting

FAK Focal Adhesion Kinase

FBS Foetal Bovine Serum

FGF Fibroblast Growth Factor

FITC Fluorescein Isothiocynate

GABA Gamma-Aminobutyric Acid

GYM Gymnodimine

H2O Water

HBSS Hanks Balanced Salt Solution

HEPES 4-(2-Hydroxyethyl)-1-Piperazineethanesulfonic Acid

HMEC-L Human Microvascular Endothelial Cells – Lung

IAP Inhibitor of Apoptosis Protein

IC50 Half Maximal Inhibitory Concentration

ICC Immunocytochemistry

IgG Immunoglobulin G

KCl Potassium Chloride

KH2PO4 Potassium Di-Hydrogen Orthophosphate

LD10 Lethal Dose to 10% of Individuals

LPS Lipopolysaccharide

mAChR Muscarinic Acetylcholine Receptor

MAPK Mitogen-Activated Protein Kinases

MCF Michigan Cancer Foundation

MECA Mecamylamine

MEK Mitogen-Activated Protein Kinase

MeSH 2-Mercaptoethanol

Page 14: A Novel Mechanism for Nicotinic Acetylcholine Receptor

xiv

MgSO4 Magnesium Sulphate

MLA Methyllycaconitine

MOPS 3-(N-Morpholino) Propanesulfonic Acid

mTOR Mammalian Target of Rapamycin

MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide

Na+ Sodium Ion

Na2HPO4 Di-Sodium Hydrogen Orthophosphate

nAChR Nicotinic Acetylcholine Receptor

NaCl Sodium Chloride

NAD Nicotinamide Adenine Diculeotide

NaHCO3 Sodium Bicarbonate

NDNI N-n-Decylnicotinium Iodide

NF-κB Nuclear Factor-Kappa B

NIC Nicotine

NMDA N-methyl-D-aspartate

NNK Nicotine-Derived Nitrosamine Ketone

NNN N-Nitrosonornicotine

NOD/SCID Non-Obese Diabetic/Severe Combined Immunodeficiency

NOS Nitric Oxide Synthase

NSCLC Non-Small Cell Lung Carcinoma

PBS Phosphate Buffered Saline

PEG-PLA Poly(Ethylene Glycol)-Poly(D,L-Lactide)

PI Propidium Iodide

PI3K Phosphatidylinositol 3-Kinase

PKC Protein Kinase C

PnTX Pinnatoxin

Page 15: A Novel Mechanism for Nicotinic Acetylcholine Receptor

xv

Raf Rapidly Accelerated Fibrosarcoma Protein

Ras Rat Sarcoma Protein

RNA Ribonucleic Acid

RPMI Roswell Park Memorial Institute

SCLC Small Cell Lung Carcinoma

SDS Sodium Dodecyl Sulfate

SEM Standard Error of the Mean

SLURP Secreted Ly-6/uPAR-Related Protein

SMAC Second Mitochondria-Derived Activator of Caspase

TEMED Tetramethylethylenediamine

TMRE Tetramethylrhodamine, Ethyl Ester, Perchlorate

TRAIL TNF-Related Apoptosis-Inducing Ligand

uPAR Urokinase Plasminogen Activator Receptor

VAChT Vesicular Acetylcholine Transporter

VEGF Vascular Endothelial Growth Factor

XIAP X-linked Inhibitor of Apoptosis Protein

zVAD-FMK N-Benzyloxycarbonyl-Val-Ala-Asp-Fluoromethyl Ketone

α-BTX α-Bungarotoxin

α-CbT α-Cobratoxin

Page 16: A Novel Mechanism for Nicotinic Acetylcholine Receptor

1

1 Introduction

1.1 Nicotinic Acetylcholine Receptors

Nicotinic acetylcholine receptors (nAChRs) are common ligand-activated neurotransmitter

receptors located throughout the body. nAChRs respond to the binding of the endogenous

neurotransmitter, acetylcholine (ACh), causing fast ionotropic cationic transmission through the cell

membrane (Albuquerque et al., 2009). The receptors are expressed on both neuronal and non-

neuronal cells, resulting in a diverse range of functions. Consequently, the receptors are linked to

many human disease states, making them one of the most intensely researched areas of molecular

biology and therapeutics.

1.1.1 Structure, Diversity and Localisation

The acetylcholine receptor family is comprised of two major sub-groups of receptors;

nAChRs and muscarinic acetylcholine receptors (mAChRs). Unlike mAChRs that relay

intracellular messages though G-protein-coupled transmission, nAChRs gate the flow of various

ions through the cell membrane to induce a cellular response (Albuquerque et al., 2009). The ability

of nAChRs to regulate ion transmission stems from their pentagonal structure of five subunits,

arranged around a central pore. Each subtype spans the length of the cell membrane, with both the

N- and C-terminal of the subtype protein located extracellularly. The nAChR is comprised of an

arrangement of different α, β, γ, δ and ε subtypes that dictate the flow of ions through the receptor

pore (Kalamida et al., 2007).

As with all members of the acetylcholine receptor family, receptor function is regulated by the

interaction between ACh and the ligand-binding site. Binding of ACh to the receptor occurs at the

interface between two subunits, with at least one subunit residing from the α-subunit family, on the

extracellular domain of the receptor, as represented in Figure 1. ACh binding causes a

conformational shift in the pentagonal structure of the receptor as a result of changes to the van der

Page 17: A Novel Mechanism for Nicotinic Acetylcholine Receptor

2

Waal interactions between the receptor subunits. Opening of the transmembrane pore allows the

movement of positively charged ions, including sodium and calcium (Ca2+) into the cell, and

potassium out of the cell, to elicit the cellular response (Miyazawa et al., 2003).

Figure 1: Protein ribbon structure of a nicotinic acetylcholine receptor, as viewed from the synaptic cleft (left) and

parallel with the cell membrane (right). The diagram displays the pentagonal structure of a common heteromeric

(α)2βγδ-nAChR, configured around a central pore with sections of each subunit located within the intracellular (I) and

extracellular (E) space. The αTrp149 ligand binding region of the α-subunit has been displayed in yellow. Images

adopted from (Unwin, 2005).

The localisation of the receptor also plays a role in the expression of specific nAChR

subunits. The sub-group of nAChRs is broadly split into two types, muscle-type and neuronal-type

nAChRs. Muscle-type nAChRs are located at the neuromuscular junction of muscle terminals,

where they play an important role in muscle contraction. Muscle-type nAChRs are comprised

strictly of a (α1)2β1εδ receptor configuration in the adult muscle (embryonic muscle contains the γ-

subunit in place of the ε-subunit, with the transition of γ- to ε-subunit expression occurring during

development) (Tzartos et al., 1998). Neuronal-type nAChRs demonstrate a higher degree of

diversity in terms of subunit expression. Receptor pentamers can be comprised of homomeric

Page 18: A Novel Mechanism for Nicotinic Acetylcholine Receptor

3

configurations of α-subunits (e.g. (α7)5), or heteromeric configurations consisting of both α- and β-

subunits (e.g. (α4)2(β2)3). As a result, the function, ionic permeability and responsiveness to various

exogenous compounds of the neuronal-type nAChRs can differ greatly between receptor

configurations (Sargent, 1993).

Traditionally, the expression of nAChRs consisting of homo- and heteromeric configurations

of α- and β-subunits was thought to be localised to within the nervous system. However, recent

research has identified the expression of non-neuronal nAChRs on various cells outside of the

nervous system. Various nicotinic α- and β-subunits are expressed on non-neuronal tissues,

including endothelial cells, epithelial cells, mesenchymal cells and a variety of immune cells

(reviewed in (Arias et al., 2009). The function of the different neuronal and non-neuronal nAChRs

is discussed in more detail in the following sections.

1.1.2 Neuronal nAChR Function

Neuronal nAChRs are widely spread throughout the nervous system, including certain areas

of the brain and within the peripheral ganglia. The nAChRs are comprised of combinations of nine

α (α2-α10) and three β (β2-β4) subunits, forming operational ion channels at the pre-terminal, pre-

synaptic and post-synaptic sites of neuronal cells (Dajas-Bailador et al., 2004). Pre-synaptically,

nAChRs influence neurotransmitter release through membrane depolarisation, or by altering

intracellular Ca2+ concentration due to the high permeability of some nAChR subunit configurations

to Ca2+. Post-synaptically, nAChRs contribute to the fast excitatory transmission of signals within

the central nervous system (CNS), enhancing post-synaptic excitation. However, the post-synaptic

effects of neuronal nAChRs are thought to be less relevant to their overall function in the CNS

(Kalamida et al., 2007).

Based on the expression of particular subunits on the pre-synaptic terminal, nAChRs in the

CNS can regulate the release of particular neurotransmitters. For example, dopamine release has

been found to be regulated by the expression of α4β2 and α6β2 subtypes in the mesolimbic pathway

of the CNS (Gotti et al., 2010); while, α7 subtype expression is linked to glutamatergic

Page 19: A Novel Mechanism for Nicotinic Acetylcholine Receptor

4

neurotransmission in various regions of the brain (Cui et al., 2013). The ability of nAChRs to

regulate neurotransmission is due to their ability to cause an increase in intracellular Ca2+ levels at

the pre-synaptic terminal. Non-α7 subtype receptor configurations, such as the α4β2, have relatively

low permeability to Ca2+ when activated. However, these receptors are functionally coupled to

voltage-gated calcium channels, resulting in an influx of Ca2+ into the pre-synaptic terminal

following ACh binding (Garduño et al., 2012). Alternatively, nAChRs, such as the homomeric α7

receptor, can induce glutamine release without the involvement of coupled ion channels, due to its

high permeability to Ca2+ through the central pore of the receptor once activated (Garduño et al.,

2012).

The ability of nAChR to regulate intracellular Ca2+ levels within the nervous system plays

another important role in the regulation of neuronal gene expression. The influx of Ca2+ into the cell

following nAChR activation of Ca2+ permeable subunits results in the downstream activation of

transcription factors (Nakayama et al., 2006). One example is the established role of nAChRs in

memory and plasticity events. The activation of nAChRs, especially those containing the highly

Ca2+ permeable α7 subunits, regulates the ERK/MAPK intracellular signalling cascade. The

prolonged activation of this signalling cascade results in the phosphorylation of various gene

transcription factors, such as the cAMP response element binding protein (CREB), Sp1 and c-myc.

The increase in gene transcription results in the expression of several proteins involved in memory

formation and long-term plasticity within the nervous system (Nishimoto et al., 2011). Furthermore,

the activation of nAChRs via exogenous agonists such as nicotine plays an important role in

addiction through the activation of similar intracellular signalling cascades (Changeux, 2010).

Finally, neuronal nAChR activation has been shown to be linked to neuroprotection within the

CNS, while contributing to protection against neuronal toxicity. nAChRs containing the α7 subtype

have been linked to the intracellular activation of the phosphatidylinositol 3-kinase (PI3K) cascade.

The neuronal activation of the PI3K pathway results in downstream phosphorylation of Akt, and

subsequently Bcl-2 and Bcl-xl proteins, a subset of anti-apoptotic proteins that can prevent cell

Page 20: A Novel Mechanism for Nicotinic Acetylcholine Receptor

5

death during neuronal toxicity (Kawamata et al., 2012). The interaction of neuronal nAChRs

interaction with N-methyl-D-aspartate (NMDA) glutamate receptors has also been shown to protect

against Ca2+ induced neurotoxicity. NMDA receptor stimulation induces an influx in Ca2+ during

ischaemic brain injury, resulting in the over-activation of intracellular nitric oxide synthase and

subsequent glutamate-related cell death (Akaike et al., 1994). However, activation of nAChRs

during glutamate-induced neurotoxicity has demonstrated potential neuroprotective effects of the

nAChRs. Pre-treatment with nicotine, a potent nAChR agonist, prior to glutamate exposure

significantly increased cell viability in cultures of rat cortical neurons, compared to glutamate

exposure alone (Kaneko et al., 1997). The neuroprotective effects were shown to be related to the

inhibition of NOS through the activation of α4β2 and α7 nAChRs (Takada et al., 2003).

1.1.3 Non-Neuronal nAChR Function

ACh is synthesised by a large range of non-neuronal cells, with functions that differ greatly

from the neuronal neurotransmitter role of the cholinergic system. The role of ACh in non-neuronal

cells plays an intermediate role between the cells and the surrounding environment, facilitating cell

proliferation, differentiation and migration, amongst other physiological processes. Blockage of

these receptors leads to cellular dysfunction and death, highlighting their importance in normal

cellular processes of non-neuronal cells (Wessler et al., 2008). The expression of specific nAChR

subtypes on non-neuronal cells varies according to the cell phenotype and the intra- and

intercellular environment. Analogous to the presence of nAChRs in the nervous system, both

homomeric and heteromeric combinations of α- and β-nAChR subunits are expressed amongst non-

neuronal cells (Arias et al., 2009).

The major function of nAChRs in non-neuronal cells is the regulation of intercellular signal

transduction through auto- and paracrine secretions of ACh (Chen et al., 2002). One example of

non-neuronal nAChR signal transduction is the role of these receptors in epidermal cells, where the

concentration of ACh is found at levels significantly higher than in other non-neuronal regions of

the body (Grando et al., 2006). Real-time polymerase chain reaction and immunostaining analysis

Page 21: A Novel Mechanism for Nicotinic Acetylcholine Receptor

6

of keratinocytes within the epidermis has eluded to the presence of α3, α5, α7, α9, α10, β2 and β4

subunits; in addition to the expression of the functional enzymes required for the synthesis and

breakdown of ACh (Kurzen et al., 2004; Nguyen et al., 2000). Autocrine and paracrine secretions

of ACh activate keratinocyte α7-nAChRs, utilising the influx of Ca2+ into the cell as a second

messenger. The downstream effect of the intracellular influx of Ca2+ results in the regulation of

cellular differentiation, such as cytoskeleton reorganisation and expression of migratory integrins.

Interestingly, the removal of the α7 signalling pathway in keratinocytes not only prevented

differentiation of these cells, but also decreased the expression of pro-apoptotic signalling

molecules, suggesting a secondary role of nAChR activation in keratinocyte apoptosis (Arredondo

et al., 2002).

Much like the role of nAChRs in epidermal signalling transduction, non-neuronal nAChRs

are critical in the formulation of new blood vessels. Non-neuronal nAChRs have been shown to

contribute to angiogenesis through the expression of homomeric α7 nAChRs and several

heteromeric nAChRs, including α3, α5, β2 and β4 subunits, on endothelial cells (Arias et al., 2009).

Stimulation of nAChRs of endothelial cells with the non-selective nAChR agonist, nicotine, induces

endothelial tube formation in vitro (Heeschen et al., 2002). Investigation into the activation of

endothelial cell nAChRs established that α7 homomeric receptors were the primary receptor

involved following activation with a α7-selective agonist, dimethoxybenzylidene (Li et al., 2006);

while tube formation was significantly reduced in the presence of the α7-selective antagonist,

methyllycaconitine (Beckel et al., 2006). Furthermore, the non-selective blockade of nAChRs with

α-bungarotoxin (α-BTX) reduces endothelial cell migration by significantly inhibiting fibroblast

growth factor (FGF) and vascular endothelial growth factor (VEGF), important factors for

regulating the adhesion between endothelial cells (Ng et al., 2007).

Research has also shown the ability of ACh to modify the immune response through the

expression of non-neuronal nAChRs on various immune cells via similar auto- and paracrine

functions. The presence of ACh during in vitro immune models has demonstrated the role of ACh

Page 22: A Novel Mechanism for Nicotinic Acetylcholine Receptor

7

in the induction of CD4+ T-cell maturation and in the differentiation of CD8+ T-cells into cytolytic

T-cells (Basso et al., 2008; Zimring et al., 2005). Similarly, several nAChR subtypes have been

identified on B lymphocytes, with both α4α5β2 and α7α5β4 receptor configurations identified.

Murine knockouts of the α4, α7 or β2 subtypes showed that the nAChR subtypes are critically

involved in supporting B lymphocyte survival and cellular growth (Skok et al., 2007). Conversely,

knockdown of the α7 subunit in a murine model has been shown to increase the expression of IgG1

antibodies in response to an antigen, compared to wild-type mice, suggesting a possible immune

suppression role of nAChRs (Kawashima et al., 2007).

1.1.4 nAChRs in Disease

Due to the vast array of neuronal and non-neuronal processes that are governed by nAChRs, a

number of pathological conditions can arise when substantial alterations of the neuronal and non-

neuronal cholinergic system occur. Neuronal nAChR’s are involved in the fine control of complex

behaviours within the CNS, consequently, alterations to their function can contribute to the

development of several behavioural diseases (Gotti et al., 2006). In contrast, the role of the

cholinergic system of non-neuronal cells in disease is not well known.

Alzheimer’s disease, the most common cause of dementia, is characterised by a distinct loss

of cholinergic function within parts of the CNS. Recent research has alluded to the reduced numbers

of nAChRs in Alzheimer’s patients, with the α7 subunit being of greatest importance. Treatment

with nAChR agonists was shown to protect and delay the loss of neurons (Parri et al., 2011).

Similarly, changes in neuronal nAChR function have been associated with schizophrenia disease.

Studies investigating the post-mortem brains of schizophrenia patients have observed a decrease in

cholinergic receptor expression involving the α7 and α4β2 subunits in the hippocampal and striatal

regions of the brain (Durany et al., 2000; Freedman et al., 2000). Changes in α7 and α4β2 subunit

expression within the CNS have also been associated with Parkinson’s disease (Meyer et al., 2009),

autism (Martin-Ruiz et al., 2004) and attention-deficit hyperactivity disorder (Wilens et al., 2007),

with potential selective agonists offering potential treatment benefits in these disease states.

Page 23: A Novel Mechanism for Nicotinic Acetylcholine Receptor

8

Alterations in the non-neuronal cholinergic system, such as small changes in the expression

pattern of selected subtypes, result in similar cellular stress outside of the nervous system (Wessler

et al., 2008). Studies have indicated a possible role of nAChRs in dermatitis, with an increase in

acetylcholine of 14-fold in superficial and 3-fold in the underlying portion of patients with atopic

dermatitis. In the same patients, a significant increase in choline-acetyltransferase, the enzyme

responsible for the production of ACh, was seen in the various skin and immune cells surrounding

the affected site (Wessler et al., 2003). In contrast, ACh content is substantially reduced in the

blood cells and bronchi of cystic fibrosis patients (Wessler et al., 2007). The cholinergic

dysfunction is thought to contribute to the characteristic alteration in water and ion balance during

cystic fibrosis; while agonists of the α7-nAChR have been shown to produce anti-inflammatory

effects in the epithelium of patients with cystic fibrosis (Greene et al., 2010). Finally, the

overexpression of nAChR on cancer cells stimulates tumour growth when activated in various

cancer types, with the role of nAChR in cancer development discussed in more detail in the

following section (Paleari et al., 2008).

1.2 nAChRs in Cancer Development

Cancer is defined as the uncontrolled growth of abnormal cells within the body. The process

by which healthy cells develop into malignant cancerous cells is complex, with a number of

endogenous and exogenous factors contributing to cancer development. However, recent research

has identified a range of common hallmarks acquired by cells during carcinogenesis, which allow

the abnormal cells to proliferate and spread throughout the body. In the most simplistic form, these

hallmarks include the ability of the cancer to sustain proliferation signalling, evade growth

suppressors, resist cell death, enable replicative immortality, induce angiogenesis, and activate

invasion and metastasis (Hanahan et al., 2011). The following section details the role of neuronal

and non-neuronal nAChRs in cancer development through facilitating some of these key functional

changes, with the key intracellular pathways summarised in Figure 2.

Page 24: A Novel Mechanism for Nicotinic Acetylcholine Receptor

9

Figure 2: The regulation of intracellular pathways and pro-carcinogenic effects following nAChR activation.

Endogenous and exogenous ligands bind to the specific nAChRs, causing an influx of Ca2+and other cations through

the nAChRs and voltage-gated Ca2+channels. The large Ca2+influx triggers the activation of various second

messengers, resulting in angiogenesis, cell proliferation and the inhibition of apoptosis. Figure adopted from (Schuller,

2009).

1.2.1 nAChR Expression in Cancer

Similarly to the expression of nAChRs on normal healthy cells, multiple nAChR subtypes are

expressed on both neuronal and non-neuronal cancer cells. The expression pattern of these subtypes

may vary greatly between normal and cancerous cells, where variations in nAChR expression levels

facilitate cancer development and proliferation. Extensive research has been conducted on the

nAChR patterns of various lung cancer types, due to the link of nicotine exposure and lung cancer

development of smokers (Egleton et al., 2008). Comparison of the expression patterns of nAChR

Page 25: A Novel Mechanism for Nicotinic Acetylcholine Receptor

10

subtypes between normal and cancerous lung cell types have revealed a significant upregulation of

the β4 subunit in non-small cell lung carcinoma (NSCLC) cells. Conversely, a decrease in α4 levels

was shown across the same comparison (Lam et al., 2007). Interestingly, an increase in the

expression of nAChRs has also been observed in lung cancer cells of non-smokers. A comparison

between NSCLCs in smokers and non-smokers showed variations in the expression patterns of

some nAChR subunits, with a higher expression of α6β3 receptors on NSCLCs of non-smokers,

compared to those from smokers (Lam et al., 2007). One hypothesis is that other exogenous

compounds act on nAChRs in a similar fashion to nicotine. N-nitrosodiethylamine (DeN), a

compound commonly found in food, contains a similar structure to ACh, with a high affinity for

both homo- and heteromeric nAChRs (Schuller, 2007). Compounds such as DeN may lead to the

non-tobacco-regulated modulations of nAChRs and development of lung cancer in non-smokers

(Niu et al., 2014).

The up-regulation of nAChRs subunits has also been observed in a number of cancer types,

with the majority of research focused on the α7 and α4β2 subtypes due to their abundance across all

cell types (Le Novere et al., 1995). Significant levels of either receptor subtype have been identified

on human mesothelioma (Trombino et al., 2004), colon (Wong et al., 2007), lung (Lam et al.,

2007), oesophageal (Arredondo et al., 2007), breast and pancreatic cells (Dasgupta et al., 2009).

Nicotine affinity studies have shown the ability of nicotine to bind with a higher affinity to α4β2

receptors, than to α7 nAChRs. The higher affinity of nicotine to α4β2 receptors causes a long-term

desensitisation of the receptor following chronic exposure, while having no effect on the sensitivity

of the α7 subtype (Kawai et al., 2001). Consequently, the biological function of the α7 nAChRs are

increased following nicotine exposure, resulting in an increase in cancer cell proliferation due to the

pro-carcinogenic effects of α7 nAChRs. Alternatively, the α4β2 receptor is mainly involved in

inhibitory cellular functions, with desensitisation of the receptor contributing to an environment

ideal for cancer development and proliferation (Schuller, 2009).

Page 26: A Novel Mechanism for Nicotinic Acetylcholine Receptor

11

Interest surrounding the overexpression of another nAChR subtype, the α9-subunit, has grown

recently with the subunit being linked to the development of breast cancer. Microdissected breast

tumours of 50 human breast cancer samples have shown an average 7.84-fold increase in α9-

nAChR mRNA expression in 67.3% of the samples tested. In vivo studies inducing overexpression

of the α9-nAChR of MCF-10A cell xenografts in nude mice have revealed a substantial increase in

tumour growth following treatment with nicotine (mean 84% increase in tumour size) (Lee et al.,

2010). The up-regulation of the α9-nAChR expression in breast cancer has been linked to the

activation of the estrogen receptor (ER). Prolonged exposure to estrogen and nicotine are known

risk factors for the formation of breast cancer (Daniell, 1980). Recent studies have shown the ability

of nicotine to act in a similar fashion to estrogen, binding to the ER and inducing gene transcription.

The activated ER, following nicotine or estrogen exposure, can specifically bind the activating

protein 1 (API)-binding site of the α9-nAChR gene, inducing transcription and overexpression of

the α9-nAChR. The results indicate that the ER-induced α9-nAChR overexpression may play a

central role in the association between nicotine or hormonal exposure and human breast cancer

formation (Lee et al., 2011).

The expression of nAChRs in cancers of the CNS is less characterised than the expression

on non-neuronal cancer types. The expression of nAChRs on normal healthy neuronal cells is well

known, with multiple subtypes expressed, playing an important part in many physiological

processes (see Neuronal nAChR Function) (Kalamida et al., 2007). One common form of CNS

cancer is glioblastoma multiforme, an aggressive type of malignant glial cancer originating from

healthy glial cells that comprise the supportive tissue within the brain (Parsons et al., 2008).

Isolation of various glioblastoma cell types, including U87, LN18 and U373 cells, have allowed for

effective in vitro studies into the characteristics of this aggressive tumour. However, due to the

difficulty of delivering anti-cancer drugs through the blood brain barrier and the widespread role of

nAChRs in many other normal CNS functions and pathological diseases, only limited investigations

into the role of nAChRs in the development of glioblastoma have been conducted. A study

Page 27: A Novel Mechanism for Nicotinic Acetylcholine Receptor

12

undertaken by (Zhan et al., 2011), has exploited the expression of nAChRs on U87 cells to facilitate

the delivery of the chemotherapy agent paclitaxel to glioblastoma cells. Poly(ethylene glycol)-

poly(D,L-lactide) (PEG-PLA) micelles encapsulating paclitaxel were coupled with candoxin

(CDX), a selective α7-nAChR antagonist, and injected intravenously into nude mice bearing

intracranial U87 glioblastoma. The paclitaxel-encapsulated PEG-PLA micelles significantly

prolonged average survival time by 7 days when coupled with CDX, compared to PEG-PLA-

Paclitaxel treatment alone. The study demonstrated the possible mechanism for exploiting

differential expression of nAChRs on the surface of glioblastoma cancer cells (Zhan et al., 2011).

1.2.2 nAChRs in Cancer Development and Proliferation

One of the most fundamental characteristics of cancer cells to develop and grow is their

ability to sustain rapid proliferation. Normal tissue cells regulate the production and release of

various growth factors that ensure controlled progression through the cell cycle, limiting tissue

growth. However, cancer cells lose the ability to regulate these growth factors, resulting in

sustained and uncontrolled proliferative signalling (Hanahan et al., 2011). nAChRs have been

shown to aid in the proliferation of cancer cells through changes in receptor expression and

downstream signalling pathways of various receptor subtypes that contribute to the loss of

homeostatic regulation of cell growth.

Investigation into the proliferative signalling of nAChRs in cancer cells using potent nAChR

agonists, such as nicotine-derived nitrosamines, has revealed several pathways important for tumour

growth. Selective binding of nitrosamines to α7-nAChRs of pulmonary neuroendocrine cells and

small cell lung carcinoma (SCLC) cells activates multiple intracellular signalling messengers,

including protein kinase C (PKC), Raf1, the mitogen-activated kinases ERK1 and ERK2, and

several transcription factors (FOS, JUN and MYC) (Jull et al., 2001). The transcription factors

FOS, JUN and MYC induce transcription of specific promoter elements located on proliferative

genes, such as c-erbB-1 (Volm et al., 1998). The proliferative effects following nAChR activation

were significantly reduced through the use of a selective α7-nAChR inhibitor, Ca2+ channel

Page 28: A Novel Mechanism for Nicotinic Acetylcholine Receptor

13

blockers or pharmacological inhibition of the ERK1 and ERK2 kinases, further validating the

central role of nAChRs in cancer cell proliferation (Sheppard et al., 2000; Xu et al., 2004).

Similar proliferative pathways related to nAChR-activation have been identified in other

cancer models. Studies using NSCLC and other lung epithelial carcinomas have shown the

nitrosamine-induced activation of the PI3K-Akt pathway, resulting in the phosphorylation of the

proliferation specific p70S6-kinase messenger. The effects on cancer cell proliferation were

comparable between selective agonists of nAChRs containing α3, α4 or α7 subunits (West et al.,

2003). Furthermore, the activation of nAChRs in breast cancer cell lines, MCF10A and MCF7 cells,

enhances the activity of PKC-α and cell division control protein 42 (cdc42) involved in cellular

proliferation and migration (Guo et al., 2008). While studies have suggested that the nAChR-

proliferative effects on breast cancer cells are dependent on the expression of the α9 subunit (Chen

et al., 2011). Interestingly, the desensitisation of α4β2 receptors during prolonged exposure to

nicotine or estrogen, as previously discussed, contributes to the proliferative state of a cancer cell.

Studies have shown the ability of the α4β2-nAChR to regulate the release of gamma-aminobutyric

acid (GABA), an important neurotransmitter with tumour suppressing functions. A reduction in

GABA release has been demonstrated in nitrosamine-stimulated pulmonary adenocarcinoma cells,

suggesting a link between α4β2 desensitising and reduced GABA expression (Schuller et al., 2008).

Furthermore, reduced GABA levels have been observed in the brains of smokers (Epperson et al.,

2005); while a reduction in the RNA levels of the α4 subunit gene have been shown in pulmonary

adenocarcinoma cells (Lam et al., 2007).

nAChR signalling can influence cancer cell proliferation through synergistic functions with

various growth factors. Stimulation of nAChRs of bovine adrenal pheochromatocytes induces the

production and release of FGF-2, a mitogenic neurotrophic protein that controls the development

and plasticity of various neuronal cell types. Induction of the FGF-2 growth hormone is controlled

through the cAMP/PKC signalling pathway, a common cascade linked to nAChRs and cancer cell

growth; while selective inhibition of the α7-nAChRs significantly reduces FGF-2 expression

Page 29: A Novel Mechanism for Nicotinic Acetylcholine Receptor

14

(Brown et al., 2012; Moffett et al., 1998). The activation of nAChRs by nicotine has also been

shown to increase the expression of epidermal growth factor receptor (EGFR), which when

activated by extracellular growth factors, signals cancer cell proliferation. Nicotine treatment of

MDA-MB-231 breast cancer cells was shown to stimulate EGFR expression, via the ERK1/2

signalling pathway, and promote breast cancer growth (Nishioka et al., 2011). Likewise, nicotine

induced glioma cell growth and colony formation in U87 and GBM12 cell lines through the

increased phosphorylation of EGFR, AKT and ERK. The proliferative effects were reduced

following the inhibition of the EGFR, PI3K and MEK pathways, suggesting the importance of

nAChRs in cancer cell sensitisation to extracellular growth signals (Khalil et al., 2013).

1.2.3 Role of nAChRs in Cancer Angiogenesis and Metastasis

Analogous to normal tissues, tumours require constant access to nutrients and oxygen in order

to proliferate. The physiological process of angiogenesis in which new vasculature is formed

provides the required circulation for a tumour to grow. During controlled processes, such as wound

healing, the angiogenic response is transiently activated; however, during tumour progression, the

process of angiogenesis is poorly regulated. A phenomenon called the ‘angiogenic switch’ occurs

where prolonged and sustained signalling for the development of new vasculature within the tumour

microenvironment results in continuous vessel formation (Hanahan et al., 1996; Hanahan et al.,

2011). Similar to the regulation of apoptosis, angiogenesis is thought to be regulated by the

counterbalance of pro- and anti-angiogenic factors. Several angiogenic inducers have been

identified in the process of both healthy and tumour-associated angiogenesis. VEGF, a key player in

the initiation of new blood vessel formation, is up-regulated in response to hypoxia and oncogene

signalling. In addition, the pro-angiogenic factor, FGF, has been linked to sustaining tumour

angiogenesis during prolonged expression (Baeriswyl et al., 2009; Ferrara, 2009; Hanahan et al.,

2011).

The role of nAChRs in both normal physiological angiogenesis and tumour-associated

angiogenesis is well established. Studies inducing nAChR activation through nicotine

Page 30: A Novel Mechanism for Nicotinic Acetylcholine Receptor

15

administration in both the Lewis lung cancer model and colon cancer xenografts in mice have

demonstrated an increase in tumour vascularity, resulting in accelerated tumour growth (Heeschen

et al., 2001; Natori et al., 2003). Mecamylamine has been shown to partially inhibit the nicotine

induced angiogenic response and return capillary density to control levels, suggesting the

importance of the α7-subunit (Zhu et al., 2003). The pro-angiogenic effects mediated by the α7-

nAChR are linked to the induction of VEGF, involving various nAChR-linked signalling pathways.

nAChR activation significantly increases VEGF expression by >2-fold in NSCLC cells (A549 and

H157) in the presence of nicotine. The pharmacological blocking of various nAChR-mediated

signalling pathways, including Ca2+/calmodulin, c-Src, PKC, PI3K/Akt, MAPK/ERK and mTOR,

significantly reduced the nicotine induced up-regulation of the pro-angiogenic factor (Zhang et al.,

2007). Comparable effects have also been observed in gastric cancers by up-regulating both COX2

and VEGF-receptors in response to nAChR stimulation (Shin et al., 2005).

Once established, malignant tumour cells begin a process of local invasion and formation of

distant colonies, termed metastasis. Cancer cells of the primary tumour follow a sequence of key

events that characterise the process of metastasis. First, the cells invade the surrounding tissue, and

enter the microvasculature of the lymph and blood systems (intravasation). The cancer cells then

migrate from the bloodstream into distant tissues (extravasation), where the cells must adapt to the

new environment in order to form new colonies and proliferate (Chaffer et al., 2011). An integral

process in tumour metastasis is the ability of cancer cells to undergo the epithelial-mesenchymal

transition (EMT). EMT is a mechanism by which cancer cells lose their endothelial characteristics

and develop a more migratory mesenchymal phenotype, stimulating invasion and migration. The

initiation of EMT involves the action of several transcription factors that regulate genes involved in

the migratory process, allowing cancer cells to down-regulate adhesive junctions with neighbouring

cells and intravate the local vasculature (Grando, 2014).

Signalling through α9-containing nAChRs is critical in the migration of several cancer cell

types. The expression of the α9 subunit in breast cancer cell lines, MCF-7 and MDA-MB-231,

Page 31: A Novel Mechanism for Nicotinic Acetylcholine Receptor

16

increases cell migration in response to nicotine; while the effects are blocked through the selective

inhibition of the α9 subunit both in vitro and in vivo (Hung et al., 2011; Lee et al., 2010). The

stimulation of α9-containing nAChRs results in the phosphorylation of several adhesion and

intercellular junction proteins, such as FAK, paxillin and β-catenin, key steps in the EMT

(Chernyavsky et al., 2007). Furthermore, the proinvasive effects of nAChRs have been linked to

α7-subunit expression on NSCLCs, breast cancer and pancreatic cancer cell lines. Nicotine

stimulation of the receptor facilitated cellular invasion across all cell lines, and most importantly

induced changes in gene expression consistent with the EMT. The reduction in epithelial markers,

such as E-cadherin, and the increased expression of mesenchymal proteins, such as vimentin and

fibronectin, in response to nAChR stimulation, further validate the role of nAChRs in the EMT and

tumour metastasis (Dasgupta et al., 2009).

1.2.4 Anti-Apoptotic Effects of nAChRs in Cancer Survival

Apoptosis serves as a natural barrier against the development of cancerous cells. Some of the

notable signals that induce apoptosis include uncontrolled cellular signalling and DNA damage,

both of which are essential precursors to the development of cancer. The intracellular activation of

apoptosis is controlled by the balance of pro- and anti-apoptotic regulator messengers, that when

altered towards a pro-apoptotic state, induce a cascade of downstream effectors that result in

programmed cell death. One family of anti-apoptotic molecules, the Bcl-2 family, are of particular

relevance to cancer. Bcl-2 acts as an inhibitor of apoptosis, by binding to and suppressing the

effects of the pro-apoptotic proteins Bax and Bak. When activated, the Bax and Bak proteins disrupt

the outer mitochondrial membrane, causing the release of pro-apoptotic factors, such as cytochrome

c, that induce the cascade of proteolytic caspases, resulting in apoptosis. Overexpression of Bcl-2 in

cancer cells inhibits the downstream effector molecules of apoptosis, thus allowing the cancer cells

to continue to proliferate (Adams et al., 2007; Hanahan et al., 2011).

Recent studies into the effects of nicotine on human MCF10A and MDA-MB-231 breast

cancer cells have shown an association between nAChR activation and Bcl-2 expression. Treatment

Page 32: A Novel Mechanism for Nicotinic Acetylcholine Receptor

17

of breast cancer cells with 0.5 μM nicotine for 48 hours produced a significant 2-fold increase in

Bcl-2 expression, coinciding with a 2-fold increase in cell colony formation. Interestingly, the

increase in expression of Bcl-2 was inhibited by the co-administration of KP372-1, a selective Akt

inhibitor. The inhibition of Bcl-2 up-regulation in the presence of nicotine was again reflected in the

number of cancer cell colonies, with the number of colonies returning to baseline in the presence of

KP372-1. The results demonstrated that persistent activation of nAChRs induces an up-regulation in

Bcl-2, enhancing cancer cell survival through the induction of anti-apoptotic pathways, with the

effects regulated by the intracellular messaging of the Akt pathway (Nishioka et al., 2011).

Additional studies have further added to the role of nAChR in inducing Bcl-2 expression, with

nicotine treatment significantly prolonging cell survival and protecting breast cancer cells against

doxorubicin induced cell death through mechanisms involving Bcl-2 phosphorylation (Mai et al.,

2003).

Similar increases in Bcl-2 expression of various other cancer cell types have been observed

following nAChR activation. Human colorectal cancer cell lines treated with 1 μM nicotine induced

a significant increase in phosphorylated Akt and Bcl-2. Further investigation into the specific

nAChR subtypes involved in this process found that co-treatment with α-Bungarotoxin, a selective

α7-nAChR antagonist, inhibited both cell growth and the anti-apoptotic pathways (Cucina et al.,

2012). Similarly, the anti-apoptotic activity of human bronchial epithelial cells is thought to be

mediated via the expression of α3-, α4- and α7-containing nAChRs, resulting in the downstream

activation of the Akt pathway (West et al., 2003). Exposure of lung epithelial and cancer cells to

nicotine and nicotine-derived nitrosamines promotes cell survival through the increased expression

of Bcl-2, complemented by an increased resistance to cisplatin-induced apoptosis. Comparable

effects were observed between the healthy lung epithelial cells and the cancer cells, further

consolidating the idea that nAChRs play a key role in the development of cancer through the

induced expression of anti-apoptotic factors (Nishioka et al., 2010).

Page 33: A Novel Mechanism for Nicotinic Acetylcholine Receptor

18

1.2.5 nAChRs and Cancer Cell Death

The anti-apoptotic effects of nAChRs in cancer survival are well defined in current literature.

nAChR stimulation during cancer development induces the Akt signalling pathway, increasing the

phosphorylation of the known anti-apoptotic messenger, Bcl-2. These anti-apoptotic effects of

nAChRs expressed on various cancer cell types have been discussed. However, the relationship

between nAChR blockade via antagonists and the apoptotic activity of cancer cells is less well

defined by the current literature.

Recent studies have demonstrated that various lung cancer cell lines express all the functional

components of the ACh autocrine pathway, including acetylcholinesterase (AChE), choline

acetyltransferase (ChAT), vesicular acetylcholine transporter (VAChT) and choline transporter 1

(CHT1). The complete functional loop of ACh signalling allows cancer cells to promote

proliferation through the autocrine activation of the nAChRs (Lau et al., 2013; Song et al., 2008).

Lau and colleagues investigated the possible pro-apoptotic effects of inhibiting the ACh autocrine

pathway in bronchioalveolar carcinoma cells. Initially, A549 and H358 cells treated with 100 nM

nicotine demonstrated a significant increase in cancer cell ACh secretion, through the up-regulation

of VAChT and ChAT, acting specifically through the α7-, α3β2 and β3-containing nAChR

subunits. The increase in AChR-related enzymatic activity and cell signalling coincided with an

increase in bronchioalveolar carcinoma cell proliferation. The carcinoma cells were then co-treated

with 50 μM vesamicol, a VAChT antagonist, to investigate whether the nicotine-ACh induced

growth of human bronchioalveolar carcinoma cells could be alleviated. Vesamicol caused

significant apoptosis in A549 and H358 cells, with a 2.5 to 3-fold increase in apoptosis relative to

nicotine-only treated carcinoma cells. The pro-apoptotic effects of vesamicol were linked to a

significant decrease in phosphorylated Akt. The findings can be explained by decreasing nAChR

activation swaying the balance of apoptotic mediators towards a pro-apoptotic state, thus inducing

cancer cell death (Lau et al., 2013).

Page 34: A Novel Mechanism for Nicotinic Acetylcholine Receptor

19

The homomeric α7 nAChR is of particular interest given its abundant expression and well-

established role in the activation of signalling transduction pathways with anti-apoptotic and

proliferative effects. APS8, a polymeric alkypyridinium salt, was investigated by (Zovko et al.,

2013) for its ability to induce NSCLC cell apoptosis through selective antagonistic binding to α7

nAChRs. APS8 showed a concentration dependent decrease in lung cancer cell viability (IC50 =

~370 nM), while having little effect on normal fibroblast cells. The decrease in cancer cell viability

was correlated with a prominent induction of apoptotic morphology, with ~40% of cells found to be

apoptotic following exposure to 500 nM of APS8 for 48 hours. Radioligand displacement studies

demonstrated that the pro-apoptotic effects were mediated via antagonising the α7 nAChRs through

displacing the specific binding of 125I-α-BTX to the receptors. The effects of APS8 on a number of

apoptotic proteins was also investigated using a Human Apoptosis Antibody Array. APS8 induced

the up-regulation of several pro-apoptotic proteins, including bad, bax, cytochrome C, SMAC,

TRAIL-R1, Fas, and various caspases, while down-regulating anti-apoptotic proteins, such as Bcl-2

and Bcl-W. The ratio between the levels of bax and Bcl-2 proteins determines whether apoptosis

will be induced, with an increase in the bax/Bcl-2 ratio causing a switch towards mitochondrial

membrane permeabilisation and activation of apoptosis. Therefore, the change in bax and Bcl-2

expression following α7 nAChR antagonism may facilitate the observed APS8 induced cancer cell

death (Zovko et al., 2013). Given the effects of agonists and antagonists of nAChR in controlling

cellular growth, they are being explored as potential therapeutics for a number of diseases.

1.3 nAChR-Modulators and nAChR-Based Therapies

An improved understanding of various neurological and inflammatory diseases has implicated

the potential role of nAChRs in these conditions. Furthermore, the differences in the expression

pattern of the various nAChR subtypes within a disease state allows for selective pharmacological

targeting of these receptors. The following section explores some of the nAChR-modulators being

developed to target a number of nAChR-associated diseases.

Page 35: A Novel Mechanism for Nicotinic Acetylcholine Receptor

20

1.3.1 nAChR-Based Therapies for Common Diseases

Agonists of α7-nAChRs have been identified as pharmacologically relevant compounds for

the treatment of several neuronal or inflammatory associated diseases. Examples of nAChR-

modulators currently under investigation for use in various disease states, as reviewed by (Pohanka,

2012), are presented in Table 1. The association between α7 nAChRs and the accumulation of

Amyloid β peptide within the CNS of Alzheimer’s patients has been well characterised. The

development of agonists to target the nAChR subtype has shown promise in the treatment of

Alzheimer’s disease by inducing neuroprotection and slowing disease progression (Lombardo et al.,

2014). A potent α7 nAChR agonist, SEN1233, is of current interest due to its ability to cross the

blood brain barrier when administered orally, thus alleviating some of the common problems

associated with current CNS-associated treatments. ABT-107, another agonist of α7 nAChR has

shown promise in the treatment of Alzheimer’s disease through protecting rat cortical cultures from

glutamate induced toxicity (Pohanka, 2012).

The use of α7 nAChR modulators in the treatment of schizophrenia is another avenue of

current research. Reduced levels of α7-subtype expression have been observed in the post-mortem

brains of schizophrenic patients (Martin-Ruiz et al., 2003). GTS-21, a partial α7 nAChR agonist, is

one of the most commonly explored treatments for schizophrenia. Clinical studies of GTS-21 have

reported improvements in neurocognitive function and gating of P50 auditory evoked potentials in

schizophrenic patients (Olincy et al., 2006). Promising results have also been observed in

schizophrenic models with another full agonist of the α7 nAChRs, TC-5619; however, these

compounds currently have limited clinical ability (Lieberman et al., 2013). Nevertheless, studies

demonstrating an improvement of cognition in schizophrenic patients following nicotine treatment

reinforce the possibility of developing selective nAChR treatments (Young et al., 2013).

The development of compounds that target the anti-inflammatory pathways of the cholinergic

system is another emerging area of therapeutic drug research. Several diseases associated with an

overstimulation of the immune system have led to the theory that pharmacological targeting of the

Page 36: A Novel Mechanism for Nicotinic Acetylcholine Receptor

21

cholinergic anti-inflammatory pathways may alleviate disease symptoms and progression (Pohanka,

2012). The GTS-21 compound, currently being investigated in the treatment of schizophrenia, also

has implications in preventing inflammatory injury through reducing TNF-α and the production of

other pro-inflammatory cytokines (Rosas-Ballina et al., 2009). Vc1.1, an α9-nAChR modulator, has

demonstrated clear analgesic effects in a variety of animal pain models with encouraging pre-

clinical results (Livett et al., 2006). Another compound, CNI-1493, is a potential pro-inflammatory

cytokine inhibitor via its selective binding to the α7 nAChR. CNI-1493 has been investigated

clinically for use as a treatment in Crohn’s disease, with chronic dosing having positive effects on

disease progression (Dotan et al., 2010). Selective α7 nAChR agonists may also have implications

in other clinical conditions of the digestive tract, such as in the treatment of postoperative ileus. The

α7 agonist, AR-R17779, potently prevents postoperative ileus in mice, restoring the propulsive

function of the entire gastrointestinal tract following abdominal surgery (The et al., 2007). Although

considerable promise with α7 nAChR compounds has been demonstrated in the laboratory, there

are currently no clinically approved drugs that target the cholinergic system of neurological and

inflammatory pathways.

Page 37: A Novel Mechanism for Nicotinic Acetylcholine Receptor

22

Table 1: Examples of Selective α7 nAChR-Modulators Investigated for the Treatment of Various

Disease States

Name Structure Use

ABT-107 5-(6-[(3R)-1- azabicyclo[2.2.2]oct-3-

yloxy]pyridazin-3-yl)-1H-indole

Tested for the treatment of Alzheimer’s

disease and cognitive deficits associated

with schizophrenia, not commercially

available.

SEN12333 5-morpholin-4-yl-pentanoic acid (4-pyridin-3-

yl-phenyl)-amide

TC-5619

N-[(2S,3S)-2-(pyridin-3- ylmethyl)-1-

azabicyclo[2.2.2]oct- 3-yl]-1-benzofuran-2-

carboxamide

Vc1.1 Peptide structure similar to α-conotoxin Potential analgesic effect for the

treatment of neuropathic pain.

CNI-1493

N,N′-bis[3,5-bis[N-

(diaminomethylideneamino)-

Cmethylcarbonimidoyl]phenyl] decanediamide

tetrahydrochloride

Investigated under clinical trials (as

Semapimod) for its ability to inhibit

inflammation and NO production.

GTS-21

3-[(3E)-3-[(2,4-

dimethoxyphenyl)methylidene]- 5,6-dihydro-

4H-pyridin-2-yl] pyridine

Under clinical testing for the treatment

of Alzheimer’s, cognitive deficits

associated with schizophrenia and anti-

inflammatory properties.

AR-R17779 (2S)- 2′H-spiro[4-azabicyclo[2.2.2]octane-2,5′-

[1,3]oxazolidin]-2’-one

Preclinical testing to prevent

postoperative ileus following abdominal

surgery.

1.3.2 nAChR-Modulators for Cancer Treatment

The connection of nAChRs with cancer development is well established with various nAChR

subtypes contributing to tumour proliferation, survival and metastasis. Antagonism of the nAChRs

has the potential to inhibit these effects and induce cancer cell apoptosis, as discussed in Section

1.2.5 nAChRs and Cancer Cell Death. Many experimental compounds, including those investigated

for the use in various disease states, may also have important implications in cancer treatment. The

following section explores the novel compounds used to selectively target the nAChR pathway to

regulate cancer development.

Overexpression of the nAChR in human NSCLC tissues has led to a number of studies

attempting to modulate the nAChRs to limit tumour progression. MG624, a synthetic α7-nAChR

antagonist has displayed both anti-proliferative and anti-angiogenic properties in SCLC. Human

microvascular endothelial cells derived from the lung (HMEC-L) co-treated with both MG624 and

Page 38: A Novel Mechanism for Nicotinic Acetylcholine Receptor

23

nicotine supressed the nicotine-associated proliferation of the cancer cells, consolidating the

nAChR-action of MG624. In vivo studies administering MG624 in a SCLC tumour xenografted

murine model demonstrated potent anti-angiogenic effects, supressing angiogenesis through

reducing both EGR-1 and FGF2 promoters (Brown et al., 2012; Thornhill et al., 2013). Similar

effects have also been observed when co-administering the α7-nAChR antagonist, α-BTX, with

cisplatin (alkylating-like chemotherapy drug). The cytotoxic effectiveness of α-BTX alone and in

combination with cisplatin was investigated in A549 lung adenocancer and SK-MES-1 lung

squamous cells. The combination of 0.1 μM cisplatin with low levels of α-BTX (1.0 ρM)

significantly increased the expression of the apoptotic factors Bax, and Caspase-3, while decreasing

Bcl-2 expression, compared to individual treatment (Balkan et al., 2012).

Naturally occurring compounds have been implicated as possible anticancer treatments due to

their interaction with the overexpressed α9-nAChRs in breast cancer. The green tea-polyphenol (–)-

epigallocatechin-3-gallate (EGCG) has been explored as a nAChR-blocker of nicotine or estradiol

(E2) induced breast cancer proliferation. MCF-7 human breast cancer cells pre-treated with EGCG

completely abolished the nicotine and E2 induced α9-nAChR activity. Co-treatment with EGCG

also inhibits the binding activity of 3H-nicotine to the α9-nAChRs of breast cancer cells. EGCG

(1 μM) was shown to competitively bind the α9-nAChR and induce receptor down-regulation,

reducing the nAChR proliferative effects of nicotine and E2 (Tu et al., 2011). Continual studies

have compared additional green and black tea extracts to the inhibitory effects of EGCG α9-

nAChRs. Human breast cancer cells with different ER status, MDA-MB-231 (ER–) and MCF-7

(ER+) were pre-treated with the extracts, having prominent effects on the inhibition of nicotine-

induced α9-nAChR protein expression. Extract treatment also inhibited colony formation of MCF-7

cells, with some extracts demonstrating a higher degree of α9-nAChR regulation than that observed

by EGCG treatment (Ho et al., 2013). Similar results have also been observed with studies treating

MDA-MB-231 cells with polyphenolic compounds, luteolin and quercetin, by significantly down-

Page 39: A Novel Mechanism for Nicotinic Acetylcholine Receptor

24

regulating α9-nAChR expression (Shih et al., 2009). The results demonstrate the potential of diet-

based extracts as possible natural modulators of the α9-nAChRs to limit cancer progression.

In addition to the naturally occurring polyphenols, various endogenous compounds have

demonstrated the potential to regulate nAChR activity. The endogenous secreted mammalian Ly-

6/urokinase plasminogen activator receptor-related protein-1 and -2 (SLURP-1 and SLURP-2),

responsible for signal transduction, immune activation and cell adhesion, has shown α7-nAChR

positive allosteric functions with possible anti-carcinogenic properties (Pettersson et al., 2009).

HT29 human colon carcinoma cells incubated with 1 μM of SLURP-1 and SLURP-2 significantly

reduced cell numbers by ~54 and 63%, respectively, compared to controls. Neither necrotic nor

apoptotic cell death was observed following fluorescent microscopy, with the researchers

concluding that the anti-proliferative effects of SLURP-1 and SLURP-2 were manifested through

the interactions with the α7-nAChR (Lyukmanova et al., 2014). The endogenous neurotransmitter,

serotonin (5-hydroxytryptamine, 5-HT), also displays possible anti-carcinogenic activity with links

to nAChR function. Treatment of HT29 and LS1034 carcinoma cells with the selective serotonin

reuptake inhibitor, sertraline, induced caspase-3 activation and Bcl-2 inhibition, causing a dose-

dependent decrease in cancer cell viability. In vivo, sertraline significantly inhibited tumour growth

in CD1 nude mice xenografted with HT29 cells (Gil-Ad et al., 2008). While the effects of serotonin

reuptake inhibitors have been shown to eliminate the effects of nicotine and nicotine-derived

nitrosamines, suggesting a link between increased serotonin levels and nAChR inhibition (Schuller,

2009).

The use of nAChR modulators as possible cancer sensitisers to current chemotherapy drugs

has been investigated. The nAChR antagonist adiphenine (ADI) was investigated in combination

with the chemotherapy drugs, doxorubicin and docetaxel. A vast range of cancer cell lines were

explored, with cell lines grouped according to their sensitivity (based on the IC50 value from the

chemotherapy treatment) to discriminate between drug-sensitive and -resistant cell lines. 250 μM

ADI treatment of resistant breast carcinoma cell lines did not affect cell proliferation; however,

Page 40: A Novel Mechanism for Nicotinic Acetylcholine Receptor

25

when combined with 0.1 μM doxorubicin + 100 μM docetaxel treatment, caused a significant ~40%

reduction in cell viability, compared to doxorubicin + docetaxel treatment alone. The sensitising

effects were observed at 4-fold lower doses of ADI, even when combined with half of the

doxorubicin + docetaxel dose. Similar effects were also observed with the nAChR antagonists

proadifen and NDNI. The researchers claimed that the chemo-sensitising effects of the nAChR

antagonists were operating through the α9 subunit, having the potential to reduce the amount of

toxic chemotherapy agents required to induce tumour cell death in vivo (Hallett et al., 2012).

Neurotoxins, commonly used to distinguish between nAChR subunit combinations through

their selectivity to distinct receptor configurations, demonstrate potential cancer modulating effects.

Studies have suggested that cancer cell proliferation can be arrested through the use of snake

neurotoxins or snail conotoxins due to their selective antagonistic effects at the α7 nAChRs (Wu et

al., 2011). One particular toxin of interest, α-cobratoxin (α-CbT), has produced anticancer effects in

various cancer cell models, both in vitro and in vivo. The treatment of mesothelioma MPP89 cells

showed a notable induction of apoptosis following treatment with α-CbT, significantly inducing

caspase-3 cleavage and down-regulating several anti-apoptotic proteins (survivin, XIAP, IAP1,

IAP2 and Bcl-xl). In vivo, orthotopically transplanted NOD/SCID mice treated with a 1/1000 LD10

dose of α-CbT (0.12 ng/kg) significantly reduced tumour growth without any signs of significant

toxicity to healthy cells (Catassi et al., 2008). Comparable effects have also been observed in A549

NSCLC orthotopically grafted nude mice and MCF-7 breast tumour-bearing nu/nu mice following

treatment with α-CbT (CATASSI et al., 2006; Mei et al., 2015).

1.3.3 Marine Toxins

The potential anticancer effects of various marine-derived agents has been long known, with

several compounds approved for the use in treating various human cancers, including cytarabine,

eribulin and monomethylauristatin E (Newman et al., 2014). However, the discovery of marine

toxins that have regulatory effects on the nAChR-system has been a new avenue for marine-derived

anticancer therapeutic research. One of the first marine toxins investigated for their potential

Page 41: A Novel Mechanism for Nicotinic Acetylcholine Receptor

26

anticancer effects were the polymeric alkylpyridinium salts (poly-APS), isolated from the marine

sponge Reniera sarai, for their AChE inhibitory activity. Fluorescence-activated cell sorting

analysis of NSCLC cells treated with poly-APS showed a selective cytotoxicity of the marine salts

towards cancer cells, compared to normal lung fibroblasts. Non-toxic doses of poly-APS also

reduced NSCLC cell-to-cell adhesion in vitro, while having no significant organ toxicity in healthy

C57BL/6N mice. The study indicated the possibility of regulating cancer cell proliferation through

the cholinesterase inhibitory effects of marine toxins (Paleari et al., 2006).

Since the discovery of poly-APS, several toxins have been extracted from marine shellfish

that act directly on nAChRs, possessing both potent and selective properties. Extracts related to the

cyclic imine group of toxins, such as gymnodimine (GYM) and spirolides, potently antagonise both

homomeric and heteromeric nAChRs (Bourne et al., 2010). Pinnatoxin (PnTX), a newly discovered

group of cyclic imines, has demonstrated high antagonistic affinity towards α7-nAChR. Recordings

of gamma oscillations in response to tetanic stimulation of hippocampal CA1 slices of male

Sprague-Dawley rats showed a reduction in gamma pulse shape discrimination and spike count in

the presence of PnTX E and F, similar to that seen with the selective α7 nAChR antagonist

methyllycaconitine (Hellyer et al., 2011). Furthermore, studies suggest that cyclic amines

specifically target the α7-subtype, with relatively less potent effects at the heteromeric subtypes,

such as α4β2-nAChRs (Hellyer et al., 2011; Kharrat et al., 2008). The potential nAChR-

antagonistic effects of pinnatoxins suggest their possible use in elucidating the contribution of

nAChRs to cancer development and as therapeutics for regulating cancer cell proliferation.

1.4 Aims and Hypothesis

Current literature has identified the various nAChR pathways that contribute to the

development of cancer. The majority of the published studies focus on the effects of nAChR

agonists, such as nicotine, on enhancing the carcinogenic properties of various lung, breast and

Page 42: A Novel Mechanism for Nicotinic Acetylcholine Receptor

27

colon cancer cell lines. However, investigation into the role of nAChRs in the development of

glioblastoma is limited.

Recent studies investigating the use of nAChR antagonists for regulating cancer development

have highlighted the possible advantages of targeting these receptors directly. The use of these

antagonists has often been investigated in the presence of nicotine, where the main effects of the

investigational compounds are to negate the effects of nicotine on progressing cancer development.

However, many carcinoma cell types, such as mesothelioma and colon cancer types, display an

increase in nAChR expression independent of exogenous influence from compounds such as

nicotine (Egleton et al., 2008). These carcinoma cell types also contain the complete functional loop

of ACh signalling that allows cancer cells to promote proliferation through the autocrine activation

of nAChRs (Lau et al., 2013; Song et al., 2008). The present study looked to target these

endogenous functions of nAChRs using a range of pilot studies to better understand the mechanism

behind nAChR antagonist-induced cancer cell death, where clinical application of the compounds is

not an immediate focus of the various investigations performed.

In this research, the effects of both established and novel nAChR antagonists in regulating

carcinogenesis have been explored directly, and in combination with common chemotherapy

agents, utilising a variety of different methods. GYM and PnTX are considered novel compounds

due to the lack of published literature relating to their effects on carcinogenesis. Details regarding

the various compounds, selectivity towards nAChRs and the concentration ranges investigated have

been provided in Table 3 (Section 2.2.2. Cell Viability Assay). Specifically the aims of this research

were:

Aim I: To examine the cytotoxic effects of nAChR modulators on a variety of carcinoma cell

types, and to determine the key nAChR subtypes involved in mediating these

responses.

Page 43: A Novel Mechanism for Nicotinic Acetylcholine Receptor

28

Aim II: To investigate the mechanism(s) of the newly identified role of nAChR modulators in

inducing carcinoma cell death.

Aim III: To determine whether inhibiting common apoptotic pathways would reduce the

cytotoxic effects of nAChR modulators.

Aim IV: To explore the synergistic capacity of the most effective nAChR modulators

identified in Aims I-III to sensitise cancer cells to common chemotherapy agents.

The results of this research suggest a possible mechanism for nAChR antagonist induced cancer cell

cytotoxicity, as highlighted in the following sections. The implications of which are discussed in

context with the current literature.

Page 44: A Novel Mechanism for Nicotinic Acetylcholine Receptor

29

2 Methods

2.1 Cell Culture

2.1.1 Materials

Dulbecco’s Modified Eagle Medium (DMEM), foetal bovine serum (FBS), 0.5% Trypsin

EDTA, and Penicillin/Streptomycin were purchased from Gibco, Life Technologies (California,

USA). Phosphate buffered saline (PBS) was purchased from Invitrogen, Life Technologies

(California, USA). Haemocytometer, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

(MTT) and Roswell Park Memorial Institute (RPMI) 1640 Medium was purchased from Sigma-

Aldrich (St Louis, MO, USA). Dimethyl sulfoxide (DMSO) was purchased from Scharlau

(Sentmenat, Barcelona, Spain). GraphPad Prism 5 was purchased from GraphPad Software (La

Jolla, CA, USA).

2.1.2 Cell Lines and Cell Culture

A total of eight human carcinoma cell lines were used in this research, as described in Table

2. MDA-MB-231, SKBR3, HT29 and PC3 cell lines were a gift from Associate Professor R.

Rosengren of the Pharmacology Department, University of Otago (Dunedin, NZ). U87, U373 and

LN18 cell lines were a gift from Dr. K. Greish of the Pharmacology Department, University of

Otago (Dunedin, NZ). A549 cells were a gift from Dr. G. Giles of the Pharmacology Department,

University of Otago (Dunedin, NZ).

Page 45: A Novel Mechanism for Nicotinic Acetylcholine Receptor

30

Table 2: Details of the Carcinoma Cell Lines

Cell Line Disease State Cell Type Tissue

MDA-MB-231 Breast adenocarcinoma Epithelial

Mammary gland/breast,

derived from metastatic

site (pleural effusion)

HT29 Colorectal adenocarcinoma Epithelial Colon

PC3 Grade IV prostate

adenocarcinoma Epithelial

Prostate, derived from

metastatic site (bone)

SKBR3 Breast adenocarcinoma Epithelial

Mammary gland/breast,

derived from metastatic

site (pleural effusion)

A549 Lung carcinoma Epithelial Lung

U87 Grade IV astrocytoma Epithelial Brain

LN18 Grade IV astrocytoma Epithelial Brain/cerebrum, right

temporal lobe

U373 Grade IV astrocytoma Epithelial Brain

MDA-MB-231, HT29, PC3, A549 and SKBR3 cells were cultured in DMEM with 10% heat

inactivated FBS, 100 units/mL penicillin and 100 µg/mL streptomycin. U87, U373 and LN18 cells

were cultured in RPMI medium with 10% heat inactivated FBS. Cells were incubated at 37°C in

5% CO2 humidified conditions. Cell lines grew to ~80% confluence in 75 cm2 culture flasks before

passaging. The sub-culturing process involved discarding the media, washing with 2 mL trypsin

solution (1x trypsin, 0.5% EDTA, PBS), and incubated in 2 mL trypsin solution until cells no

longer adhered. 5 mL fresh media was added to the trypsin-cell solution, transferred to a 15 mL

falcon tube and centrifuged at 1200 rpm for 3 minutes at 4°C. The supernatant was discarded, with

the cell pellet re-suspended in 5 mL fresh medium. Cells were counted visually using a

haemocytometer under 20x magnification. The number of cells were counted in all four quadrants,

averaged and multiplied by 104, to give the cell number per mL of media. Cells were then seeded

according to experiment requirements.

2.1.3 MTT Assay

The MTT colourmetric assay was employed to determine cell viability following treatment

(refer below). The assay utilises the reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl

tetrazolium bromide) (MTT) by NAD-dependant enzymatic activity, to form formazin crystals, as

Page 46: A Novel Mechanism for Nicotinic Acetylcholine Receptor

31

an indicator of metabolically active viable cells (Mosmann, 1983). The MTT solution was prepared

at 5 mg/mL in PBS and combined with cells at a final concentration of 0.5 mg/mL. The MTT-media

solution was incubated for ~3 hours in the humidified incubator, solution removed, and replaced

with an equal volume of DMSO (to dissolve the formazin crystals, forming a purple solution). The

reduced MTT solution was analysed spectrophotometrically at 560 nm and processed using

microplate manager 5.2.1 software to quantify the amount of viable cells.

2.1.4 Statistical Analysis

For all cellular viability MTT assay results, GraphPad Prism software was utilised to average

and normalise the absorbance values as a percentage of the control mean ± the standard error of the

mean (SEM) following technical replicates of the method. Significance was determined by

analysing the difference of the means, using either a one-way or a two-way analysis of variance

(ANOVA) with a Bonferroni post-hoc test, where the p-value between the compared groups was

less than 0.05 (p<0.05).

2.2 Cell Viability Assay

2.2.1 Materials

Acetylcholine chloride, (–)-nicotine hydrogen tartrate salt, methyllycaconitine citrate,

serotonin creatine sulphate complex, adiphenine hydrochloride and isopropanol were purchased

from Sigma-Aldrich (St Louis, MO, USA). Mecamylamine hydrochloride was purchased from

Abcam (Cambridge, UK). Pinnatoxin G and Gymnodimine were gifted from Assoc. Prof. S. Kerr of

the Pharmacology Department, University of Otago (Dunedin, NZ), and Dr Paul McNabb and Dr

Andrew Selwood of the Cawthron Insititute (Nelson, NZ).

2.2.2 Cell Viability Assay

MDA-MB-231, HT29, PC3, U87, U373 and LN18 cell lines were seeded in 96-well plates at

a density of 3,000 cells/100µL/well in their respective media, 24 hours prior to treatment. Cells

were treated with acetylcholine (ACh), nicotine (NIC), mecamylamine (MECA), pinnatoxin G

Page 47: A Novel Mechanism for Nicotinic Acetylcholine Receptor

32

(PnTX), gymnodimine (GYM), methyllycaconitine (MLA), adiphenine (ADI) or serotonin (5-HT)

at final well concentration ranging from 0.1 µM to 1000 µM. The vehicle control contained the

highest concentration of the solvent for the corresponding treatment. Treatment profiles are

displayed in Table 3. Treated cells were analysed by MTT assay at 24, 48 and 72 hours post

treatment. The absorbance values for the treated samples were normalised against the untreated

group for each corresponding time point prior to statistical analysis.

Table 3: Cell Viability Assay Treatment Profile

Treatment nAChR Selectivity Solvent

Maximum

Volume of

Vehicle

Control (µL)

Minimum

Final Well

Concentration

(µM)

Maximum

Final Well

Concentration

(µM)

Acetylcholine Broad-Spectrum

Agonist H2O 10 0.1 1000

Nicotine Broad-Spectrum

Agonist H2O 10 0.1 1000

Mecamylamine Broad-spectrum

Antagonist H2O 10 0.1 1000

Pinnatoxin G α7, α4β2 Antagonist Isopropranolol 0.193 0.1 5

Gymnodimine α7, α4β2, α3β2

Antagonist H2O 10 0.1 50

Methyllycaconitine α7 Antagonist H2O 10 0.1 100

Adiphenine

Antagonist at α3 or

α4 Containing

Heteromeric

nAChRs

H2O 10 0.1 1000

Serotonin

Secondary Effects

on Decreasing

nAChR Expression

H2O 10 0.1 1000

2.3 Western Blot

2.3.1 Materials

Albumin from bovine serum (BSA), 3-(N-morpholino) propanesulfonic acid (MOPS),

Tween® 20, sodium dodecyl sulphate (SDS), Trizma®-base, Trizma®-hydrochloride, Triton® X-100,

Page 48: A Novel Mechanism for Nicotinic Acetylcholine Receptor

33

2-mercaptoethanol (MeSH) and tetramethylethylenediamine (TEMED) were purchased from

Sigma-Aldrich (St Louis, MO, USA). 30% acrylamide/bis solution 19:1 and ammonium persulfate

(APS), were purchased from Bio-Rad Laboratories Ltd (Hercules, CA, USA).

Ethylenediaminetetraacetic acid as purchased from BDH/VWR International Ltd (Radnor, PA,

USA). PageRuler™ Prestained Protein Ladder, CL-XPosure Film, Pierce® Protein Inhibitor Tablets,

Pierce® BCA Protein Assay Kit, and dithiothreitol (DTT) were purchased from Thermo-Fisher

Scientific (Waltham, MA. USA). NuPage LPS Sample Buffer and iBlot® Gel Nitrocellulose transfer

Stacks were purchased from Novex, Life Technologies (California, USA). Amersham ECL Prime

Western Blotting Detection Reagents were purchased from GE Healthcare (Little Chalfont, UK).

2.3.2 Antibodies

Rabbit monoclonal anti-nicotinic acetylcholine receptor alpha 4 antibody, rabbit polyclonal

anti-nicotinic acetylcholine receptor alpha 7, rabbit polyclonal anti-CHRNA9 antibody (alpha 9),

goat polyclonal secondary antibody to mouse IgG, and goat polyclonal secondary antibody to rabbit

IgG were purchased from Abcam, Inc (Cambridge, UK). Mouse monoclonal anti-β-actin antibody

was purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The primary anti-α4-

nAChR, anti-α7-nAChR, and anti-β-actin antibodies were used at 1:25,000 dilution; while primary

anti-α9-nAChR was used at 1:12,500. The secondary antibodies were incubated at 1:10,000

dilution. Antibodies were diluted in 10 mL PBS containing 1% w/v BSA and 0.1% v/v Tween.

2.3.3 Protein Extraction

MDA-MB-231, HT29, PC3, U87, U373 and LN18 cell lines were seeded in 6-well plates at a

density of 200,000 cells/mL/well in their respective media. For baseline levels of nAChR

expression, cells were grown until ~80% confluence before harvesting. Treated samples were

allowed to adhere for 24 hours prior to treatment, and then harvested 72 hours post treatment, with

appropriate untreated controls. MDA-MB-231 cells used in the time-course study were treated after

24 hour adherence with 10 µM nicotine, and harvested at 30 min, 60 min, 2 h, 6 h, and 24 h post-

treatment, with corresponding untreated controls. Cell samples were harvested by removing media,

Page 49: A Novel Mechanism for Nicotinic Acetylcholine Receptor

34

washing with PBS and incubating in 200 µL lysis buffer (NaCl 150 mM, Tris Base 50 mM, SDS

0.05%, Triton X100 1%, AEBSF, aprotinin, bestatin, E64, leupeptin, pepstatin A, pH 8.0) on ice

for 30 min. Cell lysate samples were collected, sonicated, and homogenised by centrifugation

(18,000 rpm, 8 min, 4°C). The protein concentration of the lysate supernatant was measured using

the Pierce® BCA Protein Assay Kit, combined with loading buffer, reducing buffer (DTT 500 mM)

and heat denatured at 70°C for 10 min.

2.3.4 Western Blotting

Loading of 5 µg protein per 20 µL per well and protein reference ladder (7 µL per well) was

resolved by 8% sodium dodecyl sulphate-polyacrylamide gel electrophoresis in MOPS buffer

(MOPS 50 mM, Tris Base, 50 mM, SDS 1%, EDTA 1 mM, pH 7.7) at 150 V for 65 min. Resolved

proteins were electroblotted onto a nitrocellulose membrane (20 V for 1 min, 23 V for 4 min, 25V

for 2 min) and blocked (5% nonfat milk solution in PBS-Tween 0.1%) for 100 min at room

temperature. The membranes were then incubated in the anti-nAChR primary antibody overnight at

4°C with gentle agitation, washed in PBS-Tween 0.1% (6 x 5 min), and incubated in the appropriate

secondary antibody at room temperature for 60 min. Membranes were washed as described above,

visualised with enhanced chemiluminescence reagent (used at a 1:1 concentration, as per the

manufacturer’s instructions) and immediately exposed to X-ray film for developing. The

membranes were then stripped (Tris HCl 62.5 mM, SDS 2%, MeSH 0.1 M, pH 6.8) for 30 min at

50°C, washed, blocked and re-probed with anti-β-actin primary antibody (for normalisation of

loading). Developed films were scanned using an imaging densitometer and densitometry data for

the individual Western blot bands determined by Quantity One software. GraphPad Prism software

was used to determine the mean density of each band.

Page 50: A Novel Mechanism for Nicotinic Acetylcholine Receptor

35

2.4 Immunocytochemistry

2.4.1 Materials

Paraformaldehyde and Hoechst BisBenzimide H33342 trihydrochloride were purchased from

Sigma-Aldrich (St Louis, MO, USA). Alexa Fluor® 488 Goat Anti-Rabbit IgG (H+L) Antibody and

ProLong® Gold antifade reagent were purchased from Molecular Probes, Life Technologies

(California, USA).

2.4.2 Sample

MDA-MB-231 cells were seeded at 10,000 cells/500µL/well onto sterile micro-circular

coverslips in 24-well plates for 72 hours. Cells were fixed with 4% paraformaldehyde- PBS pH 7.4

for 15 min at room temperature. The paraformaldehyde fixed samples were treated with either

0.25% Triton X-100-PBS (for permeabilisation of cell membranes) or PBS (non-permeabilised) for

10 min. Following this step, samples were blocked with 1% BSA in PBS-Tween for 30 min, then

incubated with primary antibody (nAChR-α7 or nAChR-α9 at 1:100 in PBS-Tween) overnight at

4oC. The probed samples were incubated with fluorescent secondary antibody (488 Goat Anti-

Rabbit at 1:500 in PBS-Tween) for 1 hour under constant oscillation at room temperature in the

dark. The coverslips were briefly incubated with 1 µg/mL Hoescht stain for 1 min under dark

conditions, dried, and transferred to microscope slides, bound by ProLong Gold gel for

preservation. The prepared microscope slides were dried and stored in the dark until visualisation.

2.4.3 Fluorescence Microscopy

Cells were visualised using an upright Nikon Eclipse Ni-E Motorised microscope with

ultraviolet or blue illumination from a mercury fibre illuminator lamp through a 100x Nikon

florescence oil immersion objective (numerical aperture 1.3). Images were captured using NIS-

Elements Microscope Imaging Software through a 4,6-diamidino-2-phenylindole (DAPI) or

Fluorescein isothiocynate (FITC) filter. For image adjustments, the captured images were merged

and examined using ImageJ software.

Page 51: A Novel Mechanism for Nicotinic Acetylcholine Receptor

36

2.5 Flow Cytometry

2.5.1 Materials

Propidium iodide (PI), citric acid and 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

(HEPES) were purchased from Sigma-Aldrich (St Louis, MO, USA). Tetramethylrhodamine, ethyl

ester, perchlorate (TMRE) and Annexin V (Alexa Fluor® 488 conjugate) were purchased from

Molecular Probes, Life Technologies (California, USA). Di-sodium hydrogen orthophosphate

(Na2HPO4) and calcium chloride (CaCl2) was purchased from BDH/VWR International Ltd

(Radnor, PA, USA). Ribonuclease A was purchased from Invitrogen, Life Technologies (California,

USA). Ethanol and sodium chloride (NaCl) were purchased from Scharlau (Sentmenat, Barcelona,

Spain). Cisplatin (CDDP) was purchased from Ebewe Pharma (Unterach, Austria).

2.5.2 Cell Treatment and Staining

MDA-MB-231, HT29, PC3, and LN18 cell lines were seeded in 6-well plates at a density of

100,000 cells/mL/well in their respective media. Samples were allowed to adhere for 24 hours prior

to treatment with either MECA 1000 µM, PnTX 5 µM, ADI 1000 µM, 5-HT 1000 µM or CDDP

20 µM, with appropriate untreated controls. Cell samples were harvested using trypsin at 72 hours

post-treatment, and probed using various fluorescent stains, including PI, TMRE, or a combination

of Annexin V and PI.

Propidium Iodide

Cells were washed in cold PBS and spun at 2000 rpm for 3 min. The samples were fixed by

discarding the supernatant and mixing the cell plate with 500 µL per tube of 4°C ethanol 70%, then

stored at 4°C for 30 minutes. The fixed cell solution was washed twice in phosphate-citrate buffer

(Na2HPO4 0.192 M, Citric Acid 0.004 M), spinning at 2000 rpm for 3 min between each wash. The

cell supernatant was removed then treated with 50 µL per tube of 100 µg/mL Ribonuclease A,

followed by 200 µL per tube of 50 µg/mL propidium iodide, and kept on ice until analysis.

TMRE

Page 52: A Novel Mechanism for Nicotinic Acetylcholine Receptor

37

The treated cell samples were re-suspended in 500 µL per tube of appropriate media

following trypsin harvesting. 2 µL per tube of 10 µM TMRE was added to the samples under dark

conditions, and incubated at 37°C protected from light for 10 minutes. Cell samples were washed in

PBS, spun at 2000 rpm for 3 min, and re-suspended in 300 µL per tube of media. Stained samples

were kept on ice, protected from light until analysis.

Annexin V/PI

The cell samples were washed twice in cold PBS, with spinning at 2000 rpm for 3 min, and

re-suspended in 100 µL per tube Annexin V binding buffer (HEPES 0.1 M, NaCl 1.4 M, CaCl2 25

mM, pH 7.4). 2 µL per tube Annexin V conjugate and 2 µL per tube of 50 µg/mL PI were added to

the cell solution and incubated at room temperature for 15 min protected from light. 400 µL per

tube of additional Annexin V binding buffer was combined to each sample and stored on ice until

analysis.

2.5.3 Fluorescence-Activated Cell Sorting (FACS)

Stained samples were loaded into FACS tubes and run through the FACSCalibur™. Data was

acquired using CellQuestPro software with an acquisition criteria of 10,000 events per sample, and

analysed using Flowing Software 2.5.0. The PI cell samples were measured using the yellow 585/42

nm (FL2-H) detector and analysed to generate cell cycle phase distribution data. The data was gated

to ensure only single cells were analysed, with the cell cycle phases (sub-G1, Go/G1, S and G2/M)

determined manually based on the methods described in the literature (Wlodkowic et al., 2011).

TMRE samples were measured via the same yellow 585/42 nm detector and histograms produced to

quantify the change in TMRE peaks between control and treated samples. Double-stained, Annexin

V and PI samples were measured using the green 530/30 nm (FL1-H) and yellow 585/42 nm (FL2-

H) detectors, graphed accordingly on a dot-plot with quadrants for double stain and individual

stains measured. Values were entered into GraphPad Prism software and analysed using a one-way

ANOVA with a Bonferroni post-hoc test, where the significance was determined by p<0.05.

Page 53: A Novel Mechanism for Nicotinic Acetylcholine Receptor

38

2.6 Intracellular Calcium

2.6.1 Materials

Fura-2 acetoxymethyl ester (AM) was purchased from Abcam, Inc (Cambridge, UK).

Potassium chloride (KCl) was purchased from Scharlau (Sentmenat, Barcelona, Spain). Glucose,

and sodium bicarbonate (NaHCO3) were purchased from Sigma-Aldrich (St Louis, MO, USA).

Potassium di-hydrogen orthophosphate (KH2PO4) was purchased from BDH/VWR International

Ltd (Radnor, PA, USA). Magnesium sulphate (MgSO4) was purchased from Merck Millipore

(Darmstadt, Germany).

2.6.2 Fura-2 AM Fluorescence Measurement

MDA-MB-231, HT29 and PC3 cells were seeded at 40,000 cells/100µL/well in 96-well black

plates for fluorescence. Cells were allowed to adhere overnight before being loaded with 100 µL

per well of 4 µM of the Ca2+ indicator Fura-2 AM (from 1 mM stock in DMSO) in Hanks Balanced

Salt Solution (HBSS; NaCl 0.137 M, KCl 5.4 mM, Na2HPO4 0.25 mM, glucose 0.01%, KH2PO4

0.44 mM, CaCl2 1.3 mM, MgSO4 1.0 mM, NaHCO3 4.2 mM) for 60 min in a humidified incubator

protected from light. The cells were then washed in HBSS and incubated in fresh HBSS 100 µL per

well for 30 min at room temperature protected from light to allow Fura-2 AM cleavage. Cells

transfected with Fura-2 were treated with Ach 10 µM, MECA 1000 µM, PnTX 5 µM, GYM 10

µM, ADI 1000 µM, MLA 100 µM or 5-HT 1000 µM at time = 0 min. Fura-2 fluorescence was then

measured every minute for the first 20 min, 1 h, 2 h, 4 h, and 6 h post-treatment. Softpro Max

Software 4.8 was used to determine Fura-2 fluorescence using a fluorescent plate reader at an

excitation of 340 nm (λ1) and 380 nm (λ2), with an emission of 510 nm, at each time point. The

fluorescence intensity ratio (R = Fλ1/Fλ2) of Fura-2 fluorescence detected at 510 nm for each time

point was then calculated, indicating the ratio of Fura-2 in a completely Ca2+-saturated state against

Fura-2 in a Ca2+-free state. GraphPad Prism software was used to construct time-course graphs of

the fluorescence intensity ratio to display the difference between treatment groups over time.

Page 54: A Novel Mechanism for Nicotinic Acetylcholine Receptor

39

2.7 Cell Death Inhibitors

2.7.1 Materials

1,2-bis(o-aminophenoxy)ethane-N,N,N’,N’-tetraacetic acid – acetoxymethyl ester (BAPTA-

AM), N-Acetyl-Leu-Leu-Nle-al (ALLN) and N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl

ketone (zVAD-FMK) were purchased from Abcam, Inc (Cambridge, UK). Cyclosporin A (CsA)

was purchased from Sigma-Aldrich (St Louis, MO, USA).

2.7.2 Cell Culture and Inhibitors

MDA-MB-231, HT29, PC3, and LN18 cell lines were seeded in 96-well plates at a density of

3,000 cells/100µL/well in their respective media. Cells were allowed to adhere for 24 hours, then

treated with specific concentrations of various cell death inhibitors, as shown in Table 4, alone and

in combination with varying concentrations of PnTX, MECA, MLA, ADI or 5-HT. Treated cell

viability was measured via MTT assay at 48 and 72 hours post-treatment.

Table 4: Cell Death Inhibitor Treatment Profile

Cell Line

Final Well Concentration (µM)

CsA ALLN BAPTA-AM zVAD

MDA-MB-231 10 5 1 30

HT29 1 1 1 20

PC3 1 5 10 30

LN18 1 1 0.5 10

2.7.3 Isobologram

Following treatment of the various cancer cell types with cell death inhibitors, as described in

Section 2.7.2, an isobologram was constructed to investigate the relationship between CsA and

MECA in reducing cell viability. MDA-MB-231 cells were seeded in 96-well plates at a density of

3,000 cells/100µL/well in DMEM. The cells were allowed to adhere for 24 hours before treatment.

Cells were treated with CsA at a range of 0.1 to 40 µM, MECA at a range of 10 to 1500 µM, and

combinations of the two (CsA 0.1, 1.0 or 5.0 µM; MECA 100 or 500 µM). The treated MDA-MB-

Page 55: A Novel Mechanism for Nicotinic Acetylcholine Receptor

40

231 cells were then analysed by MTT assay 72 hours post treatment. IC50 values for CsA and

MECA were calculated for the individual compounds and in combination to determine the overall

cytotoxic relationship between the two compounds. An isobologram was constructed according to

the methods described by (Tallarida, 2006) from the IC50 values calculated using OriginPro

software (OriginLab Corporation, MA, USA).

2.8 Combination Therapy

2.8.1 Materials

Doxorubicin (DOXO) and Docetaxel (DTX) were purchased from Cayman Chemical Company

(Michigan, USA).

2.8.2 Chemotherapy Combination Therapy

The methods for chemotherapy combination treatment of various cancer cell lines were

adopted from (Hallett et al., 2012). MDA-MB-231, HT29, PC3, A549 and SKBR3 cells were

seeded in 96-well plates at a density of 20,000 cells/100µL/well in their respective media. Cells

were allowed to adhere for 4 hours, followed by compound addition of various concentrations of

chemotherapy agents and nAChR antagonists, with appropriate vehicle controls. Chemotherapy

final well concentrations included doxorubicin at 0.1-1.0 µM and docetaxel at 100 µM. After 24

and 48 hours post-treatment, cell viability was measured via the MTT assay.

Page 56: A Novel Mechanism for Nicotinic Acetylcholine Receptor

41

3 Results

3.1 Effects of nAChR Antagonists on Cell Viability

The link between nAChR activation and cancer development is well established. Several

nAChR subtypes have been shown to contribute to tumour proliferation, survival and metastasis

(refer to Section 1.2 nAChRs in Cancer Development). To test the hypothesis that the more selective

modulators of the nAChRs would display the most potent cytotoxic effects, a colourimetric cell

viability assay was used to measure the effects of exposure of a range of carcinoma cell types (refer

Table 2) to both established and novel nAChR modulators (Aim I).

3.1.1 Cell Viability Assay

Initial experiments were undertaken using the cell viability assay to establish an effective

modulating concentration for each test compound. Contrary to expectations, the nAChR agonists

acetylcholine (ACh) and nicotine (NIC) did not exhibit a consistent change in cell viability across

the range of concentrations studied (0.1 µM to 1000 µM), with the exception of HT29 cells (data

not shown). ACh and NIC concentrations of 10 µM were therefore chosen based on the observed

effects on HT29 cells, an established history of increased cancer cell viability and a potential

modification of nAChR expression observed at this concentration in published studies (Al-Wadei et

al., 2012; Dasgupta et al., 2009). Following treatment with ACh 10 µM for 72 hours, a significant

increase (P < 0.001) in HT29 viability was detected, corresponding to a 28.50% increase in cell

viability compared to untreated control cells (Figure 3A). A similar increase in cell viability was

seen following treatment of HT29 cells with NIC 10 µM (19.91% increase in cell viability) (Figure

3B). No significant increase in cell viability was seen in any other cell line when treated with either

ACh or NIC at 10 µM. Furthermore, visual analysis of MDA-MB-231, HT29 and PC3 cell lines,

provided for reference only, at 72 hours post treatment showed no changes in cell morphology or

confluency, compared to control cells (refer to microscopic images in Figure 3A and Figure 3B).

Page 57: A Novel Mechanism for Nicotinic Acetylcholine Receptor

42

A

0 24 48 720

20

40

60

80

100

120

140

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 ns ns ns

HT29 ns ns ***

PC3 ns ns ns

U87 ns ns ns

LN18 ns *** *

U373 ns ns ns

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

B

0 24 48 720

20

40

60

80

100

120

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 ns ns ns

HT29 ns ns **

PC3 ns ns ns

U87 ns * ***

LN18 * ns *

U373 ns ** ***

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 3: The determination of MDA-MB-231, HT29, PC3, U87, LN18 and U373 cell viability following treatment with

ACh 10 µM (A) or NIC 10 µM (B). Cells were treated following a 24 hour seeding period, with cell viability tested at 24

hour intervals over a 72 hour study timeframe, with viability established using the MTT assay. Bright-field microscopic

images of MDA-MB-231, HT29 and PC3 treated cells (primary image) versus control cells (inset image) at 72 hours

are shown (20x magnification). Values are displayed as the mean percent of the viability of control (untreated) cells,

with error bars representing SEM (n = 2). A two-way ANOVA with a Bonferroni post-hoc test was used to determine

significance (* P < 0.05, ** P < 0.01, *** P < 0.001).

MDA-MB-231

HT29

PC3

MDA-MB-231

HT29

PC3

ACh 10 μM

NIC 10 μM

Page 58: A Novel Mechanism for Nicotinic Acetylcholine Receptor

43

An array of nAChR antagonists were investigated for their effects on cancer cell viability. The

selectivity of the antagonists towards specific nAChR subtypes ranged from relatively non-selective

to highly selective, as summarised in Table 3. Again, concentration dependence studies were

conducted on each compound to determine which concentration had the greatest effect on cell

viability (data not shown). Contrasting effects on cell viability, compared to ACh and NIC, were

seen for the majority of compounds tested. Mecamylamine (MECA) 1000 µM, pinnatoxin G

(PnTX) 5 µM, serotonin (5-HT) 1000 µM and adiphenine (ADI) 1000 µM caused a significant

decrease in cell viability after 72 hours of treatment for all cell lines tested (P < 0.01 for all

treatments and cell lines). MECA 1000 µM (Figure 4A) and 5-HT 1000 µM (Figure 4D) produced

the largest range of viability results; causing a 21.53% (U87) to 83.28% (HT29) and 16.66% (U373)

to 72.74% (PC3) decrease in cell viability, respectively. ADI 1000 µM (Figure 4E) treatment

caused at least 50% decrease in cell viability by the 48 and 72 hour time points in all cell lines

tested, with MDA-MB-231s demonstrating the greatest susceptibility to ADI treatment (97.62%

decrease in cell viability at 72 hours compared to control). PnTX 5 µM (Figure 4B) was the most

potent of the antagonists studied, causing between 28.11% (U87) to 68.32% (HT29) decrease in cell

viability at a concentration 200-fold lower than all other nAChR antagonists tested. Gymnodimine

(GYM) 10 µM (Figure 4C) and methyllycaconitine (MLA) 100 µM (Figure 4F) produced the least

significant change in cell viability of the compounds tested.

Bright-field microscopic images of MDA-MB-231, HT29 and PC3 cell lines, as a

representation of all the cell lines tested, at 72 hours post treatment showed obvious visual changes

in cell morphology and confluency, consistent with the viability results. MECA, 5-HT and ADI

treatment showed a widespread decrease in cell viability, compared to control (untreated) cells. A

similar change in cell viability was seen following PnTX treatment, with a significant depletion in

viable cells compared to control cells. Microscopic images following GYM and MLA treatment

were consistent with the viability assay results, with little observable changes to cell morphology or

confluency.

Page 59: A Novel Mechanism for Nicotinic Acetylcholine Receptor

44

A

0 24 48 720

10

20

30

40

50

60

70

80

90

100

110

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 *** *** ***

HT29 *** *** ***

PC3 *** *** ***

U87 ns * **

LN18 *** *** ***

U373 * *** ***

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

B

0 24 48 720

10

20

30

40

50

60

70

80

90

100

110

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 ns ** ***

HT29 *** *** ***

PC3 ns *** ***

U87 *** *** ***

LN18 *** *** ***

U373 ** *** ***

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

MDA-MB-231

HT29

PC3

MDA-MB-231

HT29

PC3

MECA 1000 μM

PnTX 5 μM

Page 60: A Novel Mechanism for Nicotinic Acetylcholine Receptor

45

C

0 24 48 720

10

20

30

40

50

60

70

80

90

100

110

120

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 ns * **

HT29 ns ns ns

PC3 ns ns ns

U87 ns ns ns

LN18 ns ns ns

U373 ns ns ns

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

D

0 24 48 720

10

20

30

40

50

60

70

80

90

100

110

120

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 *** *** ***

HT29 ns *** ***

PC3 ns *** ***

U87 *** *** ***

LN18 *** ns ***

U373 ** *** ***

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

MDA-MB-231

HT29

PC3

MDA-MB-231

HT29

PC3

GYM 10 μM

5-HT 1000 μM

Page 61: A Novel Mechanism for Nicotinic Acetylcholine Receptor

46

E

0 24 48 720

10

20

30

40

50

60

70

80

90

100

110

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 *** *** ***

HT29 *** *** ***

PC3 *** *** ***

U87 ns *** ***

LN18 *** *** ***

U373 *** *** ***

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

F

0 24 48 720

20

40

60

80

100

120

140

MDA-MB-231

HT29

PC3

U87

LN18

U373

Time (Hours)

24 48 72

MDA-MB-231 ns ns ns

HT29 ns ns ns

PC3 ns ns **

U87 ns ** ns

LN18 ns ns ns

U373 ns ns *

ns = not significant, * = p<0.05, ** = p<0.01, *** = p<0.001

Time (Hours)

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 4: The evaluation of MDA-MB-231, HT29, PC3, U87, LN18 and U373 cell viability following treatment with

MECA 1000 µM (A), PnTX 5 µM (B), GYM 10 µM (C), 5-HT 1000 µM (D), ADI 1000 µM (E) or MLA 100 µM (F).

Cells were treated following a 24 hour seeding period, with cell viability tested using the MTT assay at 24 hour

intervals over a 72 hour study timeframe. Bright-field microscopic images of MDA-MB-231, HT29 and PC3 treated

cells (primary image) versus control cells (supplementary image) at 72 hours are shown (20x magnification). Values

are displayed as the mean percent of the control viability, with error bars representing SEM (n = 2). A two-way

ANOVA with a Bonferroni post-hoc test was used to determine significance (* P < 0.05, ** P < 0.01, *** P < 0.001).

MDA-MB-231

HT29

PC3

MDA-MB-231

HT29

PC3

ADI 1000 μM

MLA 100 μM

Page 62: A Novel Mechanism for Nicotinic Acetylcholine Receptor

47

3.1.2 Western Blot - Basal nAChR Levels

To investigate the possible correlation between nAChR subtype expression and susceptibility

to nAChR antagonism-induced changes in cell viability, the basal levels of nAChR subtypes in the

cell lines tested were determined using Western blot analysis. The relative densities of the bands on

the Western blots were quantified using laser densitometry in comparison to β-actin.

As described in the literature, the relative abundance of the different nAChR subtypes within

a cell varies depending on cell type. HT29 cells demonstrated the greatest level of nAChR

expression across all cell lines, as shown in Figure 5. HT29 cell expression of the α4, α7 and α9

nAChR subtypes were shown to be at least 142.6%, 7.43% and 86.80%, higher than the expression

observed in all other cell lines for the respective subtypes. PC3 cells showed a similar level of α7

nAChR expression as the HT29 cells; while LN-18 possessed the greatest total nAChR expression

among the glioblastoma cell lines.

Page 63: A Novel Mechanism for Nicotinic Acetylcholine Receptor

48

MD

A-M

B-2

31

HT29

PC3

U87

LN

18

U37

3

0.00

0.25

0.50

0.75

1.00

1.25

1.50

4 nAChR

7 nAChR

9 nAChR

Den

sity

Rati

o

Figure 5: Western blot analysis showing the basal level of α4, α7 and α9 nAChR expression in MDA-MB-231, HT29,

PC3, U87, LN18 and U373 cancer cell lines. β-actin protein was used as a loading control to ensure equal loading of

proteins. Each well was adjusted according to the corresponding β-actin control. The columns in the graph represent

the density ratio for each nAChR subtype.

3.1.3 Immunocytochemistry

To further demonstrate the presence and localisation of the nAChR subtypes on cancer cells,

immunocytochemistry was performed on untreated MDA-MB-231 cells. The immunocytochemistry

images for non-permeabilised and permeabilised cell samples stained for the nAChR α7 and α9

subtypes are presented in Figure 6 and Figure 7, respectively. Staining with the α7 nAChR subtype

antibody in non-permeabilised cell samples showed widespread expression (Figure 6A). Subtype

expression was clearly displayed on the surface of the cancer cells, with much of the α7 labelling

Page 64: A Novel Mechanism for Nicotinic Acetylcholine Receptor

49

following the numerous cell processes. However, significant unknown staining of the α7 antibody

was seen in the extracellular environment. A high cluster of staining was also observed around the

nucleus of the carcinoma cells, predominately in the permeabilised MDA-MB-231 samples (Figure

6B).

A diverse pattern of receptor subtype expression was observed for MDA-MB-231 cells

treated with the α9 antibody. Non-permeabilised cell samples showed specific staining of receptor

subtypes that mimic the flow of the cellular cytoskeleton (Figure 7A). A similar trend was seen in

the permeabilised cell samples (Figure 7B). Furthermore, the α9 antibody of permeabilised samples

stained the region of mitotic spindles within the nucleus of replicating cells. Slight extracellular and

high-density staining was seen with the α9 antibody; however, not as predominately as compared to

that seen for the α7 antibody.

Page 65: A Novel Mechanism for Nicotinic Acetylcholine Receptor

50

A

B

Figure 6: Immunocytochemistry images of non-permeabilised (A) and permeabilised (B) MDA-MB-231 cells showing

α7 nAChR subtype expression. Cells were treated with the primary α7 nAChR antibody with a fluorescent Alexa Fluor®

488 secondary antibody (green) and the Hoechst nuclear stain (blue). The treated cells were visualised using an upright

Nikon Eclipse Ni-E Motorised microscope with a 100x objective lense. DAPI and FITC images were overlayed using

Image J software, with the corresponding bright-field microscopy images shown.

10 μm

10 μm

Page 66: A Novel Mechanism for Nicotinic Acetylcholine Receptor

51

A

B

Figure 7: Immunocytochemistry images of non-permeabilised (A) and permeabilised (B) MDA-MB-231 cells showing

α9 nAChR subtype expression. Cells were treated with the primary α9 nAChR antibody with a fluorescent Alexa Fluor®

488 secondary antibody (green) and the Hoechst nuclear stain (blue). The treated cells were visualised using an upright

Nikon Eclipse Ni-E Motorised microscope with a 100x objective lense. DAPI and FITC images were overlayed using

Image J software, with the corresponding bright-field microscopy images shown.

10 μm

10 μm

Page 67: A Novel Mechanism for Nicotinic Acetylcholine Receptor

52

3.1.4 Discussion

Currently, there is only limited information in the literature describing the effects of nAChR

antagonists on cancer cell development. The present studies examined the effects of both

established and novel nAChR modulators on cell viability of a variety of carcinoma cell types using

a cell viability assay. In addition, the study aimed to determine the key nAChR subtypes involved in

mediating these responses. It was predicted that the selective nAChR antagonists, such as the

marine toxins, would display the most potent effects on viability of the carcinoma cell types; while

the α7 and α9 receptor subtypes were anticipated to be the most prominently expressed of the

nAChR subtypes.

Both ACh and NIC were used as positive controls to investigate whether the characteristic

increase in cell viability and expression of nAChRs could be replicated due to their strong agonistic

effects at nAChRs. Viability assay results demonstrated an ~20-30% increase in cell viability of

HT29 cells over 72 hours following treatment with 10 μM ACh or NIC. However, the change in

viability following treatment with ACh or NIC was not reflected in the remaining carcinoma cell

types investigated. Previous studies have demonstrated the ability of NIC to increase cancer cell

proliferation, with the most significant effects seen in lung cancer cell lines (not tested in the

present study) (Dasgupta et al., 2009; Tsurutani et al., 2005). The current literature has shown that

changes in lung cancer cell proliferation and cellular activity could be observed within 5 hours of

NIC treatment. However, it is possible that the extensive nAChR expression on lung cancer cells

differs from the expression patterns of the carcinoma cell types tested in this research. This theory is

supported by the Western Blot results, where lower levels of the key nAChR subtypes linked to cell

proliferation, α7 and α9, were shown compared to the highly responsive HT29 cells. The 10 μM

concentration of ACh and NIC was continued due to the possibility of the compounds having an

underlying effect on nAChR expression or changes in intracellular calcium concentration ([Ca2+]),

based on previous literature findings.

Page 68: A Novel Mechanism for Nicotinic Acetylcholine Receptor

53

Similar effects on HT29 cell proliferation have been previously shown with NIC promoting

cell proliferation, with a dose-dependent increase in DNA synthesis and adrenaline production

(Wong et al., 2007). The study suggested a link between the expression of the α7-nAChR subtype

and NIC-induced proliferation, with selective α7-nAChR blockage reversing the stimulatory effects

on cell proliferation. The present study is consistent with these findings, in that a significant

increase in HT29 cell proliferation was observed following nAChR stimulation that correlated with

a higher level of α7 expression in HT29 cells, compared to all other cell lines tested.

Significant decreases in cell viability were observed following MECA, PnTX, 5-HT and

ADI treatment. PnTX was the most potent of the compounds tested, causing as much as a ~70%

decrease in cell viability at a concentration 200-fold lower than that of the other nAChR

antagonists, with the most prominent effects seen in HT29 cells. PnTX A and G have been

previously shown to potently antagonise human α7 and α4β2 nAChRs (Araoz et al., 2011). Human

nAChR-expressing Xenopus oocytes showed significant blocking of α7 nAChR with nM

concentrations of PnTX A that was not reversible after 10-15 minutes washout. While PnTX A also

blocked the human α4β2 nAChR at higher concentrations, indicating that the toxin may be more

selective for the α7 subtype. PnTX G shows similar effects in oocyte-expressing nAChRs, but at

significantly higher concentrations than PnTX A (Araoz et al., 2011). The present results suggest

that PnTX is inducing carcinoma cell cytotoxicity through similar mechanisms, with both α7 and α4

nAChR subtypes being widely expressed amongst the cell lines tested. In addition, the

responsiveness of the cell lines was also correlated to the expression of the receptor subtypes, with

the highly expressive HT29 cells being the most sensitive to PnTX treatment. Conversely, the U87

cells had low nAChR expression and were the least sensitive to PnTX treatment.

The results of the non-selective nAChR antagonist, MECA, showed similar levels of

viability decrease compared to PnTX, but at a 200-fold higher concentration. It is reported in the

literature that MECA inhibits various nAChRs, including α4β2, α3β2, α3β4 and α7, with the least

potent effects observed at α7 nAChRs (Papke et al., 2001). Due to this relatively low selectivity for

Page 69: A Novel Mechanism for Nicotinic Acetylcholine Receptor

54

the α7 nAChR, the present study suggests that higher concentrations of MECA are required to

induce α7-related cytotoxicity, compared to the selective α7-antagonist, PnTX. A potent decrease

in cell viability was also observed following ADI treatment of the various carcinoma cell types,

similar to that seen for MECA. ADI acts via blocking heteromeric nAChR containing α3 or α4

subunits (eg α3β4, α4β2, α4β4) suggesting the cytotoxic effects may be mediated through the α4

expression on carcinoma cell types (Gentry et al., 2001). The comparative effects of MECA and

ADI, compared to PnTX, demonstrate the importance of both the α7 and α4 nAChR subtypes in

inducing cell cytotoxicity. However, the high concentrations of both MECA and ADI required to

induce a decrease in carcinoma cell viability question the mechanism behind the observed results.

Both MECA and ADI exhibit IC50 values <15 μM at nAChRs. The use of concentrations well

above the IC50 for both compounds in the present study may reduce the selectivity of these

compounds at the specific nAChRs, potentially inducing changes in cell viability through alternate

mechanisms. Additional studies using siRNA knockdown of critical nAChRs would be required to

confirm the mechanism behind the changes in carcinoma cell viability as a result of MECA and

ADI treatment.

Significant effects on cell viability were also observed following treatment with 5-HT, with

a significant decrease in cell viability across all cell lines tested at 72 hours post treatment. The

effects were consistent with those previously observed with 5-HT reuptake inhibitors (Gil-Ad et al.,

2008; Schuller, 2009). It is possible that these effects of 5-HT are mediated through the expression

of chimeric nACh–5-HT receptors that are expressed within the nervous system. Over-activation of

5-HT receptors has been shown to induce a significant downregulation of 5-HT receptors on the cell

surface of neurons (Gray et al., 2001). The treatment of 5-HT in the present study may induce

similar effects on chimeric nACh–5-HT receptors, reducing the expression of functional nACh–5-

HT receptors on the cell surface through receptor internalisation. The decrease in the nAChR

component may cause a fatal decrease in [Ca2+] levels, relating to the observed cancer cell

cytotoxicity at high doses of 5-HT. This theory would explain why previous literature has shown

Page 70: A Novel Mechanism for Nicotinic Acetylcholine Receptor

55

the ability of 5-HT reuptake inhibitors to eliminate the effects of nicotine and nicotine-derived

nitrosamines.

Interestingly, both MLA and GYM treatment had little effect on cancer cell viability with

only MDA-MB-231 and U373 cells showing a significant decrease in cell viability at 72 hours.

MLA exhibits a very high selectivity towards homomeric α7 nAChR, with the ability to bind to one

of five identical binding sites present on the α7 nAChR with an IC50 in the picomolar range (Palma

et al., 1996). MLA is thought to share the same binding sites to the α7 AChRs as the endogenous

agonist ACh, with the binding of a single MLA molecule to the receptor sufficient to prevent its

channel opening. However, unlike many antagonistic toxins that irreversibly bind the nAChR,

recovery of nAChR function is observed following MLA treatment (Palma et al., 1996). These

findings suggest that MLA may only have a significant effect on cellular function in the presence of

an agonist, such as ACh, compared to treatment with the agonist alone due to the competitive

binding nature of the compound. This idea was further investigated in Chapter 3 where [Ca2+] levels

of MDA-MB-231 cells were measured following MLA treatment. In addition, the reversible

binding of MLA may explain the lack of cytotoxicity observed following high concentrations of

MLA treatment, with 72 hours providing sufficient time for the cancer cells to recover and

proliferate.

The treatment of cancer cells with GYM was predicted to induce similar effects on cell

viability as PnTX due to their similarity in chemical structure and nAChR selectivity. GYM has

been previously shown to selectively block homomeric α7 nAChRs expressed in Xenopus oocytes

and heteropentameric α3β2 and α4β2 nAChRs during competition-binding assays (Kharrat et al.,

2008). Despite GYM displaying similar IC50 values of 0.33 and 15.5 nM at human α7 and α4β2

nAChRs, respectively, compared to PnTX, the effects on in vitro cell viability have been opposing

(Shin et al., 2005). Treatment of Neuro2a cells incubated with 10 μM GYM for 24 hours showed no

significant change in cell viability by MTT assay, while GYM at concentrations 10-fold higher have

also been relatively non-toxic on primary cultures of mouse cortical neurons (Alonso et al., 2011;

Page 71: A Novel Mechanism for Nicotinic Acetylcholine Receptor

56

Dragunow et al., 2005). The result of the present study show similar effects on cell viability, with

no significant changes in cell viability, with the exception of MDA-MB-231 cells, following

treatment with 10 μM GYM for 72 hours. The difference in effects between GYM and PnTX may

be due to the reversible binding nature of GYM, showing full recovery within 30 minutes of

washout in hemidiaphragm preparation, compared to the slow and incomplete recovery of tissue

following PnTX treatment (Hellyer, 2014; Hellyer et al., 2011).

The effects of the various compounds of cancer cell viability have been demonstrated. As

noted in Section 2.1.3 (MTT Assay), the assay method measures viable cell activity through the

enzymatic reduction of MTT. However, the method is unable to distinguish between a decrease in

cell activity due to cytotoxicity or an inhibition of cell proliferation. Therefore, the data from the

cell viability assay cannot clarify whether nAChR antagonism is having a cytotoxic effect on the

cells, or preventing the cells from replicating. Microscopic imaging of the treated cells indicate that

some compounds, such as PnTX, may be preventing cell proliferation due to a lack of cell death

observed. However, additional tests, such as flow cytometry analysis of the treated cells discussed

in the following sections, was required to further investigate the effects of these compounds on cell

viability.

To further investigate the presence and localisation of the α7 and α9 nAChR subunits on

cancer cells, immunocytochemistry (ICC) was performed on untreated MDA-MB-231 breast

carcinoma cells. The α9 nAChR is an integral subunit in the development of breast cancer due to its

upregulation following activation of the estrogen receptor, having implications in cancer growth

and metastasis (Lee et al., 2011). Furthermore, the importance of the α7 nAChR subunit has been

linked to several aspects of cancer development. ICC images from non-permeabilised cells treated

with the α7 antibody displayed significant staining on the surface of the cells, including the

numerous cell processes extending from the cell body. Significant levels of expression were also

observed with the α9 antibody, with a pattern that more closely resembles the cell cytoskeleton,

rather than small clusters of receptors on the cell surface. According to current knowledge, this is

Page 72: A Novel Mechanism for Nicotinic Acetylcholine Receptor

57

the first attempt to identify the expression of the α9 nAChR using an ICC method in human breast

cancer cells, making a comparison of the expression pattern with current literature difficult. Overall,

the ICC images exhibit the widespread expression of the key nAChR subtypes linked to

carcinogenesis on the cell surface of human cancer cells, with expression levels that are consistent

with those seen during the Western blot analysis for MDA-MB-231 cells. However, a lack of

reproducibility of the experiments due to difficulty with the method and optimisation of the

antibodies means further experiments would be required to confirm these conclusions.

The significant amount of non-specific binding of both antibodies within the extracellular

region and nucleus of the cancer cells questions the specificity of the antibodies to selectively detect

the α7 and α9 nAChR subunits. It is possible that extracellular staining may be abridged by changes

to the blocking and washing steps, reducing the non-specific expression and binding of the antibody

to cell fragments. Alternatively, the observed expression of the antibodies within the nucleus may

be due to known nuclear binding characteristic of the ER in breast carcinomas and link to α9

nAChR expression (Lee et al., 2011).

Having demonstrated an effect with various nAChR modulators on carcinoma cell viability,

the next step was to explore possible mechanisms of action.

3.2 Investigation of nAChR Antagonist Induced Cell Death

In the previous chapter it was demonstrated that selective nAChR antagonists induce cell

death in carcinoma cell lines. To investigate the mechanism(s) of nAChR modulator-induced

cytotoxicity (Aim II), Western blotting and flow cytometry were used to further characterise the

treated carcinoma cells.

3.2.1 Western Blot – α7 nAChR Levels following Treatment

Western blot analysis was used to investigate the effects of the various nAChR modulators on

α7 nAChR subtype expression (Figure 8). The α7 nAChR receptor was the main focus of the

Western blot study due to its widespread expression and involvement in cancer development.

Page 73: A Novel Mechanism for Nicotinic Acetylcholine Receptor

58

Treatment with the nAChR agonists, ACh 10 µM and NIC 10 µM, showed little effect on α7

expression compared to untreated cells. Of the four cell lines tested, only slight increases in α7

expression were seen in MDA-MB-231. MDA-MB-231 treatment with various nAChR antagonists,

including MECA, PnTX, GYM, MLA and ADI, produced a similar increase in α7 expression

ranging from 1.32 to 1.47-fold compared to untreated cells. No consistent increases in α7

expression were seen across all other cell lines or treatments. LN-18 cells were the least responsive

to treatment with nAChR modulators, with α7 expression decreasing following treatment with all

compounds, with the exception of MECA 1000 µM.

Page 74: A Novel Mechanism for Nicotinic Acetylcholine Receptor

59

Untr

eate

d M

AC

h 10

M

NIC

10

M

MEC

A 1

000

M

PnTX

5

M

GY

M 1

0 M

5-H

T 1

000

M

MLA

100

M

AD

I 25

0

0.0

0.5

1.0

1.5

2.0MDA-MB-231

HT29

PC3

LN18

a7 n

AC

hR

Den

sity

Rati

o

(Fold

Ch

an

ge)

Figure 8: Western blot analysis investigating the effects of various nAChR modulating compounds on the expression

level of the α7 nAChR subtype in MDA-MB-231, HT29, PC3 and LN18 cancer cell lines. Cells were either left untreated

or treated with ACh 10 µM, NIC 10 µM, MECA 1000 µM, PnTX 5 µM, GYM 10 µM, 5-HT 1000 µM, MLA 100 µM and

ADI 250 µM for 72 hours prior to harvesting and analysis. β-actin protein was used as a loading control to normalise

loading of proteins. The columns in the graph represent the fold change in density ratio between the treatment group

and the untreated control (n = 1).

Page 75: A Novel Mechanism for Nicotinic Acetylcholine Receptor

60

3.2.2 Flow Cytometry - Cell Cycle Analysis

Cell cycle analysis was conducted using the propidium iodide (PI) DNA stain to determine

the proportion of cells in each stage of the cell cycle. The percentage of cells in the sub-G1 stage of

the cell cycle was measured using Fluorescent Activated Cell Sorting (FACS) following treatment

with the various nAChR modulators, as displayed in Figure 9. An increase in sub-G1 percentage

represents an increase in fractional DNA content, characteristic of apoptotic cells. CDDP was

chosen as it is known to induce cell apoptosis through mitochondrial depolarisation,

phosphatidylserine translocation and caspase activation (Cregan et al., 2013). Treatment with ACh

10 µM, NIC 10 µM and MLA 100 µM had no effect on the percentage of cells in sub-G1. A

significant (P < 0.001) increase in DNA fragmentation was seen for all other treatments in all cell

lines compared to untreated controls, with the exception of GYM 10 µM and ADI 250 µM

treatment of MDA-MB-231 cells (no significant change in DNA fragmentation).

MECA 1000 µM and 5-HT 1000 µM produced consistent increases in the percentage of cells

in sub-G1 compared to untreated cells, at similar levels to the positive control (CDDP 20 µM)

(Figure 9). The significant change in cells in sub-G1 was consistent with the cell viability results for

each treatment, where significant decreases in cell death were seen over the same concentrations

(Figure 4A and Figure 4D). A significant ~2-fold increase in the sub-G1 cells was also seen

following PnTX 5 µM treatment of all cell lines (Figure 9). However, despite similar viability

losses between the treatments (Figure 4B), PnTX sub-G1 levels were substantially lower than those

seen following MECA (>10-fold) and 5-HT (>12-fold) treatment. Treatment with GYM 10 µM

produced a significant increase in apoptotic HT29 (Figure 9B) and PC3 (Figure 9C) cells, with

levels similar to PnTX treatment, which was not reflective of cell viability results (Figure 4C).

Page 76: A Novel Mechanism for Nicotinic Acetylcholine Receptor

61

A

Untr

eate

d M

AC

h 10

M

NIC

10

M

MEC

A 1

000

M

PnTX

5

M

GY

M 1

0 M

5-H

T 1

000

M

AD

I 25

0 M

MLA

100

M

CD

DP 2

0

0

1

2

3

4

5

20

30

40

50

***

***

***

***

Cell

s in

Su

b-G

1 (

%)

B

Untr

eate

d M

AC

h 10

M

NIC

10

M

MEC

A 1

000

M

PnTX

5

M

GY

M 1

0 M

5-H

T 1

000

M

AD

I 25

0 M

MLA

100

M

CD

DP 2

0

0

5

10

15

2030

40

50

60

70

80 ***

***

***

***

***

***

Cell

s in

Su

b-G

1 (

%)

C

Untr

eate

d M

AC

h 10

M

NIC

10

M

MEC

A 1

000

M

PnTX

5

M

GY

M 1

0 M

5-H

T 1

000

M

AD

I 25

0 M

MLA

100

M

CD

DP 2

0

0

2

4

6

8

1030

40

50

60

***

***

***

***

***

***

Cell

s in

Su

b-G

1 (

%)

Figure 9: Cell cycle analysis of antagonist treated MDA-MB-231 (A), HT29 (B) and PC3 (C) cell lines. Cells were

treated with ACh 10 µM, NIC 10 µM, MECA 1000 µM, PnTX 5 µM, GYM 10 μM, 5-HT 1000 µM, ADI 250 µM, and

MLA 100 µM for 72 hours prior to ethanol-fixation, propidium iodide staining and fluorescence-activated cell sorting

(FACS). Additionally, untreated cells were used as a negative control, while CDDP 20 µM treatment was used as a

positive control. The mean percentage of cells in the sub-G1 stage of the cell cycle has been calculated for each sample.

Values are displayed as the mean percent of cells in sub-G1, with error bars representing SEM (n = 5). A one-way

ANOVA with a Dunnett’s post-hoc test was used to determine significance compared to the untreated control (*** P <

0.001).

In addition to the Sub-G1 apoptotic group, the percentage of cells within each stage of the

cell cycle, including, G0/G1, S and G2/M, were determined to examine whether nAChR treatment

caused mitotic inhibition during any stage of cell division. The various stages of the cell cycle were

elucidated from the interactions of DNA with the fluorescent PI stain. Cells preparing for cell

division will contain increasing amounts of DNA; therefore, displaying a comparative increase in PI

fluorescence (Wlodkowic et al., 2011).

Page 77: A Novel Mechanism for Nicotinic Acetylcholine Receptor

62

No consistent change in the percentage of cells within each cell cycle region was seen

following GYM 10 μM and MLA 100 μM treatment of MDA-MB-231 (Figure 10), HT29 (Figure

11) and PC3 (Figure 12) cells. MECA 1000 μM treatment produced a significant decrease in the

percentage of cells in G0/G1, S and G2/M of the cell cycle, compared to untreated cells. The

observed decrease correlated with a large increase in sub-G1 cells following MECA treatment.

Similar effects were observed following 5-HT treatment of MDA-MB-231, HT29 and PC3 cells.

Changes to the number of cells within each stage of the cell cycle were variable following

PnTX 5 μm and ADI 250 μm treatment. PnTX treatment caused a significant increase (P < 0.001) in

MDA-MB-231 (Figure 10B) and PC3 (Figure 12B) cells in the G0/G1 region of the cell cycle,

suggesting PnTX may induce cell growth inhibition through G0/G1 arrest. This effect was not

observed in HT29 cells following PnTX treatment. Interestingly, PnTX treatment of HT29 cells did

produce a significant increase in cells within the S and G2/M regions, compared to untreated cells

(P < 0.001) (Figure 11B). Comparable results were seen following ADI 250 μm treatment of MDA-

MB-231 (Figure 10E), HT29 (Figure 11E) and PC3 (Figure 12E) cells.

Page 78: A Novel Mechanism for Nicotinic Acetylcholine Receptor

63

A Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

MECA 1000 M

***

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

B Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

PnTX 5 M

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

C Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

GYM 10 M

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

D Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

5-HT 1000 M

***

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

E

Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100 Untreated

ADI 250 M

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

F Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Unteated

MLA 100 M

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

Figure 10: Cell cycle analysis of treated MDA-MB-231 cells measured via flow cytometry. Cells were treated with

MECA 1000 µM (A), PnTX 5 µM (B), GYM 10 μM (C), 5-HT 1000 µM (D), ADI 250 µM (E), and MLA 100 µM (F) for

72 hours prior to ethanol-fixation, propidium iodide staining and fluorescence-activated cell sorting. Untreated cells

were used as a negative control. The mean percentage of cells in each stage of the cell cycle has been calculated for

each sample. The error bars represent SEM (n = 5). A two-way ANOVA with a Bonferroni post-hoc test was used to

determine significance compared to the untreated control (*** P < 0.001).

Page 79: A Novel Mechanism for Nicotinic Acetylcholine Receptor

64

A Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

MECA 1000 M******

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

B Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

PnTX 5 M

***

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

C Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

GYM 10 M

***

**

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

D Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

5-HT 1000 M

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

E

Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100 Untreated

ADI 250 M

***

******

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

F Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Unteated

MLA 100 M

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

Figure 11: Cell cycle analysis of treated HT29 cells measured via flow cytometry. Cells were treated with MECA 1000

µM (A), PnTX 5 µM (B), GYM 10 μM (C), 5-HT 1000 µM (D), ADI 250 µM (E), and MLA 100 µM (F) for 72 hours

prior to ethanol-fixation, propidium iodide staining and fluorescence-activated cell sorting. Untreated cells were used

as a negative control. The mean percentage of cells in each stage of the cell cycle has been calculated for each sample.

The error bars represent SEM (n = 5). A two-way ANOVA with a Bonferroni post-hoc test was used to determine

significance compared to the untreated control (*** P < 0.001).

Page 80: A Novel Mechanism for Nicotinic Acetylcholine Receptor

65

A Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

MECA 1000 M

***

*

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

B Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

PnTX 5 M

***

***

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

C Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

GYM 10 M

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

D Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Untreated

5-HT 1000 M

***

***

**

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

E

Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100 Untreated

ADI 250 M

***

**

***

***

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

F Sub-G1

G0/

G1 S

G2/

M

0

20

40

60

80

100Unteated

MLA 100 M

Pro

po

rtio

n o

f cell

s in

ea

ch

ph

ase

(%

)

Figure 12: Cell cycle analysis of treated PC3 cells measured via flow cytometry. Cells were treated with MECA 1000

µM (A), PnTX 5 µM (B), GYM 10 μM (C), 5-HT 1000 µM (D), ADI 250 µM (E), and MLA 100 µM (F) for 72 hours

prior to ethanol-fixation, propidium iodide staining and fluorescence-activated cell sorting. Untreated cells were used

as a negative control. The mean percentage of cells in each stage of the cell cycle has been calculated for each sample.

The error bars represent SEM (n = 5). A two-way ANOVA with a Bonferroni post-hoc test was used to determine

significance compared to the untreated control (* P < 0.05, ** P < 0.01, *** P < 0.001).

Page 81: A Novel Mechanism for Nicotinic Acetylcholine Receptor

66

3.2.3 Flow Cytometry - TMRE

To further investigate early apoptosis of the various cancer cell lines using flow cytometry,

the TMRE stain was used on cell samples treated with the most cytotoxic nAChR modulating

compounds (MECA, PnTX, 5-HT and ADI). TMRE is a positively-charged dye that accumulates in

active mitochondria due to the negatively charged nature of the mitochondrial membrane potential.

Therefore, a left shift in the FL2-H fluorescence peak compared to untreated cells indicates an

inability of the mitochondria to sequester the TMRE due to depolarisation of the mitochondrial

membrane potential, as observed during cell apoptosis..

The TMRE histograms for the individual treatments and cell types have been presented in

Figure 13A, with percentage of cells with low TMRE intensity graphed (Figure 13B). High levels

of TMRE staining were shown in the untreated cell samples, with consistent shifts in FL2-H peaks

following treatment with the CDDP 20 µM positive control (indicative of a loss in mitochondrial

membrane permeability during early apoptosis). Results following ADI 1000 µM for 72 hours

showed a complete shift in FL2-H, with diminutive staining of active mitochondria. MECA 1000

µM and 5-HT 1000 µM treatment showed shifts in FL2-H peak comparable to the CDDP positive

control. PnTX 5 µM induced only a minor shift in FL2-H while still showing considerable TMRE

staining of active mitochondria. The results seen following PnTX 5 µM treatment were

considerably less pronounced compared to all other treatments (Figure 13B), producing only a small

but significant change from the untreated control, despite the similar effects on cell viability loss

(Figure 4B).

Page 82: A Novel Mechanism for Nicotinic Acetylcholine Receptor

67

A Untreated MECA 1000 µM PnTX 5 µM 5-HT 1000 µM ADI 1000 µM CDDP 20 µM

MDA-

MB-

231

HT29

PC3

TMRE

B

Untr

eate

d M

MEC

A 1

000

M

PnTX

5

M

5-H

T 1

000

M

AD

I 10

00

M

CD

DP 2

0

0

10

20

30

40

50

60

70

80

90

100MDA-MB-231

HT29

PC3

All Combinations = ***

Cell

s w

ith

Low

TM

RE

In

ten

sity

(%

)

Figure 13: Evaluation of the mitochondrial transmembrane potential of MDA-MB-231, HT29 and PC3 cell lines

measured via flow cytometry to identify the early stages of cell apoptosis. Cells were treated with MECA 1000 µM,

PnTX 5 µM, 5-HT 1000 µM or ADI 1000 µM for 72 hours prior to TMRE staining and fluorescence-activated cell

sorting. Additionally, untreated cells were used as a negative control, while CDDP 20 µM treatment was used as a

positive control. Dot-plots represent FL2-H fluorescence versus cell number, where a shift in peak from left to right

signifies a collapse in mitochondrial transmembrane potential (A). The relative percentage of cells with low TMRE

intensity was graphed (B) with the error bars representing the mean ± SEM (n = 3). A one-way ANOVA with a

Bonferroni post hoc was used to determine significance between all treatments for each cell line (*** P < 0.001).

Page 83: A Novel Mechanism for Nicotinic Acetylcholine Receptor

68

3.2.4 Flow Cytometry - Annexin V/PI

The Annexin V/PI double staining was assessed by flow cytometry to distinguish between

early apoptotic and necrotic cell death following the treatment of cancer cells with the same nAChR

modulating compounds used in TMRE studies. Quadrants were determined based on the Annexin V

(FL1-H) and PI (FL2-H) staining of untreated cells, and kept constant across all treatments. Cells

within the left-bottom quadrant represent viable cells. A shift of cells into the right-bottom quadrant

represents phosphatidylserine translocation to the cell surface through an increase in Annexin V

staining, indicative of early apoptosis. As cells enter late apoptosis, PI may penetrate the

compromised cell wall and stain the DNA, as indicated by the presence of cells within the right-top

quadrant. Similarity, secondary necrotic cells demonstrate a similar increase in both PI and Annexin

V staining as the cell wall deteriorates during necrosis; while an increase in PI staining with no

change in Annexin V staining has not been clearly characterised in the literature (Wlodkowic et al.,

2011).

The results of Annexin V/PI double staining of treated MDA-MB-231 cells have been

presented in Figure 14. Low basal levels of apoptotic and necrotic cells were seen in the untreated

control cells (6.55%). Considerable increases in Annexin+/PI- (early apoptosis) and Annexin+/PI+

(late apoptosis) stained cells were seen across all treatments, including the CDDP positive control.

ADI 1000 µM and MECA 1000 µM produced the greatest increase in apoptotic cells, with 80.49%

and 71.53% of cells, respectively, within the positively stained Annexin V region. Lower levels of

Annexin+/PI- and Annexin+/PI+ stained cells were seen following PnTX 5 µM (41.85%) and 5-HT

1000 µM (31.13%) treatment, relative to ADI and MECA treatment. Nevertheless, the results

obtained following PnTX and 5-HT treatment were still well above the 7.57% seen within the

untreated sample. The same substantial increases seen in Annexin+/PI- and Annexin+/PI+ stained

cells for all treatments compared to untreated cells was not seen within the Annexin-/PI+ quadrant.

A maximum 3-fold increase in Annexin-/PI+ cells following MECA 1000 µM treatment was

observed compared to untreated cells.

Page 84: A Novel Mechanism for Nicotinic Acetylcholine Receptor

69

The percentage of MDA-MB-231 cells within the Annexin+/PI- bottom right quadrant has

been graphed in Figure 14G. All treatments, including the CDDP positive control, caused a

significant increase in Annexin+/PI- cells compared to control (P < 0.001). The significant increase

in Annexin+/PI- cells following treatment with MECA, PnTX, 5-HT and ADI indicates an increase

in the presence of early apoptotic cells.

Page 85: A Novel Mechanism for Nicotinic Acetylcholine Receptor

70

Untreated MECA 1000 µM PnTX 5 µM P

rop

idiu

m I

od

ide

A B C

Annexin V

5-HT 1000 µM ADI 1000 µM CDDP 20 µM

Pro

pid

ium

Io

did

e

D E F

Annexin V

G

Untr

eate

d M

MEC

A 1

000

M

PnTX

5

M

5-H

T 1

000

M

AD

I 10

00

M

CD

DP 2

0

0

5

10

15

2040

45

50

*** ***

***

***

***

Earl

y A

pop

toti

c C

ells

(%

)

Figure 14: Flow cytometric detection of apoptotic and necrotic cells by fluorescence-activated cell sorting of propidium

iodide and Annexin V labelled MDA-MB-231 cells. Cells were treated with MECA 1000 µM (B), PnTX 5 µM (C), 5-HT

1000 µM (D) or ADI 1000 µM (E) for 72 hours prior to staining and analysis. Untreated cells (A) were used as a

negative control, while CDDP 20 µM (F) treatment was used as a positive control. Dot-plots represent individual

stained cells. The left-bottom quadrant represents viable cells (Annexin-/PI-). The right-bottom quadrant represents

early apoptotic cells (Annexin+/PI-). Late apoptotic or secondary necrotic cells are represented by the right-top

quadrant (Annexin+/PI+). Early apoptotic (Annexin+/PI-) cells were graphed (G) with the error bars representing the

mean ± SEM (n = 3). A one-way ANOVA with a Dunnett’s post-hoc test was used to determine significance compared

to the untreated control (*** P < 0.001).

5.91%

62.96%

20.19%

10.94%

4.77%

14.75%

36.08%

44.41%

6.97%

40.05%

44.82%

8.15%

2.95%

89.48%

6.55%

1.02%

9.92%

18.55%

58.51%

13.02%

5.16%

53.00%

28.03%

13.82%

Page 86: A Novel Mechanism for Nicotinic Acetylcholine Receptor

71

Comparable results were seen within the HT29 treated cell samples, as shown in Figure 15,

with considerable increases in apoptotic and necrotic cells observed following the various

treatments, compared to the low basal levels of the control. ADI 1000 µM and MECA 1000 µM

treatment again produced the greatest increase in apoptotic cells, with 97.59% and 62.96% of cells

within the Annexin+ region for the respective treatments. Lower levels of apoptotic cells were seen

following PnTX 5 µM and 5-HT 1000 µM treatment. PnTX treatment produced a slight increase in

Annexin+ cells (24.96%) compared to the 7.81% for untreated cells; while 5-HT was comparable to

PnTX treatment with 28.74% of cells within the Annexin+ region. Significant increases in Annexin-

/PI+ stained cells were seen following MECA 1000 µM, 5-HT 1000 µM and CDDP 20 µM

treatment, with values of 22.57%, 19.93% and 14.26%, respectively, compared to untreated cells

(3.90%). Such large increases in Annexin-/PI+ stained cells was unique to HT29 cells, and not seen

in the other cell lines tested.

Significant increases in early apoptotic Annexin+/PI- cells were seen following treatment with

MECA (P < 0.001), PnTX (P < 0.001) and ADI (P < 0.05), compared to untreated cells (Figure

15G). No significant change in Annexin+/PI- cells was observed following 5-HT treatment,

compared to control. However, a large increase in secondary necrotic/late apoptotic cells following

5-HT treatment was observed, compared to untreated cells, despite the lack of early apoptotic cells.

Page 87: A Novel Mechanism for Nicotinic Acetylcholine Receptor

72

Untreated MECA 1000 µM PnTX 5 µM P

rop

idiu

m I

od

ide

A B C

Annexin V

5-HT 1000 µM ADI 1000 µM CDDP 20 µM

Pro

pid

ium

Io

did

e

D E F

Annexin V

G

Untr

eate

d M

MEC

A 1

000

M

PnTX

5

M

5-H

T 1

000

M

AD

I 10

00

M

CD

DP 2

0

0

5

10

15

20

*** ******

*

Earl

y A

pop

toti

c C

ell

s (%

)

Figure 15: Flow cytometric detection of apoptotic and necrotic cells by fluorescence-activated cell sorting of propidium

iodide and Annexin V labelled HT29 cells. Cells were treated with MECA 1000 µM (B), PnTX 5 µM (C), 5-HT 1000

µM (D) or ADI 1000 µM (E) for 72 hours prior to staining and analysis. Untreated cells (A) were used as a negative

control, while CDDP 20 µM (F) treatment was used as a positive control. Dot-plots represent individual stained cells.

The left-bottom quadrant represents viable cells (Annexin-/PI-). The right-bottom quadrant represents early apoptotic

cells (Annexin+/PI-). Late apoptotic or secondary necrotic cells are represented by the right-top quadrant

(Annexin+/PI+). Early apoptotic (Annexin+/PI-) cells were graphed (G) with the error bars representing the mean ±

SEM (n = 3). A one-way ANOVA with a Dunnett’s post-hoc test was used to determine significance compared to the

untreated control (* P < 0.05, *** P < 0.001).

19.93%

51.32%

24.33%

4.41%

1.92%

0.49%

89.16%

8.43%

14.26%

51.56%

24.76%

9.41%

3.90%

88.28%

4.24%

3.57%

22.57%

14.47%

53.11%

9.85%

7.72%

67.32%

15.02%

9.94%

Page 88: A Novel Mechanism for Nicotinic Acetylcholine Receptor

73

The cell death profiles of PC3 cells displayed a high level of apoptotic cells within the

untreated control, with 54.34% of cells within the Annexin+ region due to excessive confluency of

the cells at 72 hours (Figure 16). Increases in apoptotic cells were still however seen following

treatment with the various compounds. ADI 1000 µM and 5-HT 1000 µM produced the greatest cell

death, with respective Annexin+ values of 98.37% and 92.62 %. PnTX 5 µM treatment displayed

apoptotic results similar to the CDDP 20 µM control, with 85.57% of cells within the Annexin+

region. MECA 1000 µM treatment produced only a minor increase in Annexin+ apoptotic cells, with

a value of 62.83%, compared to untreated cells. Similar levels of Annexin-/PI+ stained cells were

observed across all treatments, contrary to results seen for the other cell lines tested.

Treatment with MECA and PnTX were the only two treatments to produce significant

increases in early apoptotic cells, compared to untreated cells (Figure 16G). However, further

analysis of the dot-plots show all treatments produced a consistent reduction in viable cells, with

large populations of cells within the secondary necrotic/late apoptotic region.

Page 89: A Novel Mechanism for Nicotinic Acetylcholine Receptor

74

Untreated MECA 1000 µM PnTX 5 µM P

rop

idiu

m I

od

ide

A B C

Annexin V

5-HT 1000 µM ADI 1000 µM CDDP 20 µM

Pro

pid

ium

Io

did

e

D E F

Annexin V

G

Untr

eate

d M

MEC

A 1

000

M

PnTX

5

M

5-H

T 1

000

M

AD

I 10

00

M

CD

DP 2

0

0

5

10

15

20

***

*

Earl

y A

pop

toti

c C

ell

s (%

)

Figure 16: Flow cytometric detection of apoptotic and necrotic cells by fluorescence-activated cell sorting of propidium

iodide and Annexin V labelled PC3 cells. Cells were treated with MECA 1000 µM (B), PnTX 5 µM (C), 5-HT 1000 µM

(D) or ADI 1000 µM (E) for 72 hours prior to staining and analysis. Untreated cells (A) were used as a negative

control, while CDDP 20 µM (F) treatment was used as a positive control. Dot-plots represent individual stained cells.

The left-bottom quadrant represents viable cells (Annexin-/PI-). The right-bottom quadrant represents early apoptotic

cells (Annexin+/PI-). Late apoptotic or secondary necrotic cells are represented by the right-top quadrant

(Annexin+/PI+). Early apoptotic (Annexin+/PI-) cells were graphed (G) with the error bars representing the mean ±

SEM (n = 3). A one-way ANOVA with a Dunnett’s post-hoc test was used to determine significance compared to the

untreated control (* P < 0.05, *** P < 0.001).

3.41%

3.98%

88.62%

4.00%

1.23%

0.40%

96.40%

1.97%

5.04%

4.68%

86.51%

3.77%

8.13%

37.53%

48.60%

5.74%

5.04%

32.13%

51.57%

11.26%

3.50%

10.93%

71.56%

14.01%

Page 90: A Novel Mechanism for Nicotinic Acetylcholine Receptor

75

3.2.5 Discussion

Western blot analyses and flow cytometry studies were used to examine the role of the most

potent nAChR modulators in inducing carcinoma cell death. It was initially hypothesised that the

selective nAChR antagonists would display the greatest cytotoxic effects on the various carcinoma

cell lines. The cell viability results previously discussed in Section 3.1 Effects of nAChR

Antagonists on Cell Viability demonstrated that the selective α7/α4β2 nAChR antagonist, PnTX,

displayed the most potent cytotoxic effects, while other selective antagonists, such as GYM and

MLA, were less toxic. The results of the cell viability studies were predicted to translate to the flow

cytometry results. In other words, the compounds with the greatest cytotoxic effects were expected

to have a significant impact on nAChR subtype expression, while inducing the greatest level of cell

apoptosis.

The Western blot analysis focused on the α7 nAChR subtype because of its widespread

expression, as observed in the previous studies. MDA-MB-231 cells showed a small increase in α7

expression following treatment with the various nAChR modulators, with the exception of 5-HT

treatment, while varying results were observed for all other cell lines. The literature reports that the

biological functions of the α7 nAChRs are increased following NIC exposure (Schuller, 2009).

Plummer and colleagues demonstrated the ability of the α7 nAChR to be significantly upregulated

in NCL-H69 human lung carcinoma cells following treatment with 100 ρM NNK. Changes in the

expression of the α7 nAChR subtype could be seen as early as 60 minutes following treatment;

however, the increase in expression was not observed at 24 hours post treatment (Plummer 3rd et

al., 2005). The present results following ACh or NIC treatment demonstrate little change in α7

expression across the cell types tested. A lack of a consistent change in α7 expression may be

attributed to the 72 hour treatment time, with changes in expression potentially occuring earlier in

the treatment period, as suggested by the literature. Similarly, no consistent changes in α7

expression were observed following treatment with the various nAChR antagonists. It was predicted

that the nAChR antagonists would produce a reduction in the α7 nAChR expression; however, the

Page 91: A Novel Mechanism for Nicotinic Acetylcholine Receptor

76

observed results showed inconsistent changes between cell lines with no clear trends in α7

expression following treatment.

Tests to investigate the mechanism of cell death for the most cytotoxic nAChR modulators

were conducted on the MDA-MB-231, HT29 and PC3 cell lines. Flow cytometry cell cycle analysis

of treated cells following PI staining showed a significant increase in fractional DNA content

following MECA, PnTX, GYM (excluding MDA-MB-231 cells), 5-HT and ADI treatment,

compared to untreated cells. Interestingly, PnTX treatment induced small, but significant increases

in the percentage of cells in the sub-G1 fraction, indicating lower levels of apoptosis compared to

the other treatments investigated. An analysis of the complete cell cycle of MDA-MB-231 and PC3

cells indicated that PnTX treatment delayed the progression of cells from the G0/G1 phase into the

S phase. Similar results were observed following TMRE staining, with all treatments investigated

inducing significant mitochondrial membrane permeability, characteristic of early apoptosis.

Annexin V/PI double stain demonstrated significant increases in the presence of early apoptotic

cells and late apoptotic/necrotic cells following a 72 hour treatment period. PnTX showed a

consistently high level of early apoptotic cells compared to the other treatments tested, however

displayed lower levels of late apoptotic/necrotic cells, indicating a delayed onset of cell apoptosis

compared to the other nAChR antagonists.

The PnTX induced cell-cycle arrest in the G0/G1 phase may result from changes in [Ca2+]

through antagonism of α7 and α4β2 nAChRs. Transient rises in [Ca2+] are integral in the process of

cell division. Rises in [Ca2+], both from intracellular pools and the extracellular environment, occur

during the early stages of the cell cycle G1 phase. Although cell division requires a vast array of

physiological responses, Ca2+ acts as a ubiquitous second messenger, initiating many of the

processes involved during the cell cycle (Kahl et al., 2003). Current literature has demonstrated that

Ca2+ channel blockers, such as amlodipine, induce G1 phase accumulation by alleviating Ca2+ entry

into the cell. Amlodipine (30 μM) treatment of A431 human epidermoid carcinoma cells

demonstrated an ~20% increase in cells within the G0-G1 phase of the cell cycle after 48 hours of

Page 92: A Novel Mechanism for Nicotinic Acetylcholine Receptor

77

treatment, compared to control cells. The reduction in [Ca2+] significantly decreased the expression

of cyclin D1 and CDK4, specific proteins involved in cell progression through the G1 phase

(Yoshida et al., 2007). The present results suggest a possible link between PnTX treatment and

reductions in [Ca2+], resulting in G0/G1 arrest and an inhibition of cell proliferation. Changes in

[Ca2+] would explain why relatively lower levels of sub-G1 cells and TMRE staining were observed

following PnTX treatment, despite possessing similar reductions in cell viability compared to other

treatments investigated. To support this theory, studies looking at the changes in [Ca2+] following

PnTX treatment were undertaken, as discussed in the next chapter.

The majority of treatments investigated induced significant levels of apoptosis in the treated

cells following 72 hours of treatment, demonstrating increases in fragmented DNA, mitochondrial

membrane permeability and phosphatidylserine translocation, indicative of early apoptosis. The

present results are consistent with those previously seen for nAChR antagonists. APS8, a non-

competitive and selective α7 nAChR antagonist, has been investigated due to its ability to inhibit

NSCLC cells growth and activate apoptotic pathways. APS8 treatment of A549 and SKMES-1 cells

caused significant mitochondrial depolarisation, leading to the release and activation of several

apoptotic factors involved in the intrinsic pathway (Zovko et al., 2013). Increased expression of

cytochrome C and SMAC, which are released following mitochondrial permeabilisation, were

observed following APS8 exposure, inducing the activation of caspase-9, a common inducer of the

intrinsic pathway (Zovko et al., 2013). PnTX, the most selective and cytotoxic α7 nAChR

antagonists investigated, induced similar mitochondrial permeabilisation in MDA-MB-231, HT29

and PC3 carcinoma cell lines. In addition, significant levels of Annexin+/PI-, indicative of early

apoptosis, suggest PnTX may induce apoptosis in a similar manner to that seen following APS8

treatment.

The cell death profile of PnTX differed from the other nAChR antagonists investigated,

despite showing similar effects in the earlier cell viability studies. PnTX treated cells displayed a

lower proportion of cells in the sub-G1 phase, lower TMRE staining and higher levels of early

Page 93: A Novel Mechanism for Nicotinic Acetylcholine Receptor

78

apoptotic cells, during the flow cytometric studies. The changes in the cell cycle of the investigated

cells suggest a possible mechanism by which PnTX treatment could reduce cancer cell proliferation.

Alternatively, the shift in TMRE staining suggests possible induction of the intrinsic apoptotic

pathway, contributing to the early apoptotic effects of PnTX. The following chapter looked at

investigating the inhibition of various downstream apoptotic messengers to further deduce the

mechanism behind the cytotoxic effects of PnTX.

3.3 Inhibition of nAChR Antagonist Induced Cell Death

In the previous studies it was shown that nAChR treatment induces potent apoptotic effects in

carcinoma cell lines. To investigate this phenomenon further, apoptotic inhibitors were used to

determine whether inhibiting common apoptotic pathways would reduce the cytotoxic effects of the

nAChR modulators (Aim III).

The apoptotic pathway, as detailed in Section 1.2.4 Anti-Apoptotic Effects of nAChRs in

Cancer Survival, is regulated by several key intermediates. Following activation of the extrinsic

death receptors, or the intrinsic response to cellular stress, the apoptotic pathway is initiated. One

key pathway is the activation of the Ca2+-dependent proteases, calpains, in response to excessive

intracellular Ca2+ levels causing cleavage of the Na+/Ca2+ mitochondrial exchanger leading to

mitochondrial Ca2+ accumulation. This induces changes in the inner mitochondrial membrane

potential, which stimulates the opening of the mitochondrial permeability transition pore (Elmore,

2007; Smith et al., 2012). Cytochrome C and small mitochondria-derived activator of caspases

proteins are released through the pore, activating several downstream mediators, such as cysteine-

dependent aspartic proteases (also known as caspases). The downstream activation of caspase-3

stimulates the final stages of apoptosis, inducing cytoskeletal reorganisation and disintegration of

the cell into apoptotic bodies (Elmore, 2007; Smith et al., 2012). Several of these integral steps

were targeted by the following studies to better understand the cytotoxic effects of nAChR

modulators on carcinoma cells.

Page 94: A Novel Mechanism for Nicotinic Acetylcholine Receptor

79

3.3.1 Cell Death Inhibitors

To investigate the possible apoptotic pathways affected by the treatment with the most potent

nAChR modulator tested, PnTX, various cell death inhibitors were used. MDA-MB-231, HT29,

PC3 and LN18 cells were treated with cell death inhibitors alone and in combination with PnTX 5

µM to examine any possible decrease in cell death caused by the PnTX treatment.

Cyclosporin A (CsA), a potent inhibitor of the opening of the mitochondrial permeability

transition pore during apoptosis through binding to the cyclophilin D protein, was the first inhibitor

investigated (Basso et al., 2008) (Figure 17). Other inhibitors tested included ALLN (Figure 18),

BAPTA (Figure 19) and zVAD (Figure 20). ALLN reversibly blocks the Ca2+-dependent neutral

cysteine protease calpain I, commonly involved in various cellular processes, including

proliferation, apoptosis and differentiation (Chen et al., 2002). BAPTA, a known Ca2+ chelator,

inhibits apoptosis through decreasing [Ca2+] levels critical in the activation of apoptotic caspases

(Zhao et al., 2005). Furthermore, zVAD inhibits the proteolytic activity of apoptotic enzymes

through irreversibly binding to the catalytic site of caspase proteases (Gamen et al., 2000).

The combination treatment of CsA + PnTX 5 µM did no improve the viability of any of the

four cell lines tested, compared to PnTX 5 µM treatment alone (Figure 17). Conversely, CsA 10

µM + PnTX 5 µM treatment produced a significant (P < 0.01) 38% decrease in cell viability of

MDA-MB-231, compared to PnTX 5 µM treatment alone (Figure 17A).

Similar results were seen across other cell death inhibitors tested with decreases in cell

viability seen for MDA-MB-231 cells following ALLN 5 µM + PnTX 5 µM (P < 0.001), BAPTA 1

µM + PnTX 5 µM (not significant) and zVAD 30 µM + PnTX 5 µM (P < 0.01), compared to PnTX

treatment alone. No significant changes in MDA-MB-231 cell viability were seen following CsA,

BAPTA or zVAD treatment alone, compared to untreated cells, with only ALLN treatment alone

showing a significant decrease in cell viability (P < 0.05).

Page 95: A Novel Mechanism for Nicotinic Acetylcholine Receptor

80

A U

ntrea

ted

DM

SO M

PnTX

5

M

CsA

10

CsA

+ P

nTX

0

20

40

60

80

100

120

**

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

BU

ntrea

ted

DM

SO M

PnTX

5

M

CsA

1

CsA

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

C U

ntrea

ted

DM

SO M

PnTX

5

M

CsA

1

CsA

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

D U

ntrea

ted

DM

SO M

PnTX

5

M

CsA

1

CsA

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 17: Examination of the effect of the cell death inhibitor Cyclosporin A (CsA) on PnTX induced cytotoxicity in

MDA-MB-231 (A), HT29 (B), PC3 (C) or LN18 (D) cell lines, following 72 hours of treatment. Cells were treated with

either PnTX 5 µM, CsA 1 μM or 10 µM, or a combination of the two following a 24 hour seeding period. Untreated

cells were used as a negative control, while DMSO was included as a vehicle control. Cell viability was established via

the MTT assay. Values are displayed as the mean viability (as a percentage) of the viability of control cells, with error

bars representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc test was used to determine significance

between treatment groups. ** represents P < 0.01 between the PnTX and the CsA + PnTX treatment groups.

Page 96: A Novel Mechanism for Nicotinic Acetylcholine Receptor

81

A

Untr

eate

d

DM

SO M

PnTX

5

M

ALLN

5

ALLN

+ P

nTX

0

20

40

60

80

100

120

***

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

B

Untr

eate

d

DM

SO M

PnTX

5

M

ALLN

1

ALLN

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

C

Untr

eate

d

DM

SO M

PnTX

5

M

ALLN

5

ALLN

+ P

nTX

0

20

40

60

80

100

120

*

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

D

Untr

eate

d

DM

SO M

PnTX

5

M

ALLN

1

ALLN

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 18: Examination of the effect of the cell death inhibitor ALLN on PnTX induced cytotoxicity in MDA-MB-231

(A), HT29 (B), PC3 (C) or LN18 (D) cell lines, following 72 hours of treatment. Cells were treated with either PnTX 5

µM, ALLN 1 μM or 5 µM, or a combination of the two following a 24 hour seeding period. Untreated cells were used as

a negative control, while DMSO was included as a vehicle control. Cell viability was then established via the MTT

assay. Values are displayed as the mean viability (as a percentage) of the viability of control cells, with error bars

representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc test was used to determine significance

between treatment groups. * represents P < 0.05 while *** represents P < 0.001 between the PnTX and the ALLN +

PnTX treatment groups.

Page 97: A Novel Mechanism for Nicotinic Acetylcholine Receptor

82

A

Untr

eate

d

DM

SO M

PnTX

5

M

BA

PTA

1

BA

PTA

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

B

Untr

eate

d

DM

SO M

PnTX

5

M

BA

PTA

1

BA

PTA

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

C

Untr

eate

d

DM

SO M

PnTX

5

M

BA

PTA

10

BA

PTA

+ P

nTX

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

D

Untr

eate

d

DM

SO M

PnTX

5

M

BA

PTA

0.5

BA

PTA

+ P

nTX

0

20

40

60

80

100

120

140

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 19: Examination of the effect of the cell death inhibitor BAPTA on PnTX induced cytotoxicity in MDA-MB-231

(A), HT29 (B), PC3 (C) or LN18 (D) cell lines, following 72 hours of treatment. Cells were treated with either PnTX 5

µM, BAPTA 1 μM or 5 µM, or a combination of the two following a 24 hour seeding period. Untreated cells were used

as a negative control, while DMSO was included as a vehicle control. Cell viability was then established via the MTT

assay. Values are displayed as the mean viability (as a percentage) of the viability of control cells, with error bars

representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc test was used to determine significance

between treatment groups. No significant difference was seen between the PnTX and the BAPTA + PnTX treatment

groups.

Page 98: A Novel Mechanism for Nicotinic Acetylcholine Receptor

83

A

Untr

eate

d

DM

SO M

CD

DP 1

0 M

PnTX

5

M

zVA

D 3

0

zVA

D +

CD

DP

zVA

D +

PnT

X

0

20

40

60

80

100

120

**

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

B

Untr

eate

d

DM

SO M

CD

DP 1

0 M

PnTX

5

M

zVA

D 2

0

zVA

D +

CD

DP

zVA

D +

PnT

X

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

C

Untr

eate

d

DM

SO M

CD

DP 1

0 M

PnTX

5

M

zVA

D 3

0

zVA

D +

CD

DP

zVA

D +

PnT

X

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

D

Untr

eate

d

DM

SO M

CD

DP 1

0 M

PnTX

5

M

zVA

D 1

0

zVA

D +

CD

DP

zVA

D +

PnT

X

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 20: Examination of the effect of the cell death inhibitor zVAD on PnTX induced cytotoxicity in MDA-MB-231

(A), HT29 (B), PC3 (C) or LN18 (D) cell lines, following 72 hours of treatment. Cells were treated with either PnTX 5

µM, zVAD 10, 20 or 30 µM, or a combination of the two following a 24 hour seeding period. Untreated cells were used

as a negative control, DMSO was included as a vehicle control, while CDDP and CDDP + zVAD was used as a

positive control. Cell viability was then established via the MTT assay. Values are displayed as the mean viability (as a

percentage) of the viability of control cells, with error bars representing SEM (n = 3). A one-way ANOVA with a

Bonferroni post-hoc test was used to determine significance between treatment groups. ** represents P < 0.01 between

the PnTX and the zVAD + PnTX treatment groups.

3.3.2 Intracellular Calcium

Intracellular calcium ([Ca2+]) is involved in various cell signalling pathways, including the

cell apoptotic pathway. Based on the early results linking nAChR modulation and Ca2+ influx

within a cell (refer to the discussion in Section 3.2 Investigation of nAChR Antagonist Induced Cell

Death), the [Ca2+] levels of MDA-MB-231, HT29 and PC3 cells were measured following

Page 99: A Novel Mechanism for Nicotinic Acetylcholine Receptor

84

treatment with the various nAChR modulators. The MDA-MB-231, HT29 and PC3 cell lines were

chosen based on their range of nAChR expression observed during Western blot studies (Figure 5).

Cells were incubated with 4 µM of the intracellular fluorescent Ca2+ indicator, Fura-2 AM, prior to

treatment. The [Ca2+] levels were then measured over 360 minutes post treatment, with the

fluorescence intensity ratio (340 nm/380 nm) displayed in Figure 21.

Characteristic results were seen following treatment with the known nAChR agonist, ACh 10

μM, in all cells lines tested. An instant increase in [Ca2+] was seen after 1-2 minutes of ACh

treatment, compared to untreated control cells. The increase in [Ca2+] was sustained over the first 15

minutes of recording, eventually returning to baseline after ~60 minutes in both HT29 and PC3 cell

lines. MDA-MB-231 [Ca2+] levels were less defined following ACh treatment. An initial increase in

[Ca2+] was seen, however results varied over the 360 minutes of recording with [Ca2+] levels

remaining above baseline.

No obvious change in [Ca2+] levels was seen in HT29 and PC3 cell lines following GYM 10

μM and MLA 100 μM treatment (Figure 21). The level of [Ca2+] for both treatments remained close

to baseline for the 300 minutes of recordings. Slight variations in [Ca2+] levels of MDA-MB-231

cells were seen following GYM and MLA treatment. However, [Ca2+] changes were inconsistent

and not characteristic of the trends seen in the other cell lines tested. The lack of [Ca2+] variation

following GYM and MLA treatment is consistent with the small changes in cell viability shown for

both treatments in Figure 4C and Figure 4F.

Despite the observed decrease in cell viability following treatment with the nAChR

antagonists MECA and ADI (Figure 4A and Figure 4E), treatment with each compound produced

an increase in [Ca2+] levels. MECA 1000 μM treatment of HT29 cells produced a noticeable

increase in [Ca2+], compared to untreated cells. The increase in [Ca2+] was delayed compared to the

response seen following ACh 10 μM treatment, and returned to baseline after ~60 minutes. A

similar, however more variable effect was seen in MDA-MB-231 cells, while the increase in [Ca2+]

levels following MECA treatment could not be repeated in PC3 cells. An obvious increase in [Ca2+]

Page 100: A Novel Mechanism for Nicotinic Acetylcholine Receptor

85

levels was also observed following treatment of MDA-MB-231, HT29 and PC3 cells with ADI

1000 μM. The increase in [Ca2+] was similar across all three cell lines, lasting for ~15 minutes

following treatment with ADI.

Treatment with PnTX 5 μM or 5-HT 1000 μM produced the only consistent decreases in

[Ca2+] levels among the compounds tested. 5-HT 1000 μM treatment in HT29 and PC3 cells

produced a noticeable decrease in [Ca2+] levels, which returned to baseline ~120 minutes following

treatment. A greater and more sustained decrease in [Ca2+] levels was observed following PnTX 5

μM treatment across all cell lines tested. [Ca2+] levels for cell lines treated with PnTX did not return

to baseline levels, remaining consistently below the untreated cell group.

MDA-MB-231 HT29 PC3

ACh 10 µM

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedACh 10 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

2.6

60 120 180 240 300 360

ACh 10 M

Untreated

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

ACh 10 M

Untreated

Time (Min)

340/3

80 R

ati

o

MECA 1000 µM

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedMECA 1000 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedMECA 1000 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedMECA 1000 M

Time (Min)

340/3

80 R

ati

o

PnTX 5 µM

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedPnTX 5 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedPnTX 5 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedPnTX 5 M

Time (Min)

340/3

80 R

ati

o

Page 101: A Novel Mechanism for Nicotinic Acetylcholine Receptor

86

MDA-MB-231 HT29 PC3

GYM 10 µM

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedGYM 10 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedGYM 10 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedGYM 10 M

Time (Min)

340/3

80 R

ati

o

5-HT 1000 µM

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

Untreated5-HT 1000 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

Untreated5-HT 1000 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

Untreated5-HT 1000 M

Time (Min)

340/3

80 R

ati

o

ADI 1000 µM

0 5 10 15

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

60 120 180 240 300 360

UntreatedADI 1000 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

1.0

1.5

2.0

2.5

3.0

3.5

4.0

60 120 180 240 300 360

UntreatedADI 1000 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedADI 1000 M

Time (Min)

340/3

80 R

ati

o

MLA 100 µM

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedMLA 100 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedMLA 100 M

Time (Min)

340/3

80 R

ati

o

0 5 10 15

0.8

1.0

1.2

1.4

1.6

1.8

2.0

2.2

2.4

60 120 180 240 300 360

UntreatedMLA 100 M

Time (Min)

340/3

80 R

ati

o

Figure 21: Ca2+ accumulation through Fura-2 AM detection within MDA-MB-231, HT29 and PC3 cells following ACh

10 µM, MECA 1000 µM, PnTX 5 µM, GYM 10 µM, 5-HT 1000 µM, ADI 1000 µM or MLA 100 µM treatment.

Untreated cells were used as the negative control Cells were allowed to adhere overnight before being treated with 100

µL/well of 4 µM of the Ca2+ indicator Fura-2 AM. Cells were then treated with one of the corresponding nAChR

modulators. Fura-2 fluorescence was measured using a fluorescent plate reader at an excitation of 340 nm (λ1) and

380 nm (λ2), with an emission of 510 nm, over 300 minutes. The fluorescence intensity ratio (R = Fλ1/Fλ2) was

calculated for each time point and graphed with error bars representing SEM (n = 3).

Page 102: A Novel Mechanism for Nicotinic Acetylcholine Receptor

87

3.3.3 Discussion

The effects of various nAChR antagonists on cancer cell viability and their capacity to induce

cell apoptosis has been discussed in the previous results sections. The current section further

investigated the mechanism of nAChR-induced cancer cell death. PnTX was chosen as the lead

compound based on its high affinity to antagonise nAChRs and interesting cytotoxic profile

compared to the other antagonists investigated. Various inhibitors of common apoptotic effectors

were employed in an attempt to reduce the cytotoxic effects of PnTX and further deduce the

mechanism of cell death. The results showed no significant inhibition of cancer cell apoptosis

following co-treatment of PnTX with CsA, ALLN, BAPTA and zVAD. Interestingly, the majority

of apoptotic inhibitors induced a further decrease in cell viability when combine with PnTX, while

having little effect on cell viability alone.

The flow cytometry results have suggested an increase in mitochondrial permeabilisation

following PnTX treatment (refer to Section 3.2.3 Flow Cytometry - TMRE). Changes in

mitcochondrial permeabilisation occur during activation of the intrinsic apoptotic pathway,

resulting in release of cytochrome C and a consequent downstream activation of the caspases. CsA

acts via inhibiting the cyclophilin D protein, preventing opening of the mitochondrial permeability

transition pore during apoptosis. It was hypothesised that co-treatment with CsA might alleviate

PnTX induced cytotoxicity through inhibition of the intrinsic apoptotic pathway. Interestingly

however, CsA treatment with PnTX induced a significant decrease in cell viability, compared to

PnTX treatment alone in MDA-MB-231 cells, while no change was observed in all other cell lines

tested. CsA possesses a secondary action through inhibiting phosphatase 2B activity, which may

explain why a further decrease in cell viability was observed (Marszalec et al., 2005).

Supplementary studies to investigate the effects of combination treatment with CsA and the broad-

spectrum nAChR inhibitor, MECA, in MDA-MB-231 cells were investigated in the following

section (Combination T).

Page 103: A Novel Mechanism for Nicotinic Acetylcholine Receptor

88

Similarly, combination treatment of the caspase inhibitor zVAD and PnTX induced a decrease

in cell viability compared to PnTX treatment alone. Caspases exhibit proteolytic activity, which

when activated play an almost irreversible role in apoptosis. The activation of effector caspases

(caspase-3, -6 and -7) facilitates the execution pathway of apoptotic cell death, resulting in

breakdown of nuclear material and the formation of apoptotic bodies (Elmore, 2007). The present

results are inconsistent with those seen previously following inhibition of the nAChRs. APS8, a

known antagonist of α7 nAChRs, induced significant activation of caspase-9 and caspase-3

following treatment of A549 and SKMES-1 cancer cells (Zovko et al., 2013). Interestingly, NIC has

also been shown to inhibit the activation of caspase-3 following the treatment of Tca8113 oral

cancer cells with the known caspase inducer, cisplatin, suggesting a link between nAChR regulation

and caspase activation (Xu et al., 2007). A lack of restoration of cell viability following caspase

inhibition in the presence of PnTX suggests PnTX-induced cancer cell cytotoxicity acts via a

caspase-independent mechanism. The mechanisms mediating caspase-independent apoptosis are not

well characterised; however, several key cellular events are capable of occurring in the absence of

caspase activity. Phosphatidylserine externalisation (as demonstrated during flow cytometry studies

with Annexin V in Section 3.2.4) can occur independently of caspase activation through changes in

intracellular Ca2+ activity, altering the activity of the key mediators of phosphatidylserine

externalisation (Cummings et al., 2004). Furthermore, nuclear fragmentation (evident from flow

cytometry studies with PI in Section 3.2.2) as a result of the activation of DNAses may be

controlled through non-caspase-related pathways involving Ca2+-dependent DNAse proteins

(Nitahara et al., 1998). These findings again suggest a role of intracellular Ca2+ alterations in PnTX

cancer cytotoxicity.

The detrimental effects of PnTX on MDA-MB-231 and PC3 cell viability increased following

treatment with the Ca2+ activated calpain inhibitor, ALLN, while having no significant effect on

HT29 and LN18 viability compared to PnTX treatment alone. Calpains play a role in the execution

process of apoptosis following activation of the enzymes due to an increase in intracellular Ca2+.

Page 104: A Novel Mechanism for Nicotinic Acetylcholine Receptor

89

Once activated, calpains degrade the membrane, cytoplasmic and nuclear structures during cell

apoptosis (Momeni, 2011). Changes in mitochondrial permeability and release from intracellular

stores during apoptosis induce an influx in the intracellular concentration of Ca2+, activating such

proteins as calpains. The present results suggest that the cytotoxic effects of PnTX may result from

decreases in intracellular Ca2+ due to the antagonistic effects at α7 and α4β2 nAChRs. This theory is

further consolidated through the observed effects of BAPTA on PnTX cytotoxicity. The Ca2+

chelator, BAPTA, has been utilised in apoptotic studies as a possible inhibitor by reducing the

intracellular influx in Ca2+ (Zhao et al., 2005). The present results demonstrated that, in some cases,

BAPTA potentiated the cytotoxic effects of PnTX on cancer cells.

Due to the results obtained in the apoptosis inhibitory studies, it was hypothesised that the

cytotoxic effects of the various antagonists, specifically PnTX, were due to changes in [Ca2+] influx

into the cell, inducing carcinoma cell apoptosis. PnTX treatment demonstrated an obvious and

sustained decrease in [Ca2+] levels of various carcinoma cells over the 360 minutes of testing

(Figure 21). Similar effects were not observed following treatment with the other antagonists, with

only 5-HT treatment producing a transient decrease in [Ca2+].

PnTX has been identified as a potent antagonist of α7 nAChRs. Recent literature analysing

the functional capacity of PnTX A and G revealed high affinity binding and potent antagonism for

the α7 subtype during binding and voltage-clamp electrophysiology studies (Bourne et al., 2015).

Activation of the α7 nAChRs causes large influxes of intracellular Ca2+, capable of inducing various

intracellular signalling pathways, without the additional coupling to voltage-gated calcium channels

(Garduño et al., 2012). This characteristic influx in [Ca2+] is responsible for many of the pro-

carcinogenic effects of nAChRs. The effects of PnTX on [Ca2+] were first identified during studies

conducted on P388 leukaemia cancer cells (Geiger et al., 2013); however, limited discussions of the

effects on PnTX on [Ca2+] have since been reported. The present results suggest that PnTX’s ability

to antagonise the highly Ca2+ permeable α7 nAChRs may be responsible for the observed decrease

in [Ca2+] levels. These changes in [Ca2+] levels following PnTX treatment could potentially explain

Page 105: A Novel Mechanism for Nicotinic Acetylcholine Receptor

90

the differences in flow cytometric and apoptosis inhibition studies compared to the other treatments

tested. However, further studies are required to confirm the relationship between PnTX and the

decrease in [Ca2+] via α7 nAChR antagonism.

The potential effects of Ca2+ in context of the PnTX induced cell-cycle arrest in the G0/G1

phase have been discussed in the previous chapter. Cells have been shown to be most susceptible to

depletions in Ca2+ during the G1 phase of the cell cycle. Ca2+ is important for the expression of the

cell cycle genes, FOS, JUN and MYC, which when depleted, reduce cell cycle progression through

the G1 phase (Kahl et al., 2003). The cytotoxic effects of PnTX may also result from the decreases

in intracellular Ca2+. A decrease in Ca2+ influx from the extracellular environment can cause the cell

to release intracellular Ca2+ reserves from the endoplasmic reticulum. In cases such as in the

presence of PnTX, stress through the chronic depletion of Ca2+ from the endoplasmic reticulum can

result in cell apoptosis via the intrinsic apoptotic pathway (Delom et al., 2007). The cytotoxic

effects of PnTX through endoplasmic reticulum stress-induced apoptosis correlates with the

increases in fragmented DNA and mitochondrial membrane permeability observed during the flow

cytometry studies.

In contrast to PnTX, MECA and ADI treatment induced a significant transient increase in

intracellular Ca2+ in the three carcinoma cell lines tested. Increases in intracellular Ca2+ can also

initiate cell apoptosis. A significant influx in intracellular Ca2+ can activate cell death effectors,

such as calpains, which stimulate the cleavage of downstream apoptotic effectors. Initiation of this

pathway ultimately results in cell apoptosis through mitochondrial permeabilisation and cytochrome

C release (Roderick et al., 2008). On the basis of this information, the various apoptotic inhibitors

investigated with PnTX would theoretically have a significant effect on MECA and API induced

cell death. However, the increase in [Ca2+] due to MECA and ADI treatment was unexpected due to

the similar antagonistic effects of both compounds on nAChRs. In addition, a similar increase in

[Ca2+] was shown following treatment with ACh, while ACh has no cytotoxic effects on carcinoma

Page 106: A Novel Mechanism for Nicotinic Acetylcholine Receptor

91

cells. This suggests that the cytotoxic effects of both MECA and ADI may operate through a Ca2+-

independent pathway, contrary to that of PnTX.

3.4 Combination Treatment

The final studies were conducted to explore the synergistic capacity of the most effective

nAChR modulators identified in Aims I-III to sensitise cancer cells to common chemotherapy

agents. It was hypothesised that the nAChR antagonists would enhance the cytotoxic nature of

chemotherapy agents, at concentrations that were relatively non-cytotoxic when administered alone.

Cell viability assay, flow cytometry and an isobologram analysis were utilised to identify the most

effective cytotoxic combination of nAChR modulators and chemotherapy agents.

3.4.1 CsA + MECA

Due to the consistent decrease in cell viability observed following treatment with the cell

death inhibitors in combination with PnTX in MDA-MB-231 cells, MECA (due to the limited

availability of PnTX) and CsA were further investigated across a range of toxic and non-toxic

concentrations to determine the combination relationship between the two compounds.

The effect of MECA 100 µM and 500 µM combination treatment with CsA 0.1 µM, 1.0 µM

and 5.0 µM on MDA-MB-231 cell viability was studied following 72 hours of exposure to the

treatment (Figure 22). No significant combinational effect was seen in cells treated with CsA 0.1

µM (Figure 22A). However, significant decreases in cell viability were seen following treatment

with CsA 1.0 µM and CsA 5.0 µM. CsA 1.0 µM + MECA 500 µM combination treatment produced

a significant decrease in cell viability (P < 0.001), compared to the single treatment with MECA

500 µM or CsA 1.0 µM (Figure 22B) alone. A similar effect was not seen with CsA 1.0 µM +

MECA 100 µM; while a strong combinational effect was seen with CsA 5.0 µM treatment (Figure

22C). CsA 5.0 µM + MECA 100 µM and CsA 5.0 µM + MECA 500 µM produced significant

decreases in cell viability, compared to the individual treatments (P < 0.001). The CsA 5.0 µM +

MECA 500 µM treatment demonstrated the greatest combinational effect, resulting in a 91.2%

Page 107: A Novel Mechanism for Nicotinic Acetylcholine Receptor

92

decrease in cell viability, compared to a 23.2% and 8.7% decrease for the respective CsA 5.0 µM

and MECA 500 µM treatments alone.

A

Untr

eate

d M

MEC

A 1

00

M

MEC

A 5

00

M

CsA

0.1

M

M +

MEC

A 1

00

CsA

0.1

M

M +

MEC

A 5

00

CsA

0.1

0

20

40

60

80

100

120

******

No

. o

f V

iab

le C

ell

s C

om

pa

red

to

Co

ntr

ol

B

Untr

eate

d M

MEC

A 1

00

M

MEC

A 5

00

M

CsA

1.0

M

M +

MEC

A 1

00

CsA

1.0

M

M +

MEC

A 5

00

CsA

1.0

0

20

40

60

80

100

120

***

***

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

C

Untr

eate

d M

MEC

A 1

00

M

MEC

A 5

00

M

CsA

5.0

M

M +

MEC

A 1

00

CsA

5.0

M

M +

MEC

A 5

00

CsA

5.0

0

20

40

60

80

100

120

* ***

******

***

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 22: The cytotoxic effect of combined MECA and CsA treatment in MDA-MB-231 cells. MDA-MB-231 cells were

treated with MECA 100 µM or 500 µM and CsA 0.1 μM (A), 1.0 µM (B) or 5.0 µM (C), or a combination of the two for

72 hours following a 24 hour seeding period. Untreated cells were used as a negative control. Cell viability was then

established via the MTT assay. Values are displayed as the mean percent of the control viability, with error bars

representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc test was used to determine significance

between the different treatment groups (* P < 0.05, *** P < 0.001).

Page 108: A Novel Mechanism for Nicotinic Acetylcholine Receptor

93

Given the substantial decrease in MDA-MB-231 viability following the combination

treatment with MECA and CsA, an isobologram was constructed to establish the relationship

between the two compounds. First, the IC50 values for each individual MECA and CsA treatment

were calculated and plotted on a dotplot. The line of additivity was then added, connecting the two

IC50 values. The line of additivity represents the theoretical effect of the two treatments, given each

treatment acts independently of the other. Next, the IC50 value for each treatment was calculated in

the presence of a known and constant concentration of the other treatment (e.g. IC50 of MECA in the

presence of CsA 5.0 μM). The various IC50 values are then plotted on the isobologram to determine

the relationship between the two treatments. In the event that the interaction between the two

treatments is more effective than the sum of the single effects (the line of additivity), then it can be

concluded that the treatments act synergistically. Alternatively, should the effect of the two

treatments be less effective than the sum of the individual effects, than an antagonistic relationship

is concluded. A sample diagram highlighting the line of additivity and the two possible

relationships between two treatments has been provided in Figure 23.

Page 109: A Novel Mechanism for Nicotinic Acetylcholine Receptor

94

Figure 23: Isobologram example displaying the line of additivity, connecting the IC50 values for two individual

treatments. The line of additivity represents the theoretical effect of the two treatments, given each treatment acts

independently of the other. The line of synergy represents an interaction between two treatments that is more effective

than the sum of the individual effects. Conversely, the line of antagonism characterises an interaction between the two

treatments that is less effective than the sum of the individual effects for each treatment.

An isobologram demonstrating the relationship between MECA and CsA treatment of MDA-

MB-231 cells, with respect to cell death, is displayed in Figure 24. The IC50 values for the single

treatment of MECA and CsA were determined to be 673.6 μM and 28.90 μM respectively. In

addition, the IC50 values for MECA and CsA were determined when in the presence of a constant

concentration of the alternate treatment. The IC50 value for CsA decreased to 11.40 μM in the

presence of MECA 100 μM, and further decreased to 0.9482 μM in the presence of MECA 500 μM.

Similar reductions in the IC50 value for MECA were seen in the presence of CsA. The IC50 value

dropped to 364.2 μM for MECA in the presence of CsA 1.0 μM, while further decreasing to 149.0

μM when in the presence of CsA 5.0 μM. The decrease in IC50 values for MECA and CsA have

been displayed visually in Figure 24. The data points for the fixed ratio treatment combinations fall

Page 110: A Novel Mechanism for Nicotinic Acetylcholine Receptor

95

well below the line of additivity, representing a pattern of synergistic effect, similar to that shown in

the example diagram in Figure 23.

Figure 24: Isobologram demonstrating the relationship between MECA and CsA treatment of MDA-MB-231 cells. Cells

were treated with a range of MECA (10 μM – 1500 μM) and CsA (0.1 μM – 40 μM) for 72 hours, following a 24 hour

seeding period, to determine the IC50 for each treatment. The line of additivity has been added between the MECA IC50

value (673.6 μM) and CsA IC50 value (28.90 μM). IC50 values for MECA and CsA, in the presence of constant

concentrations of the other treatment, are shown to determine the relationship between the two compounds, with

respect to cell death.

The Annexin V/PI double staining was assessed by flow cytometry to distinguish between

early apoptotic and necrotic cell death following the individual and combination treatment of MDA-

MB-231 cells. Cells were either left untreated, or treated with CsA 5 μM, MECA 500 μM, or the

combination of the two. The dotplots showing viable, apoptotic and necrotic cells are presented in

Figure 25, with the shift in Annexin V staining presented histographically. The results demonstrate

an increase in Annexin+/PI- apoptotic cells following the various treatments. Small levels of

apoptotic and necrotic cells were seen in the untreated control cells. A significant increase in early

apoptotic Annexin+/PI- cells was seen in all treatment groups (P < 0.001). CsA and MECA

treatment caused moderate increases in Annexin+/PI- cells, with MECA producing a greater

increase in apoptotic cells (10.50% and 20.34%, respectively). However, combination of the two

Page 111: A Novel Mechanism for Nicotinic Acetylcholine Receptor

96

treatments caused a shift in early apoptotic cells that was significantly higher than either individual

treatment (44.44%; P < 0.001). Furthermore, the cytotoxic effects of the combination treatment

caused a dramatic reduction in viable Annexin-/PI- cells to 19.44%, compared to the individual

treatments (CsA 70.52%, MECA 57.16%) and the untreated control (19.44%).

Page 112: A Novel Mechanism for Nicotinic Acetylcholine Receptor

97

Untreated CsA 5 µM P

rop

idiu

m I

od

ide

A B

Annexin V

MECA 500 µM CsA + MECA

Pro

pid

ium

Io

did

e

C D

Annexin V

E

Untr

eate

d M

CsA

5

M

MEC

A 5

00

CsA

+ M

EC

A

0

10

20

30

40

50

All Combinations = ***

Earl

y A

pop

toti

c C

ells

(%

)

Figure 25: Flow cytometric detection of apoptotic and necrotic cells by fluorescence-activated cell sorting of propidium

iodide and Annexin V labelled MDA-MB-231 cells. Cells were treated with CsA 5.0 μM (B), MECA 500 μM (C) or CsA

5.0 μM + MECA 500 μM (D) for 72 hours prior to staining and analysis. Untreated cells (A) were used as a negative

control. Dot-plots represent viable and apoptotic/necrotic cells. The bottom-left quadrant represents viable cells

(Annexin-/PI-). The bottom-right quadrant represents early apoptotic cells (Annexin+/PI-). Late apoptotic or secondary

necrotic cells are represented by the top-right quadrant (Annexin+/PI+). Early apoptotic (Annexin+/PI-) cells were

graphed (E) with the error bars representing the mean ± SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc

test was used to determine significance between all combinations (*** P < 0.001).

88.05%

7.39%

1.98%

2.58%

70.52%

14.76%

10.50%

4.22%

57.16%

15.06%

20.34%

7.44%

19.44%

30.92%

44.44%

5.20%

Page 113: A Novel Mechanism for Nicotinic Acetylcholine Receptor

98

3.4.2 Chemotherapy Agents + nAChR Antagonists

To further investigate the combinational effect of nAChR antagonists with common

chemotherapy agents in susceptible cell lines, the methods of (Hallett et al., 2012) were adopted.

A549 and SKBR3 cells were first treated with doxorubicin (DOXO) and docetaxel (DTX), and in

combination with ADI, to observe the corresponding reduction in cell viability. DOXO 1.0 μM,

DTX 100 μM and the combination of the two chemotherapy agents in A549 cells caused a moderate

reduction in cell viability, ranging from ~25-40% reduction in viable cells (Figure 26). Combination

of the treatments with a relatively weak cytotoxic concentration of ADI 200 μM caused little

change in the viability of the cell lines, with only DOXO + ADI causing a significant reduction

from the chemotherapy treatment alone (Figure 26A; P < 0.05).

Similar results were observed following individual and combination treatment with SKBR3

cells (Figure 27). A similar decrease in cell viability was seen following DOXO, DTX and DOXO

+ DTX treatment, resulting in a ~25-60% reduction in viable cells. No significant decrease in cell

viability was shown when combined with ADI 200 μM, compared to chemotherapy treatment

alone, for all combinations tested, despite the consistent ~20% reduction in cell viability caused by

ADI treatment alone.

Page 114: A Novel Mechanism for Nicotinic Acetylcholine Receptor

99

A

Untr

eate

d M

DO

XO

1.0

M

AD

I 20

0

DO

XO

+ A

DI

0

20

40

60

80

100

120

*N

o.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

B

Untr

eate

dM

DTX

100

D

MSO M

AD

I 20

0

DTX

+ A

DI

0

20

40

60

80

100

120

*

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

C

Untr

eate

d M

M +

DTX

100

DO

XO

0.1

M

AD

I 20

0

DO

XO

/DTX

+ A

DI

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

Figure 26: The cytotoxic effect of DOXO, DTX and ADI treatment in A549 cells. A549 cells were treated with DOXO

1.0 μM (A), DTX 100 μM (B) and DOXO + DTX (C), alone and in combination with ADI 200 μM for 48 hours

following a 4 hour seeding period. Untreated cells were used as a negative control, while a DMSO vehicle control was

used for DTX treatment. Cell viability was established via the MTT assay. Values are displayed as the mean percent of

the control viability, with error bars representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc test was

used to determine significance between the treatment groups (* P < 0.05).

Page 115: A Novel Mechanism for Nicotinic Acetylcholine Receptor

100

A

Untr

eate

d M

DO

XO

1.0

M

AD

I 20

0

DO

XO

+ A

DI

0

20

40

60

80

100

120N

o.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

B

Untr

eate

dM

DTX

100

D

MSO M

AD

I 20

0

DTX

+ A

DI

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

C

Untr

eate

d M

M +

DTX

100

DO

XO

0.1

M

AD

I 20

0

DO

XO

/DTX

+ A

DI

0

20

40

60

80

100

120

No.

of

Via

ble

Cel

ls C

om

pare

d t

o C

on

trol

Figure 27: The cytotoxic effect of DOXO, DTX and ADI treatment in SKBR3 cells. SKBR3 cells were treated with

DOXO 1.0 μM (A), DTX 100 μM (B) and DOXO + DTX (C), alone and in combination with ADI 200 μM for 48 hours

following a 4 hour seeding period. Untreated cells were used as a negative control, while a DMSO vehicle control was

used for DTX treatment. Cell viability was established via the MTT assay. Values are displayed as the mean percent of

the control viability, with error bars representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc test was

used to determine significance between treatment groups.

Further analysis of the combinational effects of various nAChR antagonists with the

combined cytotoxic treatment revealed similar results to those seen for ADI treatment. Relatively

mild cytotoxic concentrations of nAChR antagonists, including MECA 500 μM, PnTX 1 μM and

MLA 100 μM, were investigated in combination with DOXO 0.1 μM + DTX 100 μM treatment. No

significant decrease in cell viability was observed for A549 (Figure 28A) or SKBR3 (Figure 28B)

Page 116: A Novel Mechanism for Nicotinic Acetylcholine Receptor

101

when treated with nAChR antagonists and DOXO/DTX, compared to DOXO/DTX treatment alone

(P >0.05).

A

Untr

eate

d M

M +

DTX

100

DO

XO

0.1

M

MEC

A 5

00

M

PnTX

1

M

MLA

100

DO

XO

/DTX

+ M

EC

A

DO

XO

/DTX

+ P

nTX

DO

XO

/DTX

+ M

LA

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

B

Untr

eate

d M

M +

DTX

100

DO

XO

0.1

M

MEC

A 5

00

M

PnTX

1

M

MLA

100

DO

XO

/DTX

+ M

EC

A

DO

XO

/DTX

+ P

nTX

DO

XO

/DTX

+ M

LA

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 28: Determination of cell viability following treatment with DOXO, DTX and various nAChR antagonists in

A549 (A) and SKBR3 (B) cell lines. Cells were treated with DOXO 0.1 μM + DTX 100 μM, MECA 500 μM, PnTX 1μM,

MLA 100 μM or in various combinations for 48 hours, following a 4 hour seeding period. Untreated cells were used as

a negative control. Cell viability was established via the MTT assay. Values are displayed as the mean percent of the

control viability, with error bars representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-hoc was used to

determine significance between the treatment groups.

Page 117: A Novel Mechanism for Nicotinic Acetylcholine Receptor

102

Finally, the combination of DOXO 0.1 μM with nAChR antagonists was evaluated to

investigate any significant synergistic effects during cytotoxic combination treatment of cancer

cells. The viability of MDA-MB-231, HT29 and PC3 cells was measured following treatment with

DOXO 0.1 μM in combination with ADI 250 μM, MECA 100 μM, MLA 100 μM and 5-HT 100

μM. A significant decrease in cell viability was observed following DOXO + ADI treatment in

MDA-MB-231 cells, compared to DOXO treatment alone (P < 0.01; Figure 29A). A similar

significant decrease in DOXO + ADI and DOXO + MECA was seen in HT29 cells, compared to

DOXO treatment alone (P < 0.001; Figure 29B). However, no significant decrease in cell viability

was observed for all other treatment combinations in MDA-MB-231 and HT29 cells, and for all

combination treatments in PC3 cell lines (P > 0.05; Figure 29C).

Page 118: A Novel Mechanism for Nicotinic Acetylcholine Receptor

103

A

Untr

eate

d M

DO

XO

0.1

M

AD

I 25

0 M

MEC

A 1

00

M

MLA

100

M

5-H

T 1

00

DO

XO

+ A

DI

DO

XO

+ M

EC

A

DO

XO

+ M

LA

DO

XO

+ 5

-HT

0

20

40

60

80

100

120**

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

B

Untr

eate

d M

DO

XO

0.1

M

AD

I 25

0 M

MEC

A 1

00

M

MLA

100

M

5-H

T 1

00

DO

XO

+ A

DI

DO

XO

+ M

EC

A

DO

XO

+ M

LA

DO

XO

+ 5

-HT

0

20

40

60

80

100

120***

***

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

C

Untr

eate

d M

DO

XO

0.1

M

AD

I 25

0 M

MEC

A 1

00

M

MLA

100

M

5-H

T 1

00

DO

XO

+ A

DI

DO

XO

+ M

EC

A

DO

XO

+ M

LA

DO

XO

+ 5

-HT

0

20

40

60

80

100

120

No.

of

Via

ble

Cell

s C

om

pare

d t

o C

on

trol

Figure 29: Evaluation of cell viability following treatment with DOXO and various nAChR antagonists in MDA-MB-

231 (A), HT29 (B) and PC3 (C) cell lines. Cells were treated with DOXO 0.1 μM, ADI 250μM, MECA 500 μM, MLA

100 μM, 5-HT 100 μM or in various combinations for 48 hours, following a 4 hour seeding period. Untreated cells

were used as a negative control. Cell viability was established via the MTT assay. Values are displayed as the mean

percent of the control viability, with error bars representing SEM (n = 3). A one-way ANOVA with a Bonferroni post-

hoc was used to determine significance between the treatment groups.

Page 119: A Novel Mechanism for Nicotinic Acetylcholine Receptor

104

3.4.3 Discussion

The final studies investigated the possible synergistic effects of the most cytotoxic nAChR

modulators in combination with various chemotherapy agents. It was hypothesised that the

combination of nAChR antagonists would enhance the cytotoxic nature of the chemotherapy agents,

at concentrations that were relatively non-cytotoxic when administered alone. MECA co-treatment

with CsA was first investigated due to the synergistic effects observed during the apoptotic

inhibition studies with PnTX and CsA in various carcinoma cell lines (see Section 3.3.1 Cell Death

Inhibitors). MECA was chosen for continuation of the combination studies due to the unavailability

of PnTX. Treatment with a combination of MECA and CsA produced significant increases in

carcinoma cell death, compared to the individual treatments. The relationship between the two

compounds was determined as synergistic, with a significant reduction in IC50 value for CsA when

in the presence of relatively non-cytotoxic concentrations of MECA. From the isobologram, it

seems CsA has the greater effect on the IC50 of MECA.

The isobologram results demonstrated a synergistic effect between MECA and CsA. The

swayed nature of the IC50 points suggests that the synergistic effects are more evident under

different combinations of the two compounds. The presence of CsA has a greater effect on the IC50

of MECA, potentiating the cytotoxic effects of MECA presumably through enhancing the

downstream effects of nAChR antagonism. CsA acts via inhibiting the cyclophilin D protein,

preventing the opening of the mitochondrial permeability transition pore during apoptosis.

However, the results from studies with both MECA and PnTX suggest a secondary action of CsA,

whereby CsA acts as a phosphatase 2B inhibitor, with the ability to affect the desensitisation status

of nAChRs. Marszalec and colleagues previously investigated the desensitisation of nAChR during

modulation of kinase activation and phosphatase inhibition (Marszalec et al., 2005). The study

showed that nAChRs are present as two interconverting states of desensitisation, a rapidly

recovering D1 state, and a slower recovering D2 state. nAChRs important in cancer development,

such as the α4β2 subtype, are often driven towards the slowly recovering D2 state following

Page 120: A Novel Mechanism for Nicotinic Acetylcholine Receptor

105

continuous exposure to receptor modulators. The blocking action of CsA reduces the overall

desensitisation of nAChRs by facilitating a shift toward the faster recovering D1 receptor state

through the inhibition of nAChR dephosphorylation (Marszalec et al., 2005). The phosphatase 2B

inhibitory effects in the present combination study would effectively enhance the activity of the

potent nAChR antagonists, MECA and PnTX, resulting in the observed synergistic cytotoxic

effects. Consequently, CsA could potentially be used as a sensitising agent to enhance the effects of

nAChRs antagonists in cancer cell death.

Studies involving the co-treatment of carcinoma cell lines with nAChR modulators and

chemotherapy agents, doxorubicin and docetaxel, were conducted to further explore the possible

synergistic effects of the combination treatments. The present studies were based on those

previously performed by Hallet et al, where co-treatment of ADI with the chosen chemotherapy

agents produced a significant synergistic effect in a range of ‘sensitive’ carcinoma cell lines. The

study showed a significant synergistic effect was observed at concentrations of the two treatments

that would not elicit a cytotoxic response when administered individually. The present study used a

range of carcinoma cell lines, including the A549 and SKBR3 cell lines used in the Hallet et al

study, with similar concentrations of the treatments in an attempt to replicate and further the studies

previously reported. Contrasting results were observed during the present study, with co-treatment

of ADI and other nAChR modulators with the chemotherapy agents producing no clear synergistic

cytotoxic effects. Differences in the method used to determine cell viability between the present

studies (MTT assay), and the literature (AlamarBlue reduction method) may have contributed to the

conflicting results. The AlamarBlue viability assay is based on the reduction of the non-fluorescent

reagent resazurin, to the fluorescent compound resarufin, by viable cells. However, studies have

shown the reagent can interact and convert to the fluorescent compound through interactions with

various cell media types, independent of the presence of living cells. Incubation periods up to 4

hours produced ~15% increase in relative fluorescence when incubated in cell media without the

presence of cells (Munshi et al., 2014). This effect may have exacerbated the difference in treatment

Page 121: A Novel Mechanism for Nicotinic Acetylcholine Receptor

106

groups in the results by Hallet et al during the 24 hour incubation of AlamarBlue to determine cell

viability; while little differences were observed in the present study.

The concept of combining selective nAChR antagonists with chemotherapeutic agents is one

that has been alluded to throughout the current literature. Studies, such as that conducted with the

selective α7 antagonist, APS8, have demonstrated an ability to sensitise cells to apoptosis. NIC

treatment of SKMES-1 cells has been shown to increase the expression of a family of pro-survival

proteins, the inhibitors of apoptosis proteins (IAPs). The endogenous proteins bind and inactivate

caspases, effectively inhibiting cancer cell apoptosis. APS8 treatment was shown to be an effective

antagonist IAP activity. The results suggest that APS8 treatment was capable of preventing NIC-

induced resistance to apoptosis, by interfering with IAP activity, allowing the downstream

activation of caspases and ultimately cell apoptosis to occur (Zovko et al., 2013). Theoretically,

such effects of α7-nAChR antagonism would sensitise cancer cells to conventional chemotherapy

agents that attempt to induce apoptotic pathways. Doxorubicin and docetaxel both act via cytotoxic

mechanisms that do not directly target cancer cell apoptotic pathways. The present studies showed

no significant sensitisation to both chemotherapeutic agents when combined with various nAChR

antagonists. Combination of such antagonists with chemotherapeutic compounds that target specific

apoptotic pathways may better exploit the sensitising effects of nAChR antagonists.

Page 122: A Novel Mechanism for Nicotinic Acetylcholine Receptor

107

4 Discussion

4.1 Signalling through nAChRs is linked to Cancer Development

4.1.1 nAChR Agonist Induced Cancer Progression

In 2009, Schuller published a pivotal review highlighting the potential causal link between

smoking and nAChR driven tumour growth and metastasis. Nicotine and the highly potent nicotine-

derived nitrosamines are now recognised as potent agonists of nAChRs (Schuller, 2009). These

agonists promote carcinogenesis by stimulating angiogenesis and inhibiting apoptosis through

binding to the α7 subunit of the nAChR (Jull et al., 2001). Blocking these effects through the use of

selective antagonists offers new opportunities for the development of mechanisms that reduce

cancer development and progression.

4.1.2 nAChR Antagonist Driven Cytotoxicity

The recently identified family of shellfish toxins, PnTXs, demonstrate high antagonistic

affinity towards α7-nAChRs (Hellyer et al., 2011). In this research, PnTX-driven nAChR

antagonism demonstrated the most profound effect on cancer cell viability of all the antagonists

tested. The cytotoxic effects were more distinguishable in the adenocarcinoma cells lines,

highlighting expression of nAChRs outside of the nervous system.

Flow cytometry studies showed that treatment of the MDA-MB-231, HT29 and PC3

carcinoma cell lines with the nAChR antagonists MECA, ADI and 5-HT (through secondary

antagonistic effects) induces DNA fragmentation and opening of the mitochondrial transition pore,

indicative of early apoptosis. Interestingly, flow cytometry and apoptotic inhibition studies

suggested an alternative mechanism for PnTX induced cytotoxicity compared to the other nAChR

antagonists investigated. PnTX treatment induced a small, but significant increase in apoptotic cells

within the sub-G1 fraction of the cell cycle; while only displaying a slight increase in the

mitochondrial membrane permeability of treated cells. The flow cytometry profile of PnTX was

significantly different to that of the MECA, ADI and 5-HT treatment, despite displaying

Page 123: A Novel Mechanism for Nicotinic Acetylcholine Receptor

108

comparable cytotoxic effects during viability studies. Although a proposed mechanism for PnTX-

induced cell death of carcinoma cells has not been previously documented in the literature, the

present study suggests a cytotoxic mechanism associated with the ability of the α7-nAChR to

regulate intracellular calcium levels.

4.2 The nAChR Antagonist PnTX Induces Cytotoxicity by Altering

Intracellular Calcium Levels

The present study suggests that the cytotoxic effects of PnTX are mediated through the

observed antagonism of the highly permeable α7-, and to a lesser extent the α4β2-nAChRs,

inducing cell apoptosis through calcium-independent pathways.

Cell cycle analysis showed a potential G0/G1 phase arrest following PnTX treatment.

Transient rises in [Ca2+] are integral to the process of cell division (Kahl et al., 2003), suggesting

that changes in [Ca2+] through antagonism of α7 and α4β2 may play a role during PnTX treatment.

To investigate the mechanism of PnTX cytotoxicity further, inhibitors of common apoptotic

pathways were used in an attempt to reduce the cytotoxic effects of PnTX. PnTX-induced cell

cytotoxicity was not affected by the inhibition of the various intrinsic apoptotic processes,

suggesting an alternative mechanism for PnTXs cytotoxic effects. Intracellular calcium studies

revealed that following PnTX treatment there is a prolonged decrease in [Ca2+], suggesting a

possible cytotoxic action by inducing cell apoptosis through Ca2+-independent pathways. Although

the limited supply of PnTX available at the time of this research meant the experiments could not be

repeated, the promising initial findings warrant further investigation.

Page 124: A Novel Mechanism for Nicotinic Acetylcholine Receptor

109

4.3 Synergistic Cytotoxic Effects of Selective nAChR Antagonists with

Common Chemotherapeutic Agents

A second promising outcome of this research was the demonstration that selective nAChR

antagonists enhance the cytotoxicity of common chemotherapeutic agents. The effect of nAChR

antagonist treatment was assessed in combination with CsA and common chemotherapeutic agents

to explore the synergistic capacity of the compounds. The combination of nAChR antagonists was

expected to enhance the cytotoxic nature of the chemotherapeutic agents through sensitisation of the

cancer cells (Hallett et al., 2012). Contrary to expectations, combination of the nAChR antagonists

with doxorubicin and docetaxel displayed no synergistic effects, inconsistent with previous

literature. Alternatively, MECA treatment in combination with CsA produced a significant increase

in carcinoma cell death and early apoptotic cells, compared to the individual treatments. The

relationship between the two compounds was determined as synergistic based on the isobologram

results. The synergistic cytotoxic effects are attributed to the secondary action of CsA through

phosphatase 2B inhibition, effectively enhancing the activity of the potent nAChR antagonists. CsA

could potentially be used as a sensitising agent to enhance the effects of nAChRs antagonists in

cancer cell death. Although similar results were demonstrated when CsA was further investigated

with MECA, PnTX would have been the preferred investigational treatment.

4.4 Future Directions

The present study has established the relative cytotoxic nature of several nAChR modulators.

To further investigate the importance of nAChR in cancer development and whether nAChR

overexpression of nAChR can be used to selectively exploit cancer cells, studies on healthy

epithelial cells should be conducted. Previous studies, such as that conducted by Zovko and

colleagues on the apoptotic effects of APS8, have suggested that the antagonistic effects of selective

α7-nAChRs do not affect the proliferation of normal healthy cells. A possible theory for why

selective α7-nAChR antagonists, such as PnTX, may have a differential effect between normal and

Page 125: A Novel Mechanism for Nicotinic Acetylcholine Receptor

110

cancer cells, could be due to the number of binding sites present on the different cell types. Current

literature has repeatedly demonstrated the upregulation of the α7-nAChR in various cancer cell

types; while the present study has hinted at a significant relationship between greater levels of α7-

nAChR expression and an increased response to nAChR modulators. Studies looking at the effect of

PnTX on the viability and apoptotic induction of healthy epithelial cells would assist in determining

the possibility of exploiting nAChR overexpression in cancer treatment.

Further investigation into the cytotoxic mechanism of PnTX on the various carcinoma cell

types would play an important role in deducing the effects of nAChR in cancer cell death.

Observations from the present study have determined that decreases in intracellular calcium

following PnTX treatment are the likely cause of the cytotoxic effects. Knockout studies of various

nAChR subtypes, including the highly permeable α7 subunit, would further consolidate the present

findings. However, the complete mechanism and second messengers involved downstream of the

changes in intracellular calcium have not been determined. As discussed previously, Ca2+ is

important for the expression of the cell cycle genes, FOS, JUN and MYC. Investigation into the

changes in FOS, JUN and MYC expression during PnTX treatment would assist in determining

whether the decrease in intracellular calcium is having a significant effect on the cancer cell cycle.

Additionally, studies using PnTX to replicate the combination effect seen between MECA and CsA

treatment should be conducted, to investigate whether similar synergistic effects are observed.

Combination treatment of PnTX and CsA conducted on nAChR knockout cells would supplement

the current theory that CsA has a significant effect on the sensitivity of nAChRs to PnTX treatment.

Although the findings of this research highlight the potential application of nAChR

modulators in controlling cancer cell proliferation, further development is essential before they

could be used in a clinical setting. Naturally occurring antagonists often display potent inhibitory

actions on neuromuscular nAChRs, making them highly toxic when administered in vivo (Zovko et

al., 2013). The antagonistic behaviour at the neuromuscular junction often occurs at concentrations

below those required to induce cytotoxic effects on cancer cells, limiting the potential clinical use of

Page 126: A Novel Mechanism for Nicotinic Acetylcholine Receptor

111

such nAChR antagonists. PnTX falls into this category with potent effects on neuromuscular

nAChRs at a concentration of ~500 nM (Hellyer et al., 2011). The potential clinical use of nAChR

antagonists in cancer treatment would require alteration to the chemical structures that limit their

effects at the neuromuscular junction, while sustaining the potent effect on non-neuronal nAChRs.

Alternatively, a better understanding of how nAChR antagonists induce cancer cell cytotoxicity

may allow targeting of downstream effector molecules that eliminate the need for selective

targeting of nAChRs.

4.5 Final Conclusion

This research has identified a novel mechanism for the observed cytotoxicity of the highly

potent nAChR antagonist, PnTX, on human epithelial carcinoma cell lines. Cell cycle and

intracellular calcium studies suggest that PnTX exerts its cytotoxic effects by inducing changes in

the cell cycle via antagonism of the highly calcium permeable α7-, and to a certain extent, the α4β2-

nAChR receptors. This leads to an induction of cell apoptosis via calcium-independent pathways.

The use of nAChRs in combination with other known cytotoxic agents, such as CsA or apoptotic

inducing chemotherapy agents, may provide a sensitising tool that can reduce the dose required to

elicit a cytotoxic response.

Page 127: A Novel Mechanism for Nicotinic Acetylcholine Receptor

112

5 References

Adams JM, Cory S (2007). The Bcl-2 apoptotic switch in cancer development and therapy.

Oncogene 26(9): 1324-1337.

Akaike A, Tamura Y, Yokota T, Shimohama S, Kimura J (1994). Nicotine-induced protection of

cultured cortical neurons against N-methyl-D-aspartate receptor-mediated glutamate

cytotoxicity. Brain Res. 644(2): 181-187.

Al-Wadei MH, Al-Wadei HA, Schuller HM (2012). Pancreatic cancer cells and normal pancreatic

duct epithelial cells express an autocrine catecholamine loop that is activated by nicotinic

acetylcholine receptors α3, α5, and α7. Mol Cancer Res 10(2): 239-249.

Albuquerque EX, Pereira EF, Alkondon M, Rogers SW (2009). Mammalian nicotinic acetylcholine

receptors: from structure to function. Physiol Rev 89(1): 73-120.

Alonso E, Vale C, Vieytes MR, Laferla FM, Giménez-Llort L, Botana LM (2011). The cholinergic

antagonist gymnodimine improves Aβ and tau neuropathology in an in vitro model of

Alzheimer disease. Cell Physiol Biochem 27(6): 783.

Araoz R, Servent D, Molgó J, Iorga BI, Fruchart-Gaillard C, Benoit E, et al. (2011). Total synthesis

of pinnatoxins A and G and revision of the mode of action of pinnatoxin A. J Am Chem Soc

133(27): 10499-10511.

Arias HR, Richards VE, Ng D, Ghafoori ME, Le V, Mousa SA (2009). Role of non-neuronal

nicotinic acetylcholine receptors in angiogenesis. Int J Biochem Cell B 41(7): 1441-1451.

Arredondo J, Chernyavsky AI, Jolkovsky DL, Pinkerton KE, Grando SA (2007). Receptor-

mediated tobacco toxicity: alterations of the NF-κB expression and activity downstream of

α7 nicotinic receptor in oral keratinocytes. Life Sci 80(24): 2191-2194.

Arredondo J, Nguyen VT, Chernyavsky AI, Bercovich D, Orr-Urtreger A, Kummer W, et al.

(2002). Central role of α7 nicotinic receptor in differentiation of the stratified squamous

epithelium. J Cell Biol 159(2): 325-336.

Page 128: A Novel Mechanism for Nicotinic Acetylcholine Receptor

113

Baeriswyl V, Christofori G (2009). The angiogenic switch in carcinogenesis. Semin Cancer Biol

19(5): 329-337.

Balkan A, Avcu F, Ucar E, Gumus S, Tasci C, Elci P, et al. (2012). The evaluation of the

effectiveness of alpha bungarotoxin individually and combined with cisplatin on A549 lung

adenocancer cell line and SK-MES-1 lung squamous cell cancer cell line. Am J Respir Crit

Care Med 185: A6293.

Basso E, Petronilli V, Forte MA, Bernardi P (2008). Phosphate is essential for inhibition of the

mitochondrial permeability transition pore by cyclosporin A and by cyclophilin D ablation.

J Biol Chem 283(39): 26307-26311.

Beckel JM, Kanai A, Lee S-J, de Groat WC, Birder LA (2006). Expression of functional nicotinic

acetylcholine receptors in rat urinary bladder epithelial cells. Am J Physiol-Renal 290(1):

F103-F110.

Bourne Y, Radić Z, Aráoz R, Talley TT, Benoit E, Servent D, et al. (2010). Structural determinants

in phycotoxins and AChBP conferring high affinity binding and nicotinic AChR

antagonism. P Natl Acad Sci USA 107(13): 6076-6081.

Bourne Y, Sulzenbacher G, Radić Z, Aráoz R, Reynaud M, Benoit E, et al. (2015). Marine

macrocyclic imines, pinnatoxins A and G: structural determinants and functional properties

to distinguish neuronal α7 from muscle α1 2 βγδ nAChRs. Structure 23(6): 1106-1115.

Brown KC, Lau JK, Dom AM, Witte TR, Luo H, Crabtree CM, et al. (2012). MG624, an α7-

nAChR antagonist, inhibits angiogenesis via the Egr-1/FGF2 pathway. Angiogenesis 15(1):

99-114.

Catassi A, Paleari L, Servent D, Sessa F, Dominioni L, Ognio E, et al. (2008). Targeting α7-

nicotinic receptor for the treatment of pleural mesothelioma. Eur J Cancer 44(15): 2296-

2311.

Page 129: A Novel Mechanism for Nicotinic Acetylcholine Receptor

114

Catassi A, Viaggi S, Sessa F, Ognio E, Cilli M, Doria-Miglietta G, et al. (2006). α-cobratoxin, a

natural α 7-nicotinic receptor antagonist, as putative anticancer drug in non small cell lung

cancer and mesothelioma. Toxines Can: 99.

Chaffer CL, Weinberg RA (2011). A perspective on cancer cell metastasis. Science 331(6024):

1559-1564.

Changeux J-P (2010). Nicotine addiction and nicotinic receptors: lessons from genetically modified

mice. Nat Rev Neurosci 11(6): 389-401.

Chen CS, Lee CH, Hsieh CD, Ho CT, Pan MH, Huang CS, et al. (2011). Nicotine-induced human

breast cancer cell proliferation attenuated by garcinol through down-regulation of the

nicotinic receptor and cyclin D3 proteins. Breast Cancer Res Tr 125(1): 73-87.

Chen M, Won D-J, Krajewski S, Gottlieb RA (2002). Calpain and mitochondria in

ischemia/reperfusion injury. J Biol Chem 277(32): 29181-29186.

Chernyavsky AI, Arredondo J, Vetter DE, Grando SA (2007). Central role of α9 acetylcholine

receptor in coordinating keratinocyte adhesion and motility at the initiation of

epithelialization. Exp Cell Res 313(16): 3542-3555.

Cregan IL, Dharmarajan AM, Fox SA (2013). Mechanisms of cisplatin-induced cell death in

malignant mesothelioma cells: role of inhibitor of apoptosis proteins (IAPs) and caspases.

Int J Oncol 42(2): 444-452.

Cucina A, Dinicola S, Coluccia P, Proietti S, D'Anselmi F, Pasqualato A, et al. (2012). Nicotine

stimulates proliferation and inhibits apoptosis in colon cancer cell lines through activation of

survival pathways. J Surg Res 178(1): 233-241.

Cui W, Hu S, Chan HH, Luo J, Li W, Mak S, et al. (2013). Bis (12)-hupyridone, a novel

acetylcholinesterase inhibitor, protects against glutamate-induced neuronal excitotoxicity via

activating α7 nicotinic acetylcholine receptor/phosphoinositide 3-kinase/Akt cascade. Chem

Biol Interact 203(1): 365-370.

Page 130: A Novel Mechanism for Nicotinic Acetylcholine Receptor

115

Cummings BS, Kinsey GR, Bolchoz LJ, Schnellmann RG (2004). Identification of caspase-

independent apoptosis in epithelial and cancer cells. J Pharmacol Exp Ther 310(1): 126-

134.

Dajas-Bailador F, Wonnacott S (2004). Nicotinic acetylcholine receptors and the regulation of

neuronal signalling. Trends Pharmacol Sci 25(6): 317-324.

Daniell HW (1980). Estrogen receptors, breast cancer, and smoking. New Eng J Med 302(26):

1478.

Dasgupta P, Rizwani W, Pillai S, Kinkade R, Kovacs M, Rastogi S, et al. (2009). Nicotine induces

cell proliferation, invasion and epithelial‐mesenchymal transition in a variety of human

cancer cell lines. Int J Cancer 124(1): 36-45.

Delom F, Fessart D, Chevet E (2007). Regulation of calnexin sub-cellular localization modulates

endoplasmic reticulum stress-induced apoptosis in MCF-7 cells. Apoptosis 12(2): 293-305.

Dotan I, Rachmilewitz D, Schreiber S, Eliakim R, van der Woude CJ, Kornbluth A, et al. (2010). A

randomised placebo-controlled multicentre trial of intravenous semapimod HCl for

moderate to severe Crohn's disease. Gut 59(6): 760-766.

Dragunow M, Trzoss M, Brimble MA, Cameron R, Beuzenberg V, Holland P, et al. (2005).

Investigations into the cellular actions of the shellfish toxin gymnodimine and analogues.

Environ Toxicol Phar 20(2): 305-312.

Durany N, Zöchling R, Boissl KW, Paulus W, Ransmayr G, Tatschner T, et al. (2000). Human

post-mortem striatal α4β2 nicotinic acetylcholine receptor density in schizophrenia and

Parkinson's syndrome. Neurosci Lett 287(2): 109-112.

Egleton RD, Brown KC, Dasgupta P (2008). Nicotinic acetylcholine receptors in cancer: multiple

roles in proliferation and inhibition of apoptosis. Trends Pharmacol Sci 29(3): 151-158.

Elmore S (2007). Apoptosis: a review of programmed cell death. Toxicol Pathol 35(4): 495-516.

Epperson CN, O’Malley S, Czarkowski KA, Gueorguieva R, Jatlow P, Sanacora G, et al. (2005).

Sex, GABA, and nicotine: the impact of smoking on cortical GABA levels across the

Page 131: A Novel Mechanism for Nicotinic Acetylcholine Receptor

116

menstrual cycle as measured with proton magnetic resonance spectroscopy. Biol Psychiat

57(1): 44-48.

Ferrara N (2009). Vascular endothelial growth factor. Arterioscler Thromb Vasc Biol 29(6): 789-

791.

Freedman R, Adams CE, Leonard S (2000). The α7-nicotinic acetylcholine receptor and the

pathology of hippocampal interneurons in schizophrenia. J Chem Neuroanat 20(3): 299-

306.

Gamen S, Anel A, Pérez-Galán P, Lasierra P, Johnson D, Piñeiro A, et al. (2000). Doxorubicin

treatment activates a Z-Vad-sensitive caspase, which causes Δψm loss, caspase-9 activity,

and apoptosis in jurkat cells. Exp Cell Res 258(1): 223-235.

Garduño J, Galindo-Charles L, Jiménez-Rodríguez J, Galarraga E, Tapia D, Mihailescu S, et al.

(2012). Presynaptic α4β2 nicotinic acetylcholine receptors increase glutamate release and

serotonin neuron excitability in the dorsal raphe nucleus. J Neurosci 32(43): 15148-15157.

Geiger M, Desanglois G, Hogeveen K, Fessard V, Leprêtre T, Mondeguer F, et al. (2013).

Cytotoxicity, fractionation and dereplication of extracts of the dinoflagellate vulcanodinium

rugosum, a producer of pinnatoxin G. Marine Drugs 11(9): 3350-3371.

Gentry CL, Lukas RJ (2001). Local anesthetics noncompetitively inhibit function of four distinct

nicotinic acetylcholine receptor subtypes. J Pharmacol Exp Ther 299(3): 1038-1048.

Gil-Ad I, Zolokov A, Lomnitski L, Taler M, Bar M, Luria D, et al. (2008). Evaluation of the

potential anti-cancer activity of the antidepressant sertraline in human colon cancer cell lines

and in colorectal cancer-xenografted mice. Int J Oncol 33(2): 277-286.

Gotti C, Guiducci S, Tedesco V, Corbioli S, Zanetti L, Moretti M, et al. (2010). Nicotinic

acetylcholine receptors in the mesolimbic pathway: primary role of ventral tegmental area

α6β2* receptors in mediating systemic nicotine effects on dopamine release, locomotion,

and reinforcement. J Neurosci 30(15): 5311-5325.

Page 132: A Novel Mechanism for Nicotinic Acetylcholine Receptor

117

Gotti C, Zoli M, Clementi F (2006). Brain nicotinic acetylcholine receptors: native subtypes and

their relevance. Trends Pharmacol Sci 27(9): 482-491.

Grando SA (2014). Connections of nicotine to cancer. Nat Rev Cancer 14(6): 419-429.

Grando SA, Pittelkow MR, Schallreuter KU (2006). Adrenergic and cholinergic control in the

biology of epidermis: physiological and clinical significance. J Invest Dermatol 126(9):

1948-1965.

Gray JA, Roth BL (2001). Paradoxical trafficking and regulation of 5-HT 2A receptors by agonists

and antagonists. Brain Res Bull 56(5): 441-451.

Greene CM, Ramsay H, Wells RJ, O'Neill SJ, McElvaney NG (2010). Inhibition of toll-like

receptor 2-mediated interleukin-8 production in cystic fibrosis airway epithelial cells via the

7-nicotinic acetylcholine receptor. Mediators Inflamm 2010: 423241.

Guo J, Ibaragi S, Zhu T, Luo L-Y, Hu G-F, Huppi PS, et al. (2008). Nicotine promotes mammary

tumor migration via a signaling cascade involving protein kinase C and CDC42. Cancer Res

68(20): 8473-8481.

Hallett R, Hassell J (2012). A method of treating cancer. WIPO/PCT. WO 2012/106812 A1,

McMaster University, Canada.

Hanahan D, Folkman J (1996). Patterns and emerging mechanisms of the angiogenic switch during

tumorigenesis. Cell 86(3): 353-364.

Hanahan D, Weinberg RA (2011). Hallmarks of cancer: the next generation. Cell 144(5): 646-674.

Heeschen C, Jang JJ, Weis M, Pathak A, Kaji S, Hu RS, et al. (2001). Nicotine stimulates

angiogenesis and promotes tumor growth and atherosclerosis. Nat Med 7(7): 833-839.

Heeschen C, Weis M, Aicher A, Dimmeler S, Cooke JP (2002). A novel angiogenic pathway

mediated by non-neuronal nicotinic acetylcholine receptors. J Clin Invest 110(4): 527-536.

Hellyer SD (2014). Mechanism of action of pinnatoxins E, F and G. University of Otago, New

Zealand.

Page 133: A Novel Mechanism for Nicotinic Acetylcholine Receptor

118

Hellyer SD, Selwood AI, Rhodes L, Kerr DS (2011). Marine algal pinnatoxins E and F cause

neuromuscular block in an in vitro hemidiaphragm preparation. Toxicon 58(8): 693-699.

Ho YS, Pan MH (2013). Tea extracts confer its antiproliferating effects through inhibition of

nicotine-and estrogen-induced 9-nicotinic acetylcholine receptor upregulation in human

breast cancer cells. Nat Func Prop Food 1: 256-268.

Hung CS, Peng YJ, Wei PL, Lee CH, Su HY, Ho YS, et al. (2011). The alpha9 nicotinic

acetylcholine receptor is the key mediator in nicotine-enhanced cancer metastasis in breast

cancer cells. J Exp Clin Med 3(6): 283-292.

Jull B, Plummer H, Schuller H (2001). Nicotinic receptor-mediated activation by the tobacco-

specific nitrosamine NNK of a Raf-1/MAP kinase pathway, resulting in phosphorylation of

c-myc in human small cell lung carcinoma cells and pulmonary neuroendocrine cells. J

Cancer Res Clin Oncol 127(12): 707-717.

Kahl CR, Means AR (2003). Regulation of cell cycle progression by calcium/calmodulin-dependent

pathways. Endocr Rev 24(6): 719-736.

Kalamida D, Poulas K, Avramopoulou V, Fostieri E, Lagoumintzis G, Lazaridis K, et al. (2007).

Muscle and neuronal nicotinic acetylcholine receptors. FEBS J 274(15): 3799-3845.

Kaneko S, Maeda T, Kume T, Kochiyama H, Akaike A, Shimohama S, et al. (1997). Nicotine

protects cultured cortical neurons against glutamate-induced cytotoxicity via α7-neuronal

receptors and neuronal CNS receptors. Brain Res 765(1): 135-140.

Kawai H, Berg DK (2001). Nicotinic acetylcholine receptors containing α7 subunits on rat cortical

neurons do not undergo long‐lasting inactivation even when up‐regulated by chronic

nicotine exposure. J Neurochem 78(6): 1367-1378.

Kawamata J, Suzuki S, Shimohama S (2012). α7 nicotinic acetylcholine receptor mediated

neuroprotection in Parkinson’s disease. Curr Drug Targets 13(5): 623-630.

Page 134: A Novel Mechanism for Nicotinic Acetylcholine Receptor

119

Kawashima K, Yoshikawa K, Fujii YX, Moriwaki Y, Misawa H (2007). Expression and function of

genes encoding cholinergic components in murine immune cells. Life Sci 80(24): 2314-

2319.

Khalil AA, Jameson MJ, Broaddus WC, Lin PS, Chung TD (2013). Nicotine enhances proliferation,

migration, and radioresistance of human malignant glioma cells through EGFR activation.

Brain Tumor Pathol 30(2): 73-83.

Kharrat R, Servent D, Girard E, Ouanounou G, Amar M, Marrouchi R, et al. (2008). The marine

phycotoxin gymnodimine targets muscular and neuronal nicotinic acetylcholine receptor

subtypes with high affinity. J Neurochem 107(4): 952-963.

Kurzen H, Berger H, Jäger C, Hartschuh W, Näher H, Gratchev A, et al. (2004). Phenotypical and

molecular profiling of the extraneuronal cholinergic system of the skin. J Invest Dermatol

123(5): 937-949.

Lam DC-l, Girard L, Ramirez R, Chau W-s, Suen W-s, Sheridan S, et al. (2007). Expression of

nicotinic acetylcholine receptor subunit genes in non–small-cell lung cancer reveals

differences between smokers and nonsmokers. Cancer Res 67(10): 4638-4647.

Lau JK, Brown KC, Thornhill BA, Crabtree CM, Dom AM, Witte TR, et al. (2013). Inhibition of

cholinergic signaling causes apoptosis in human bronchioalveolar carcinoma. Cancer Res

73(4): 1328-1339.

Le Novere N, Changeux JP (1995). Molecular evolution of the nicotinic acetylcholine receptor: an

example of multigene family in excitable cells. J Mol Evol 40(2): 155-172.

Lee CH, Chang YC, Chen CS, Tu SH, Wang YJ, Chen LC, et al. (2011). Crosstalk between

nicotine and estrogen-induced estrogen receptor activation induces α9-nicotinic

acetylcholine receptor expression in human breast cancer cells. Breast Cancer Res Treat

129(2): 331-345.

Page 135: A Novel Mechanism for Nicotinic Acetylcholine Receptor

120

Lee CH, Huang CS, Chen CS, Tu SH, Wang YJ, Chang YJ, et al. (2010). Overexpression and

activation of the α9-nicotinic receptor during tumorigenesis in human breast epithelial cells.

J Natl Cancer Inst 102(17): 1322-1335.

Li X-W, Wang H (2006). Non-neuronal nicotinic alpha 7 receptor, a new endothelial target for

revascularization. Life Sci 78(16): 1863-1870.

Lieberman JA, Dunbar G, Segreti AC, Girgis RR, Seoane F, Beaver JS, et al. (2013). A randomized

exploratory trial of an alpha-7 nicotinic receptor agonist (TC-5619) for cognitive

enhancement in schizophrenia. Neuropsychopharmacol 38(6): 968-975.

Livett BG, Sandall DW, Keays D, Down J, Gayler KR, Satkunanathan N, et al. (2006). Therapeutic

applications of conotoxins that target the neuronal nicotinic acetylcholine receptor. Toxicon

48(7): 810-829.

Lombardo S, Maskos U (2014). Role of the nicotinic acetylcholine receptor in Alzheimer's disease

pathology and treatment. Neuropharmacology 96(Pt B): 255-262.

Lyukmanova E, Shulepko M, Bychkov M, Shenkarev Z, Paramonov A, Chugunov A, et al. (2014).

Human SLURP-1 and SLURP-2 proteins acting on nicotinic acetylcholine receptors reduce

poliferation of human colorectal adenocarcinoma HT-29 cells. Acta Naturae 6(4): 60.

Mai H, May WS, Gao F, Jin Z, Deng X (2003). A functional role for nicotine in Bcl2

phosphorylation and suppression of apoptosis. J Biol Chem 278(3): 1886-1891.

Marszalec W, Yeh JZ, Narahashi T (2005). Desensitization of nicotine acetylcholine receptors:

modulation by kinase activation and phosphatase inhibition. Eur J Pharmacol 514(2): 83-

90.

Martin-Ruiz C, Lee M, Perry R, Baumann M, Court J, Perry E (2004). Molecular analysis of

nicotinic receptor expression in autism. Mol Brain Res 123(1): 81-90.

Martin-Ruiz CM, Haroutunian VH, Long P, Young AH, Davis KL, Perry EK, et al. (2003).

Dementia rating and nicotinic receptor expression in the prefrontal cortex in schizophrenia.

Biol Psychiatry 54(11): 1222-1233.

Page 136: A Novel Mechanism for Nicotinic Acetylcholine Receptor

121

Mei D, Lin Z, Fu J, He B, Gao W, Ma L, et al. (2015). The use of α-conotoxin ImI to actualize the

targeted delivery of paclitaxel micelles to α7 nAChR-overexpressing breast cancer.

Biomaterials 42: 52-65.

Meyer PM, Strecker K, Kendziorra K, Becker G, Hesse S, Woelpl D, et al. (2009). Reduced

α4β2*–nicotinic acetylcholine receptor binding and its relationship to mild cognitive and

depressive symptoms in parkinson disease. Arch Gen Psychiatry 66(8): 866-877.

Miyazawa A, Fujiyoshi Y, Unwin N (2003). Structure and gating mechanism of the acetylcholine

receptor pore. Nature 423(6943): 949-955.

Moffett J, Kratz E, Stachowiak MK (1998). Increased tyrosine phosphorylation and novel cis-acting

element mediate activation of the fibroblast growth factor-2 (FGF-2) gene by nicotinic

acetylcholine receptor. New mechanism for trans-synaptic regulation of cellular

development and plasticity. Mol Brain Res 55(2): 293-305.

Momeni HR (2011). Role of calpain in apoptosis. Cell Journal (Yakhteh) 13(2): 65.

Mosmann T (1983). Rapid colorimetric assay for cellular growth and survival: application to

proliferation and cytotoxicity assays. J Immunol Methods 65(1): 55-63.

Munshi S, Twining RC, Dahl R (2014). Alamar blue reagent interacts with cell-culture media

giving different fluorescence over time: potential for false positives. J Pharmacol Toxicol

Methods 70(2): 195-198.

Nakayama H, Shimoke K, Isosaki M, Satoh H, Yoshizumi M, Ikeuchi T (2006). Subtypes of

neuronal nicotinic acetylcholine receptors involved in nicotine-induced phosphorylation of

extracellular signal-regulated protein kinase in PC12h cells. Neurosci Lett 392(1): 101-104.

Natori T, Sata M, Washida M, Hirata Y, Nagai R, Makuuchi M (2003). Nicotine enhances

neovascularization and promotes tumor growth. Mol Cells 16(2): 143-146.

Newman DJ, Cragg GM (2014). Marine-sourced anti-cancer and cancer pain control agents in

clinical and late preclinical development. Marine Drugs 12(1): 255-278.

Page 137: A Novel Mechanism for Nicotinic Acetylcholine Receptor

122

Ng MK, Wu J, Chang E, Wang B-y, Katzenberg-Clark R, Ishii-Watabe A, et al. (2007). A central

role for nicotinic cholinergic regulation of growth factor–induced endothelial cell migration.

Arterioscler Thromb Vasc Biol 27(1): 106-112.

Nguyen VT, Hall L, Gallacher G, Ndoye A, Jolkovsky D, Webber R, et al. (2000). Choline

acetyltransferase, acetylcholinesterase, and nicotinic acetylcholine receptors of human

gingival and esophageal epithelia. J Dent Res 79(4): 939-949.

Nishimoto T, Kadoyama K, Taniguchi T, Takano M, Otani M, Nakamura-Hirota T, et al. (2011).

Synaptotagmin1 synthesis induced by synaptic plasticity in mouse hippocampus through

activation of nicotinic acetylcholine receptors. Neurosci Lett 489(1): 25-29.

Nishioka T, Guo J, Yamamoto D, Chen L, Huppi P, Chen CY (2010). Nicotine, through

upregulating pro‐survival signaling, cooperates with NNK to promote transformation. J Cell

Biochem 109(1): 152-161.

Nishioka T, Kim H-S, Luo L-Y, Huang Y, Guo J, Chen CY (2011). Sensitization of epithelial

growth factor receptors by nicotine exposure to promote breast cancer cell growth. Breast

Cancer Res 13(6): R113.

Nitahara JA, Cheng W, Liu Y, Li B, Leri A, Li P, et al. (1998). Intracellular calcium, DNase

activity and myocyte apoptosis in aging Fischer 344 rats. J Mol Cell Cardiol 30(3): 519-

535.

Niu X-M, Lu S (2014). Acetylcholine receptor pathway in lung cancer: new twists to an old story.

World J Clin Oncology 5(4): 667.

Olincy A, Harris JG, Johnson LL, Pender V, Kongs S, Allensworth D, et al. (2006). Proof-of-

concept trial of an α7 nicotinic agonist in schizophrenia. Arch Gen Psychiatry 63(6): 630-

638.

Paleari L, Grozio A, Cesario A, Russo P (2008). The cholinergic system and cancer. Semin Cancer

Biol 18(3): 211-217.

Page 138: A Novel Mechanism for Nicotinic Acetylcholine Receptor

123

Paleari L, Trombino S, Falugi C, Gallus L, Carlone S, Angelini C, et al. (2006). Marine sponge-

derived polymeric alkylpyridinium salts as a novel tumor chemotherapeutic targeting the

cholinergic system in lung tumors. Int J Oncol 29(6): 1381-1388.

Palma E, Bertrand S, Binzoni T, Bertrand D (1996). Neuronal nicotinic alpha 7 receptor expressed

in Xenopus oocytes presents five putative binding sites for methyllycaconitine. J Physio

491(1): 151-161.

Papke RL, Sanberg PR, Shytle RD (2001). Analysis of mecamylamine stereoisomers on human

nicotinic receptor subtypes. J Pharmacol Exp Ther 297(2): 646-656.

Parri HR, Hernandez CM, Dineley KT (2011). Research update: Alpha7 nicotinic acetylcholine

receptor mechanisms in Alzheimer's disease. Biochem Pharmacol 82(8): 931-942.

Parsons DW, Jones S, Zhang X, Lin JC-H, Leary RJ, Angenendt P, et al. (2008). An integrated

genomic analysis of human glioblastoma multiforme. Science 321(5897): 1807-1812.

Pettersson A, Nylund G, Khorram-Manesh A, Nordgren S, Delbro DS (2009). Nicotine induced

modulation of SLURP-1 expression in human colon cancer cells. Auton Neurosci 148(1):

97-100.

Plummer 3rd H, Dhar M, Schuller HM (2005). Expression of the alpha7 nicotinic acetylcholine

receptor in human lung cells. Respir Res 6(1): 29.

Pohanka M (2012). Alpha7 nicotinic acetylcholine receptor is a target in pharmacology and

toxicology. Int J Mol Sci 13(2): 2219-2238.

Roderick HL, Cook SJ (2008). Ca2+ signalling checkpoints in cancer: remodelling Ca2+ for cancer

cell proliferation and survival. Nat Rev Cancer 8(5): 361-375.

Rosas-Ballina M, Goldstein RS, Gallowitsch-Puerta M, Yang L, Valdés-Ferrer SI, Patel NB, et al.

(2009). The selective α7 agonist GTS-21 attenuates cytokine production in human whole

blood and human monocytes activated by ligands for TLR2, TLR3, TLR4, TLR9, and

RAGE. Mol Med 15(7-8): 195-202.

Page 139: A Novel Mechanism for Nicotinic Acetylcholine Receptor

124

Sargent PB (1993). The diversity of neuronal nicotinic acetylcholine receptors. Annu Rev Neurosci

16(1): 403-443.

Schuller HM (2009). Is cancer triggered by altered signalling of nicotinic acetylcholine receptors?

Nat Rev Cancer 9(3): 195-205.

Schuller HM (2007). Nitrosamines as nicotinic receptor ligands. Life Sci 80(24): 2274-2280.

Schuller HM, Al-Wadei HA, Majidi M (2008). Gamma-aminobutyric acid, a potential tumor

suppressor for small airway-derived lung adenocarcinoma. Carcinogenesis 29(10): 1979-

1985.

Sheppard BJ, Williams M, Plummer HK, Schuller HM (2000). Activation of voltage-operated Ca2+-

channels in human small cell lung carcinoma by the tobacco-specific nitrosamine 4-

(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Int J Oncol 16(3): 513-521.

Shih YL, Liu HC, Chen CS, Hsu CH, Pan MH, Chang HW, et al. (2009). Combination treatment

with luteolin and quercetin enhances antiproliferative effects in nicotine-treated MDA-MB-

231 cells by down-regulating nicotinic acetylcholine receptors. J Agric Food Chem 58(1):

235-241.

Shin VY, Wu WK, Chu K-M, Wong HP, Lam EK, Tai EK, et al. (2005). Nicotine induces

cyclooxygenase-2 and vascular endothelial growth factor receptor-2 in association with

tumor-associated invasion and angiogenesis in gastric cancer. Mol Cancer Res 3(11): 607-

615.

Skok M, Grailhe R, Agenes F, Changeux JP (2007). The role of nicotinic receptors in B-

lymphocyte development and activation. Life Sci 80(24): 2334-2336.

Smith MA, Schnellmann RG (2012). Calpains, mitochondria, and apoptosis. Cardiovasc Res 96(1):

32-37.

Song P, Sekhon HS, Fu XW, Maier M, Jia Y, Duan J, et al. (2008). Activated cholinergic signaling

provides a target in squamous cell lung carcinoma. Cancer Res 68(12): 4693-4700.

Page 140: A Novel Mechanism for Nicotinic Acetylcholine Receptor

125

Takada Y, Yonezawa A, Kume T, Katsuki H, Kaneko S, Sugimoto H, et al. (2003). Nicotinic

acetylcholine receptor-mediated neuroprotection by donepezil against glutamate

neurotoxicity in rat cortical neurons. J Pharmacol Exp Ther 306(2): 772-777.

Tallarida RJ (2006). An overview of drug combination analysis with isobolograms. J Pharmacol

Exp Ther 319(1): 1-7.

The FO, Boeckxstaens GE, Snoek SA, Cash JL, Bennink R, LaRosa GJ, et al. (2007). Activation of

the cholinergic anti-inflammatory pathway ameliorates postoperative ileus in mice.

Gastroenterology 133(4): 1219-1228.

Thornhill BA, Brown KC, Lau JK, Dom AM, Witte TR, Hardman WE, et al. (2013). Anti-

angiogenic activity of alpha7-nicotinic receptor antagonists in human small cell lung cancer.

The FASEB Journal 27 (Meeting Abstract): 1087.14.

Trombino S, Cesario A, Margaritora S, Granone P, Motta G, Falugi C, et al. (2004). α7-nicotinic

acetylcholine receptors affect growth regulation of human mesothelioma cells role of

mitogen-activated protein kinase pathway. Cancer Res 64(1): 135-145.

Tsurutani J, Castillo SS, Brognard J, Granville CA, Zhang C, Gills JJ, et al. (2005). Tobacco

components stimulate Akt-dependent proliferation and NFκB-dependent survival in lung

cancer cells. Carcinogenesis 26(7): 1182-1195.

Tu SH, Ku CY, Ho CT, Chen CS, Huang CS, Lee CH, et al. (2011). Tea polyphenol (−)‐

epigallocatechin‐3‐gallate inhibits nicotine‐and estrogen‐induced α9‐nicotinic acetylcholine

receptor upregulation in human breast cancer cells. Mol Nutr Food Res 55(3): 455-466.

Tzartos SJ, Barkas T, Cung MT, Mamalaki A, Marraud M, Orlewski P, et al. (1998). Anatomy of

the antigenic structure of a large memberane autoantigen, the muscle‐type nicotinic

acetylcholine receptor. Immunol Rev 163(1): 89-120.

Unwin N (2005). Refined structure of the nicotinic acetylcholine receptor at 4Å resolution. J Mol

Biol 346(4): 967-989.

Page 141: A Novel Mechanism for Nicotinic Acetylcholine Receptor

126

Volm M, Rittgen W, Drings P (1998). Prognostic value of ERBB-1, VEGF, cyclin A, FOS, JUN

and MYC in patients with squamous cell lung carcinomas. Br J Cancer 77(4): 663.

Wessler I, Bittinger F, Kamin W, Zepp F, Meyer E, Schad A, et al. (2007). Dysfunction of the non-

neuronal cholinergic system in the airways and blood cells of patients with cystic fibrosis.

Life Sci 80(24): 2253-2258.

Wessler I, Kirkpatrick C (2008). Acetylcholine beyond neurons: the non‐neuronal cholinergic

system in humans. Br J Pharmacol 154(8): 1558-1571.

Wessler I, Reinheimer T, Kilbinger H, Bittinger F, Kirkpatrick CJ, Saloga J, et al. (2003). Increased

acetylcholine levels in skin biopsies of patients with atopic dermatitis. Life Sci 72(18):

2169-2172.

West KA, Brognard J, Clark AS, Linnoila IR, Yang X, Swain SM, et al. (2003). Rapid Akt

activation by nicotine and a tobacco carcinogen modulates the phenotype of normal human

airway epithelial cells. J Clin Invest 111(1): 81-90.

Wilens TE, Decker MW (2007). Neuronal nicotinic receptor agonists for the treatment of attention-

deficit/hyperactivity disorder: focus on cognition. Biochem Pharmacol 74(8): 1212-1223.

Wlodkowic D, Telford W, Skommer J, Darzynkiewicz Z (2011). Apoptosis and beyond: cytometry

in studies of programmed cell death. Methods Cell Biol 103: 55.

Wong HPS, Yu L, Lam EKY, Tai EKK, Wu WKK, Cho CH (2007). Nicotine promotes cell

proliferation via α7-nicotinic acetylcholine receptor and catecholamine-synthesizing

enzymes-mediated pathway in human colon adenocarcinoma HT-29 cells. Toxicol Appl

Pharmacol 221(3): 261-267.

Wu CH, Lee CH, Ho YS (2011). Nicotinic acetylcholine receptor-based blockade: applications of

molecular targets for cancer therapy. Clin Cancer Res 17(11): 3533-3541.

Xu J, Huang H, Pan C, Zhang B, Liu X, Zhang L (2007). Nicotine inhibits apoptosis induced by

cisplatin in human oral cancer cells. Int J Oral Maxillofac Surg 36(8): 739-744.

Page 142: A Novel Mechanism for Nicotinic Acetylcholine Receptor

127

Xu L, Deng X (2004). Tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-

butanone induces phosphorylation of μ-and m-calpain in association with increased

secretion, cell migration, and invasion. J Biol Chem 279(51): 53683-53690.

Yoshida J, Ishibashi T, Nishio M (2007). G1 cell cycle arrest by amlodipine, a dihydropyridine Ca

2+ channel blocker, in human epidermoid carcinoma A431 cells. Biochem Pharmacol 73(7):

943-953.

Young JW, Geyer MA (2013). Evaluating the role of the alpha-7 nicotinic acetylcholine receptor in

the pathophysiology and treatment of schizophrenia. Biochem Pharmacol 86(8): 1122-1132.

Zhan C, Li B, Hu L, Wei X, Feng L, Fu W, et al. (2011). Micelle‐based brain‐targeted drug delivery

enabled by a nicotine acetylcholine receptor ligand. Angewandte Chemie 123(24): 5596-

5599.

Zhang Q, Tang X, Zhang ZF, Velikina R, Shi S, Le AD (2007). Nicotine induces hypoxia-inducible

factor-1α expression in human lung cancer cells via nicotinic acetylcholine receptor–

mediated signaling pathways. Clin Cancer Res 13(16): 4686-4694.

Zhao Z, Reece EA (2005). Nicotine‐induced embryonic malformations mediated by apoptosis from

increasing intracellular calcium and oxidative stress. Birth Defects Res B 74(5): 383-391.

Zhu B, Heeschen C, Sievers RE, Karliner JS, Parmley WW, Glantz SA, et al. (2003). Second hand

smoke stimulates tumor angiogenesis and growth. Can Cell 4(3): 191-196.

Zimring JC, Kapp LM, Yamada M, Wess J, Kapp JA (2005). Regulation of CD8+ cytolytic T

lymphocyte differentiation by a cholinergic pathway. J Neuroimmunol 164(1): 66-75.

Zovko A, Viktorsson K, Lewensohn R, Kološa K, Filipič M, Xing H, et al. (2013). APS8, a

polymeric alkylpyridinium salt blocks α7 nAChR and induces apoptosis in non-small cell

lung carcinoma. Marine Drugs 11(7): 2574-2594.