Cytotoxic and Cytostatic Effect of Phenoxodiol On C6, HepG2, HT-29 and CNE1 Cancer Cell Lines -...

146
CYTOTOXIC AND CYTOSTATIC EFFECTS OF PHENOXODIOL ON C6, HEPG2, CNE1 AND HT-29 CANCER CELL LINES TEN YI YANG MASTERS OF MOLECULAR MEDICINE INTERNATIONAL MEDICAL UNIVERSITY MAY 2015

Transcript of Cytotoxic and Cytostatic Effect of Phenoxodiol On C6, HepG2, HT-29 and CNE1 Cancer Cell Lines -...

  • CYTOTOXIC AND CYTOSTATIC EFFECTS

    OF PHENOXODIOL ON C6, HEPG2, CNE1

    AND HT-29 CANCER CELL LINES

    TEN YI YANG

    MASTERS OF MOLECULAR MEDICINE

    INTERNATIONAL MEDICAL UNIVERSITY

    MAY 2015

  • DEDICATION

    To my mother,

    For her unrelenting belief, sacrifice and dedication;

    To my aunt,

    For her skepticism which fuels my motivation;

    To my high school biology teacher,

    For sparking a passion that still remains thus,

    And to my girlfriend,

    Whom provides a listening ear for when I rant.

  • i

    ABSTRACT

    Background: Genistein is a phytoestrogen flavonoids found in soy, legumes and

    other food products and is often lauded for its cytotoxic effect on various

    cancer cell types. The synthetic sterically modified derivative of genistein,

    phenoxodiol has been evaluated in phase II clinical trial in combination with

    cisplatin in treating chemo-resistant ovarian cancer. Mechanistic studies show

    that phenoxodiol disrupts the plasma membrane electron transport (PMET)

    system through the inhibition of surface tNOX protein leading to imbalanced

    NAD+ and NADH ratio. The imbalance deteriorates PMET and activates

    sphingomyelinase that generates the cytotoxic ceramide that leads to apoptotic

    pathways. This study investigates the cytotoxic and cytostatic activities of

    phenoxodiol on (a) C6, a rat glioma cell line; (b) HepG2, a human

    hepatocellular carcinoma cell line; (c) CNE1, a human highly differentiated

    nasopharyngeal carcinoma cell line and (d) HT-29, a human colorectal

    adenocarcinoma cell line. Methods used includes cell viability assay, cell cycle

    analysis, annexin V-propidium iodide apoptosis test and morphological analysis

    through ethidium bromide-acridine orange staining. Results: Results shows that

    apoptosis was induced by phenoxodiol for C6 and HepG2 cell lines only and

    the IC50 was found to be 1.4 g/mL and 2.0 g/mL for each cell line

    respectively. Cell cycle analysis shows that G1/S arrest started after 24 hours

    and increased within the next 48 hours.

  • ii

    Morphological analysis shows that the cells portraying typical apoptotic or

    necrotic signatures after the treatment of phenoxodiol. Conclusion:

    Phenoxodiol is shown to be a prominent anti-cancer agent in treating brain and

    liver cancer. Further studies should be carried out to confirm its effects in

    animals.

  • iii

    ACKNOWLEDGMENTS

    First and foremost, I would like to thank International Medical University for

    providing the research grant and facilities usage without which the project will

    not be able to proceed.

    I wish to express my most sincere gratitude to my supervisors, Dr. Fabian

    Davamani and Dr. Ho Ket Li whom both possesses remarkable qualities as a

    scientist that I wish to emulate. Their passion, guidance and discipline prove

    indispensable to my growth as a fledgling scientist. I am especially grateful to

    Dr. Ho for his devotion on assisting me on the project, particularly in

    troubleshooting errors and pointing out my mistakes.

    I am especially indebted to my fellow postgraduate colleagues, Yew Mei Yeng

    and Ng Pei Ying for providing help when I needed it the most especially during

    the work with flow cytometer. Ms. Yew and Ms. Ng both taught me on the

    operational use of the machine and provided the basis of my protocol on flow

    cytometer. A special shout out to Ms. Ng whom also provided extra reagents

    when mine ran out which allows the completion of my project.

  • iv

    I would like to extend my thanks to Dr Felicia Chung Fei Lei from the Institute

    for Research, Development and Innovation- International Medical University

    (IRDI-IMU) for kindly providing the cells used in this project.

    Sincere thanks to the laboratory staffs of the IMU research facilities particularly

    Ms. Malathi for assisting and guiding me during purchasing of project

    consumables and Ms. Yong Lee Mei whom provide assistance on flow

    cytometer and several others who kindly provided training for the lab

    equipment usage.

    And finally, I also place on record, my sense of gratitude to one and all, who

    directly or indirectly, have lent their hand in this venture.

  • v

    APPROVAL SHEETS

    I, the main supervisor to Ten Yi Yang hereby certify that the dissertation

    revisions have been made based on the recommendations by the Dissertation

    Examination Committee on 25thOf May 2015.

    _____________________________________

    Dr. Fabian Amalraj Davamani

    Lecturer

    School of Human Biology

    International Medical University

  • vi

    I certify that an Examination Committee has conducted the final examination of

    Ten Yi Yang on his name of degree dissertation entitled "Cytotoxic And

    Cytostatic Effects Of Phenoxodiol On C6, HepG2, CNE1 and HT-29 Cancer

    Cell Lines". The Committee recommended that the candidate be awarded the

    degree of Masters of Molecular Medicine.

    _______________________________

    Prof Chu Wan Loy

    Dean of School of Postgraduate Studies

    International Medical University

  • vii

    This Dissertation was submitted to the Senate of the International Medical

    University and was accepted by the Senate as having fulfilled the requirements

    for the degree of Masters of Molecular Medicine.

    _______________________________

    Prof Chu Wan Loy

    Dean of Postgraduate Studies and Research

    International Medical University

    Date:

  • viii

    DECLARATION

    I hereby declare that the dissertation is based on my original work except for

    quotations and citations which have been duly acknowledged. I also declare

    that it has not been previously or concurrently submitted for any other degree at

    the International Medical University or any other institution.

    _______________

    (Ten Yi Yang)

  • ix

    TABLE OF CONTENTS

    ABSTRACT ........................................................................................................ i

    ACKNOWLEDGMENTS ............................................................................... iii

    APPROVAL SHEETS ...................................................................................... v

    LIST OF FIGURES ........................................................................................ xii

    LIST OF ABBREVIATIONS ........................................................................ xv

    1 INTRODUCTION ..................................................................................... 1

    1.1 Background of study .......................................................................... 1

    1.2 Objectives of study ............................................................................. 3

    2 LITERATURE REVIEW ......................................................................... 4

    2.1 Cancer ................................................................................................. 4

    2.1.1 Glioma ........................................................................................... 4

    2.1.2 Hepatocellular carcinoma ............................................................. 6

    2.1.3 Nasopharyngeal carcinoma ........................................................... 8

    2.1.4 Colorectal carcinoma .................................................................... 9

    2.2 Phytoestrogens .................................................................................. 11

    2.2.1 Genistein ..................................................................................... 12

    2.3 Phenoxodiol ....................................................................................... 13

    2.3.1 Mechanism of action ................................................................... 14

    2.3.2 Other effects of phenoxodiol ...................................................... 18

    2.4 Apoptosis ........................................................................................... 19

    2.4.1 Apoptosis and cancer drug-discovery ......................................... 23

    2.5 Cell cycle ............................................................................................ 25

    2.5.1 Cell cycle regulation ................................................................... 27

    2.6 Ceramide ........................................................................................... 34

    2.6.1 Ceramide and apoptosis .............................................................. 35

    2.6.1.1 Ceramide and the extrinsic pathway ................................... 35

    2.6.1.2 Ceramide and the intrinsic pathway ................................... 36

    2.6.1.3 Ceramide induced apoptotic signals .................................... 38

    2.6.2 Ceramide and cell cycle .............................................................. 41

    3 MATERIALS AND METHODS ............................................................ 43

  • x

    3.1 Materials ........................................................................................... 43

    3.2 Methods ............................................................................................. 46

    3.2.1 Preparation of culture media and solutions ................................. 46

    3.2.1.1 DMEM .................................................................................... 46

    3.2.1.2 PBS .......................................................................................... 47

    3.2.1.3 Trypsin-EDTA ....................................................................... 47

    3.2.1.4 Phenoxodiol and Cycloheximide .......................................... 47

    3.2.1.5 MTT solution ......................................................................... 47

    3.2.2 Culture media for cell lines ......................................................... 48

    3.2.3 Maintaining and sub-culturing of cells ....................................... 49

    3.2.4 Cell seeding ................................................................................. 50

    3.2.5 Cell viability assay ...................................................................... 51

    3.2.6 Preparation of cell cycle analysis reagents ................................. 53

    3.2.6.1 Washing Solution 1 ................................................................ 53

    3.2.6.2 Washing Solution 2 ................................................................ 53

    3.2.6.3 Staining Solution ................................................................... 54

    3.2.7 Cell cycle analysis ....................................................................... 54

    3.2.8 Preparation of apoptosis test reagents ......................................... 56

    3.2.9 Annexin V-FITC apoptosis test .................................................. 56

    3.2.10 Morphological analysis by Acridine Orange (AO) and Ethidium

    Bromide (EB) dual staining ....................................................................... 58

    3.2.11 Statistical analysis ....................................................................... 59

    4 RESULTS ................................................................................................. 60

    4.1 Cell viability ...................................................................................... 60

    4.2 Flow cytometer cell cycle analysis ................................................... 63

    4.3 Annexin V-FITC apoptosis test ....................................................... 67

    4.4 EB-AO morphological analysis ....................................................... 70

    5 DISCUSSION ........................................................................................... 73

    6 CONCLUSION AND FUTURE DIRECTIONS ................................... 84

    REFERENCES ................................................................................................ 85

    APPENDIX .................................................................................................... 106

    Appendix 1: Troubleshooting ................................................................... 106

    Appendix 2: Flow cytometry data ............................................................ 121

  • xi

    LIST OF TABLES

    Table 2.1: Summary of the CDK and cyclin pair involved in the regulation of

    cell cycle phases. ............................................................................................... 28

    Table 3.1: List of chemicals used ..................................................................... 43

    Table 3.2: List of consumables used ................................................................ 44

    Table 3.3: List of apparatuses used .................................................................. 45

    Table 4.1: IC50 values of phenoxodiol treated against C6, HepG2, CNE1 and

    HT-29 cancer cell lines. .................................................................................... 61

    Table 5.1: Summary of IC50 obtained from previously reported cell lines tested

    with phenoxodiol (54). * indicates the IC50 obtained from our tested cell lines.

    ........................................................................................................................... 74

    APPENDIX

    Table A - 1: Comparison between IC50 values of phenoxodiol against

    cycloheximide on C6, HepG2, CNE1 and HT-29 cancer cell lines. ............... 114

  • xii

    LIST OF FIGURES

    Figure 2.1: Effects of phenoxodiol on apoptotic pathways and the cell cycle.

    Arrow in red represents inhibition while arrow in green represents stimulation

    ........................................................................................................................... 17

    Figure 2.2: A summary of the mammalian cell cycle with its respective

    checkpoints. Arrow in red represents inhibition and arrow in green represents

    stimulation while arrow in black indicates the approximate time point within

    the cell cycle that is regulated by its respective cyclin-CDK complex. ............ 33

    Figure 2.3: Ceramide production and metabolism pathway. ........................... 34

    Figure 4.1: Treatment of phenoxodiol on (a) C6; (b) HepG2; (c) CNE1; and (d)

    HT-29 cell lines for 24 and 48 hours. Cell viability was determined by MTT

    assay. * p< 0.05 compared to untreated cells .................................................... 62

    Figure 4.2: Cell cycle analysis of the effects of C6 cell line treated with 0

    g/mL and 1.4 g/mL phenoxodiol for (a) 24 and (b) 48 hours; (c) shows the

    cell cycle differences between 24 and 48 hours treated cells. * p < 0.05

    compared to control cells, ** p < 0.05 compared between treated cells time

    points. ................................................................................................................ 65

    Figure 4.3: Cell cycle analysis of the effects of HepG2 cell line treated with 0

    g/mL and 2.0 g/mL phenoxodiol for (a) 24 and (b) 48 hours; (c) shows the

    cell cycle differences between 24 and 48 hours treated cells. * p < 0.05

    compared to control cells, ** p < 0.05 compared between treated cells time

    points. ................................................................................................................ 66

    Figure 4.4: Annexin V Propidium Iodide flow cytometry analysis of C6 cells

    treated with 0 g/mL and 1.4 g/mL phenoxodiol for 24 and 48 hours. Cells are

    divided into four groups; viable, early apoptosis, late apoptosis and secondary

    necrosis. * p < 0.05 compared to control cells, ** p < 0.05 compared between

    treated cells time points. ................................................................................... 69

    Figure 4.5: Annexin V Propidium Iodide flow cytometry analysis of HepG2

    cells treated with 0 g/mL and 1.4 g/mL phenoxodiol for 24 and 48 hours.

    Cells are divided into four groups; viable, early apoptosis, late apoptosis and

    secondary necrosis. * p < 0.05 compared to control cells, ** p < 0.05 compared

    between treated cells time points. ..................................................................... 69

    Figure 4.6: Morphological analysis at 100x magnification of C6 cell line

    treated with 0 g/mL and 1.4 g/mL phenoxodiol for 24 hours and 48 before

    staining with 10 g/mL ethidium bromide and acridine orange. ...................... 71

    Figure 4.7: Morphological analysis at 100x magnification of HepG2 cell line

    treated with 0 g/mL and 2.0 g/mL phenoxodiol for 24 hours and 48 hours

    before staining with 10 g/mL ethidium bromide and acridine orange............ 72

  • xiii

    APPENDIX

    Figure A - 1: Determination of minimum inhibitory concentration of DMSO

    for 24 and 48 hours on (a) C6; (b) HepG2; (c) CNE1 and (d) HT-29 cell lines.

    ......................................................................................................................... 108

    Figure A - 2: Determination of optimum seeding density for C6, HepG2,

    CNE1 and HT-29 cell lines for (a) 24 and (b) 48 hours ................................. 110

    Figure A - 3: Treatment of cycloheximide on (a) C6; (b) HepG2; (c) CNE1;

    and (d) HT-29 cell lines for 24 and 48 hours. Cell viability were determined by

    MTT assay. ..................................................................................................... 113

    Figure A - 4: Unstained HepG2 cells as example in used for compensation

    setup for flow cytometry apoptosis test. Cell population was adjusted so that it

    falls within the lower left quadrant which does not contain either stain readings.

    ......................................................................................................................... 117

    Figure A - 5: Annexin-V stained HepG2 cells as example in used for

    compensation setup for flow cytometry apoptosis test. Cell population was

    adjusted so that it falls within the lower right quadrant which contains only

    Annexin V stain readings. ............................................................................... 118

    Figure A - 6: Propidium iodide stained HepG2 cells as example in used for

    compensation setup for flow cytometry apoptosis test. Cell population was

    adjusted so that it falls within the upper right quadrant which contains only

    propidium iodide stain readings. ..................................................................... 119

    Figure A - 7:Annexin V and propidium iodide stained HepG2 cells as example

    in used for compensation setup for flow cytometry apoptosis test. Cell

    population were seen in the upper right quadrant containing both stains reading

    on successful compensation setup................................................................... 120

    Figure A - 8: Cell cycle profile obtained during FACS analysis of C6 cell line

    treated with (a) 0 g/mL and (b) 1.4 g/mL phenoxodiol for 24 hours by

    plotting cell count against DNA concentration. .............................................. 121

    Figure A - 9: Cell cycle profile obtained during FACS analysis of the effects of

    C6 cell line treated with (a) 0 g/mL and (b) 1.4 g/mL phenoxodiol for 48

    hours by plotting cell count against DNA concentration. ............................... 122

    Figure A - 10: Cell cycle profile obtained during FACS analysis of the effects

    of HepG2 cell line treated with (a) 0 g/mL and (b) 2.0 g/mL phenoxodiol for

    24 hours by plotting cell count against DNA concentration. .......................... 122

    Figure A - 11: Cell cycle profile obtained during FACS analysis of the effects

    of HepG2 cell line treated with (a) 0 g/mL and (b) 2.0 g/mL phenoxodiol for

    48 hours by plotting cell count against DNA concentration. .......................... 123

    Figure A - 12: Dot plot representation of Annexin V-PI stained C6 cell line

    treated with (a) 0 g/mL and (b) 1.4 g/mL phenoxodiol for 24 hours. ........ 124

    Figure A - 13: Dot plot representation of Annexin V-PI stained C6 cell line

    treated with (a) 0 g/mL and (b) 1.4 g/mL phenoxodiol for 48 hours. ........ 125

  • xiv

    Figure A - 14: Dot plot representation of Annexin V-PI stained HepG2 cell

    line treated with (a) 0 g/mL and (b) 2.0 g/mL phenoxodiol for 24 hours. . 126

    Figure A - 15: Dot plot representation of Annexin V-PI stained HepG2 cell

    line treated with (a) 0 g/mL and (b) 2.0 g/mL phenoxodiol for 48 hours. . 127

  • xv

    LIST OF ABBREVIATIONS

    AIF Apoptosis inducing factor

    AO Acridine orange

    APC Anaphase-promoting complex

    APAF Apoptotic protease activation factor

    ATP Adenosine triphosphate

    ATR Ataxia telangiectasia and Rad3-related protein

    BAX Bcl-2-associated X protein

    CAD Caspase-activated DNAse

    CDK Cyclin-dependent kinases

    cFLIP Cellular FLICE-like inhibitory protein

    CTLA-4 Cytotoxic T-lymphocyte-associated protein 4

    CXC C-X-C motif chemokine

    DED Death effector domain

    DISC Death inducing signaling complex

    DMEM Dulbeccos Modified Eagle Medium

    DMSO Dimethyl sulfoxide

    DNA Deoxyribonucleic acid

    EB Ethidium bromide

    EDTA Ethylenediaminetetraacetic acid

    ENOX Ecto-NOX

    FADD Fas-associated death domain

    FLICE FADD-like IL-1-converting enzyme

    GCS Glucosylceramide synthase

    IAP Inhibitors of apoptosis

    JNK c-Jun N-terminal kinase

    MAP Mitogen-activated protein

    MAPK Mitogen-activated protein kinase

  • xvi

    MOMP Mitochondrial outer membrane permeabilization

    MPF M phase-promoting factor

    MPT Mitochondrial permeability transition

    MTT Thiazolyl Blue Tetrazolium Blue

    NADH Nicotinamide adenine dinucleotide

    NF- Nuclear factor

    NOX NADH-oxidase

    NPC Nasopharyngeal carcinoma

    PBS Phosphate buffer saline

    PIDD p53-induced protein with a death domain

    PI3K Phosphoinositide 3-kinase

    PIP2 Phosphatidylinositol-4,5-biphosphate

    PIP3 Phosphatidylinositol3,4,5trisphosphate

    PKC Protein kinase C

    PMET Plasma membrane electron transport

    PP2A Protein phosphatase 2A

    PPAR Peroxisome proliferator-activated receptor

    PTK Protein tyrosine kinase

    RAIDD Rip-associated protein death domain

    TNF Tumor necrosis factor

    TRADD TNFR-associated death domain

    TRAIL Tumor necrosis factor-related apoptosis-inducing ligand

    TNOX Tumor-associated NADH oxidase

    VEGF Vascular endothelial growth factor

    XIAP X-linked inhibitor of apoptosis protein

  • 1

    1 INTRODUCTION

    1.1 Background of study

    Theres a disturbing continued rising trend of global burden of cancer largely

    due to the increased adoption of unhealthy cancer-causing behaviors. In this

    project, focus will be made on four types of cancers which are: brain and

    nervous system cancer, liver cancer, nasopharyngeal cancer and colorectal

    cancer all of which are featured quite prominently in Malaysia. According to

    the latest cancer statistics in Malaysia, brain and nervous system cancer is

    ranked ninth of the most frequently diagnosed cancer type in males while liver,

    nasopharyngeal and colorectal cancer is ranked tenth, fourth and second most

    frequently diagnosed cancer types in both gender respectively.

    Phenoxodiol is a synthetic sterically modified genistein derivative, a soy

    isoflavone with antitumor effects on various cancer types such as breast,

    prostate, lung, liver and gastric cancers. Studies found phenoxodiol to be

    efficient over its parent compound and has been explored as a possible curative

    against chemo-resistant ovarian cancer when used in combination with cisplatin

    or paclitaxel in a phase II clinical trials. At the moment, phenoxodiol is

    currently also being evaluated for its side effect when combined with docetaxel

    in a phase I/II trial.

  • 2

    Phenoxodiol is shown in mechanistic studies to disrupt the plasma membrane

    electron transport system leading to imbalanced NAD+ / NADH ratio triggering

    sphingomyelinase activation that converts plasma membrane sphingomyelin to

    ceramide, an anti-apoptotic agent.

    Recent literature suggests that cytotoxicity and cytostaticity effects of

    phenoxodiol have been studied extensively in vitro in several cancer cell lines

    including ovarian, prostate, breast and leukemia. However, its effects on brain

    glioma, hepatocarcinoma, nasopharyngeal carcinoma and colorectal cancer

    have not been characterized. We hypothesized that phenoxodiol has a high

    potential in inhibiting these cancer types because the molecular target of

    phenoxodiol, the plasma membrane tumor specific NADH oxidase is associated

    with a majority of cancer types. Besides, genistein (the parent compound of

    phenoxodiol) has also been proven to work on ovary, brain, liver,

    nasopharyngeal and colon cancer. Hence, it is thought that phenoxodiol will

    also be effective against these cancer types with similar mechanism.

  • 3

    In this study, the antitumor effect of phenoxodiol on HepG2 (Homo sapiens

    hepatocellular carcinoma), HT-29 (Homo sapiens colorectal adenocarcinoma),

    CNE1 (Homo sapiens highly differentiated nasopharyngeal carcinoma) and C6

    (Rattus norvegicus glioma) were tested via cell viability assay and cell cycle

    analysis to evaluate the cytotoxic and cytostatic effect of phenoxodiol.

    Apoptosis test and morphological analysis with ethidium bromide and acridine

    orange dual staining were used to assess the apoptotic potential of phenoxodiol.

    1.2 Objectives of study

    The objective of this study is to:

    1. determine the inhibitory effect of phenoxodiol on the viability of C6,

    HepG2, CNE1 and HT-29 cancer cell lines.

    2. evaluate the effect of phenoxodiol on the cell cycle of potential cell

    lines.

    3. determine the apoptotic effect of phenoxodiol on potential cell lines.

  • 4

    2 LITERATURE REVIEW

    2.1 Cancer

    Cancer, a disease exclusive to multicellular organism, is defined as the aberrant

    cellular growth caused by disturbed expression in the genetic level leading to

    dysregulation in cellular division and differentiation leading to imbalance in the

    cell replication and death ratio which sees favorable a growth in the cancer cell

    population (1). Cancer is a common disease worldwide and Malaysia is no

    exception, where it was ranked third as the most common mortality cause after

    pulmonary disease and septicemia.

    2.1.1 Glioma

    Gliomas arise from glial cells in the central nervous system encompassing the

    spinal cord and the brain but have a higher occurrence in the latter. In Malaysia,

    it is the tenth most frequent cancer types in males with a total of 259 cases

    reported out of a total of 8123 cases and unlisted in females (2). Gliomas has a

    potential to arise from all types of glial cells however, glioblastomas arising

    from astrocytes is of the majority of malignant glioma accounting for 82% of

    cases (3). Malignant gliomas, although highly aggressive, does not metastasize

    and is largely confined to central nervous system (4).

  • 5

    Even so, the prognosis for malignant gliomas remains poor with estimated

    patient survival duration between 12 to 18 months with the best treatment (5).

    Overall, the 5-year relative survival rate is estimated to be at 34% (6).

    Progressive genetic destabilization and changes from either naturally occurring

    or various environmental factors contributes to the development of malignant

    gliomas. Gender-wise, men are more susceptible than women and white

    populations are more effected than black populations (7). A number of pre-

    existing rare heredity syndromes such as Turcot, Cowden, type 1 and 2

    neurofibromatosis, Li-Fraumeni, familial schwannomatosis and tuberous

    sclerosis may increase risk of glioma development as well (8). Genome wide

    association studies shows that polymorphic variants in the RTEL, TERT,

    CDKN2BAS, EGFR, CCDC26 and PHLDB1 genes are associated with glioma

    development albeit weakly suggesting the presence of potential multiple

    molecular subsets (9).

    Surgical resection is usually indicated with the aim of alleviating tumor mass

    effect and the extent of surgical intervention is well evidenced to affect patient

    survivability (10). Post-surgery, adjuvant radio and chemotherapy is prescribed

    with intensity-modulated radiotherapy (11) and alkylating agents (12) as the

    preferred methods respectively.

  • 6

    Other treatments being explored includes the monoclonal antibody ipilimumab

    and bevacizumab which inhibits the immune system downregulator CTLA-4

    and VEGF signalling involved in angiogenesis respectively with ipilimumab

    showing enhanced patient survivability in randomized trials (13). Recent

    discovery of specific cytomegalovirus antigens in glioblastoma multiforme

    allows the development of adoptive immunotherapy as new treatment option

    (14).

    2.1.2 Hepatocellular carcinoma

    Liver cancer originates from hepatocytes and it is associated primarily with

    hepatitis B or C viral infection (15). Following viral infection, the

    hepatocellular innate immune pathways is activatedby the release of CXCL10

    chemokine which leads to the recruitment of inflammation-causing immune

    effector cells designed to eliminate viral particles through binding and

    activation of CXCR3 receptor found on these cells (16). However, inadequate

    viral elimination in up to 85% of patients with acute viral infection leads to a

    persistent presence of proinflammatory immune cells in the liver (17) and

    subsequently causes nearby tissue destruction that links to hepatocarcinoma and

    various liver diseases. Other causes are through chronic hepatic inflammation

    due to various non-viral causes.

  • 7

    Malaysia cancer statistics shows liver cancer as the tenth most frequent cancer

    type diagnosed, contributing to 605 cases or 3.3% of total cancer cases reported

    in Malaysia and of all the cases, 443 cases are reported in men (2).

    Conventional curative treatment type for liver cancer is effective with up to

    75% 5-year survival rates (18) which includes surgical resection of liver, liver

    transplantation and local ablation therapy using chemical or thermal ablation

    methods. Even though the rate of effectiveness is promising, the low eligibility

    of patients (less than 20%) due to reasons such as donor shortage, dysfunctional

    liver or advanced hepatocarcinoma stage means the majority of liver cancer

    patients are only able to opt for palliative or symptomatic treatment with a

    much lower survival rate and duration (19). New treatment options are being

    explored for the treatment of hepatocarcinoma such as oncolytic viral therapy

    with a genetically modified poxvirus JX-594 that shows promising result in

    phase I and II clinical trials (20,21).

  • 8

    2.1.3 Nasopharyngeal carcinoma

    Nasopharyngeal carcinoma (NPC) originates from the nasopharynx which is

    the top part of pharynx lying just behind the nose. In Malaysia, there is high

    risk of nasopharyngeal cancer particularly in Chinese and Malay population,

    contributing to a total of 940 cases and 5.2% of the overall cancer cases in

    Malaysia (2) and of the total cases, 685 cases are male patients.

    Genetics and environmental factors may play a role in influencing the unique

    geographical incidence pattern of NPC. Probable environmental carcinogens

    has been found to cause the loss of alleles on chromosome 3 and 9 short arms

    which leads to inactivation of tumor suppressor genes such as p14, p15 and p16

    (22).The exact carcinogens in question is yet to be pinpoint but it is speculated

    that salted consumables particularly Chinese salted fish leads to NPC

    development (23).

    Local control of nasopharyngeal carcinoma using surgical method is not viable

    as it is highly prone to metastasis to the cervical lymph node partly due to its

    anatomical spot (24). Hence, NPC is mainly treated using radiotherapy.

    Technological advances improve the conventional two-dimensional techniques

    used to deliver radical radiotherapy.

  • 9

    When compared with two-dimensional methods, techniques such as three-

    dimensional conformal and intensity-modulated radiotherapy are reported to be

    superior in treating nasopharyngeal carcinoma (25). Furthermore, emerging

    new techniques such as Tomotherapy and RapidArc radiotherapy improves the

    dosimetric efficacy in late-stage NPC treatment (26). Radiotherapy in NPC

    treatment may be effective in controlling the primary tumor (27), however, a

    regimen of chemotherapy such as Cisplatinin combination with other agents

    such as 5-flurouracil or with docetaxel and capecitabine is still recommended to

    be prescribed together as radiotherapy alone does not inhibit lymph node

    metastasis of NPC to distant sites (28).

    2.1.4 Colorectal carcinoma

    Colorectal cancer is the formation of cancerous tumor in parts of the large

    intestine like the colon or the rectum (29). The risk of colorectal development is

    approximately 5.0% in men and slightly lower (4.7%) in women with

    diagnosed men having 30% to 40% higher mortality rate than women (30).

    Progressively higher incidence is contributed by risk factor such as age, gender

    and ethnicity (31). The exact correlation between incidence and risk factors are

    yet to known but it is speculated that complex sex hormones interactionsand

    socioeconomic status may play a role (32).

  • 10

    There is a high colorectal cancer incidence reported in Malaysia, being the

    second most frequent cancer types in both genders (2) contributing 2246 cases

    and 12.3% of all cancer cases being diagnosed in the most recent report

    published . Additionally more males (1185 cases) are being diagnosed with

    colorectal cancer than females (1011 cases).

    Prognosis for localized colorectal cancers is good with a 90.3% 5-year relative

    survival rate which declines to 12.5% if the cancer metastasized to other organs

    (6). Treatment option for colorectal cancer varies depending on the stage and

    location of cancer. Mainstay treatment for colorectal cancer is the surgical

    resection of tumour supplemented with chemotherapy treatments such as 5-

    flurorouracil and leucovorin combination that significantly improves

    survivability in a phase III trial in treatment of end-stage colorectal cancer (33)

    or radiotherapy.

  • 11

    2.2 Phytoestrogens

    Phytoestrogens are biochemically heterocyclic phenols which are a type of

    flavonoids with a similar structure and or function to endogenous mammalian

    oestrogens hence they exhibit either a similar activity to oestrogens or a weak

    antioestrogen-like property (34) and can be further classified into flavones,

    isoflavones, coumestans and ligands (35). The isoflavone genistein is one of

    such phytoestrogen. Found predominantly from soy and legumes food products

    (36), genistein is metabolized into genistein deriatives such as dihydrogenistein

    or 6-hydroxy-O-desmethylangolensin by the gut microflora through

    conjugation with glycoside that subsequently exerts oestrogenic and

    antioxidative effect (37).

  • 12

    2.2.1 Genistein

    Genistein is often lauded for its anti-cancer properties particularly in prostate

    cancer (38). Although like other flavonoids, genistein also possesses many

    health benefits as mentioned previously. Genistein is found to cause

    perturbation of cell cycle progression mostly in the G2/M phase leading to

    disrupted cancer development in several cancer types such as breast, prostate,

    lung, liver and gastric cancers (39-42). Mechanistic studies shows genistein

    inhibits protein tyrosine kinase (PTK) (43) which is deregulated in cancer cells

    leading to consecutive activation of PTK-mediated signalling pathways leading

    to uncontrolled cellular growth and proliferation (44). Sakla et al. also reported

    similar tyrosine kinase inhibition action through ER-dependent mechanism

    leading to down regulated expression of HER2 protein (45), a growth factor

    signal regulator found overexpressed in aggressive and chemoresistant breast

    cancer(46) and involved in tumorigenesis (47). Furthermore, genistein is found

    to supress the transcription factor nuclear factor (NF-) (48) which is found

    consecutively activated in many tumor types which causes expression of anti-

    apoptotic oncogenes (49). Genistein is also demonstrated to inhibit the enzyme

    topoisomerase I and II (50,51), 5- reductase (52) and the signal transductor

    protein histidine kinase (53) all of which contributed to the anti-cancer

    properties of genistein.

  • 13

    2.3 Phenoxodiol

    Phenoxodiol (2H-1-benzopyron-7-0,3-(hydroxy phenyl)) is a synthetic steric-

    modified derivative of genistein and it is found that phenoxodiol offers

    substantially improved bio-availability, lowered metabolism rate and antitumor

    potency over its parent compound genistein (54).

    The underlying foundation behind phenoxodiol development is as a chemo-

    sensitizer where it is found to reincur sensitivity in late stage tumor cells

    resistant to docetaxel and platinum-based drugs (55). Phenoxodiol is also being

    explored as a monotherapy where in vitro studies shows phenoxodiol

    effectively inhibits ovarian cancer cells and has progressed to Phase III clinical

    trial conducted by Marshall Edwards, Inc (now MEI Pharma) as orally

    administered phenoxodiol in recurrent ovarian cancer patients. However there

    is no statistically significant improvement was observed in primary or

    secondary endpoint (56). A revised study has been conducted for the evaluation

    of phenoxodiol against chemo-resistance ovarian cancer in combination with

    cisplatin or paclitaxel is currently in Phase II clinical trial which shows that the

    combination of cisplatin and phenoxodiol was well tolerated and effective

    against chemo-resistant ovarian cancer (57). Currently, phenoxodiol is

    evaluated together with docetaxel for side effects in a phase I/II trial for

    treatment against advanced stage recurrent ovarian, fallopian tube or

    gastrointestinal carcinoma (58).

  • 14

    Because phenoxodiol is an analogue of genistein, phenoxodiol shares similar

    cytotoxic mechanism. Likewise, phenoxodiol is found to disrupt the cell cycle

    and causes checkpoint arrest, however unlike genistein which found to induces

    G2/M phase arrest and G1/S phase arrest in murine fibroblast and melanoma

    cells (59), phenoxodiol is found to promote G1/S phase arrest in various cancer

    cell types (60,61). Mechanistic studies characterize the anti-tumor properties

    exerted by phenoxodiol on various cancer cell lines. It has been shown that

    phenoxodiol induces the caspase-dependent apoptotic pathway in prostate and

    ovarian cancer cell lines through degradation of XIAP and cFLIP anti-apoptotic

    proteins (62,63) of which are triggered through ceramide accumulation that

    leads to downstream cascade of events.

    2.3.1 Mechanism of action

    The hydrophobic characteristics of phenoxodiol cause its tendency to partition

    within the cellular membrane. Cellular membranous systems such as the

    mitochondrial membrane contain electron transport system that generates

    membrane potential which drives the production of ATP through oxidative

    phosphorylation. The electron shuttler, ubiqinone is involved in mediating

    redox cycling in the mitochondrial electron transport chain through the

    shuttling of electrons between complexes. Mounting evidence suggested the

    presence of a similar system in the plasma membrane (64) involved in the

    production of glycolytic ATP through oxidation of cytosolic NADH (65).

  • 15

    This suggested that lipophilic compounds like phenoxodiol may act primarily

    on the plasma membrane electron transport (PMET) leading to cellular redox

    imbalance.

    The perturbation of intracellular redox homeostasis such as the ratio of

    NADH/NAD+, balance of glutathione and ubiquinone content and state of

    redox affects cellular function such as viability and proliferation (66) thus

    ultimately leads to apoptosis (67). Phenoxodiol was reported to have a high

    affinity to purified recombinant cell surface NADH-oxidase (NOX) leading to

    truncated hydroquinone oxidation and the catalyzation of interchanging activity

    between protein disulfide and thiol (68). The cell surface ECTO-NOX protein,

    designated as tNOX due to its tumor-specificity, upon inhibition by

    phenoxodiol causes inhibition of growth followed by apoptosis in transgenic

    mice embryonic fibroblasts expressing tNOX gene but not in wild-type mice.

    Cytotoxic specificity of phenoxodiol on tumor cells may be accounted by its

    ineffectiveness on the non-tumor specific consecutive form of enzyme in

    question (CNOX) (68). The correlation between the chemosensitizing effect of

    phenoxodiol and tNOX was also explored extensively and was demonstrated to

    improve cisplatin and paclitaxel sensitization of platinum resistant HeLa cells

    tNOX activity and proliferation (69). More interestingly, tNOX which was

    previously demonstrated to have prion like properties (70) imparts phenoxodiol

    chemosensitized conformation to subsequent tNOX molecules in the absence of

    phenoxodiol (69).

  • 16

    Inhibition of tNOX by phenoxodiol causes an imbalanced ratio of NAD+ and

    NADH which subsequently leads to deterioration of PMET. The accumulation

    of excessive cytosolic NADH activates the plasma membrane

    sphingomyelinase leading to ceramide generation from metabolic hydrolysis of

    complex sphingolipids like sphingomyelin and cerebrosides (71) and

    concurrently decreases sphingosine-1-phosphate levels from the inhibition of

    sphingosine kinase at which both events contributed to potential G1 arrest and

    subsequent apoptosis (72). A summary of the effects of phenoxodiol on

    apoptotic pathways and cell cycle is seen in Figure 2.1. Furthermore, the

    reduced coenzyme Q10 level releases sphingomyelinase inhibition (73). To

    further support the statement, sphingosine kinase 1 activity in osteosarcoma cell

    line is shown to be synergistically reduced when treated with phenoxodiol and

    doxorubicin which subsequently increases cellular ceramide level and

    triggering cell death (74) plus, phenoxodiol is reported to increase ceramide

    level by around 2.3-fold in multidrug resistant tumor cell lines (75).

  • 17

    Figure 2.1: Effects of phenoxodiol on apoptotic pathways and the cell cycle.

    Arrow in red represents inhibition while arrow in green represents stimulation

  • 18

    2.3.2 Other effects of phenoxodiol

    Anti-tumor effects of phenoxodiol may not be limited to directly exerting

    cytolytic effects via induction of apoptosis on cancer cells. Phenoxodiol is

    reported to exhibit anti-angiogenic effect by inhibiting proliferation and

    migration of endothelial cells while simultaneously reduces the formation of

    capillary tube and decreases matrix metalloproteinase 2 expression (76). A

    similar observation was found in endothelial cells treated with exogenous

    ceramide analogs which its molecular mechanism was attributed to the

    decreased cyclin D1 expression through upregulation of CAV-1 expression

    which represses cyclin D1 promoter activity (77) and inhibition of ERK1/2

    phosphorylation (78).

    Phenoxodiol is also shown to improve immunomodulation through

    enhancement of natural killer cells lytic function (79). This phenomenon can be

    attributed to ceramide triggering of prosurvival NF- pathway through

    activation of calpain which cleaves the NF- inhibitor p105 (80) that

    suggested a negative feedback regulation of ceramide. The activation of NF-

    pathway not only leads to transcriptional activation of various prosurvival

    oncogenes but also genes related to innate and adaptive immune response (81).

  • 19

    Phenoxodiol is found to inhibit the catalytic activity DNA topoisomerase II, a

    key enzyme in regulating untangling of over-wounded DNA through

    stabilization of the cleavable complex (82). The interaction between ceramide

    and DNA topoisomerase II has not been elucidated, however, ceramide 1-

    sulfates 1 and 2 isolated from Japanese Bryozoa Watersipora cucullata is found

    to be a potent inhibitor of DNA topoisomerase I (83).

    2.4 Apoptosis

    Apoptosis or programmed cell death is a term used to characterize a type of

    distinct cell death with a specific morphology and biochemical processes.

    Apoptosis is a natural occurrence in cell senescence and is essential in tissue

    cell population homeostatic maintenance. However, apoptosis can also be

    induced by external stimuli such as immune reactions or cellular stress as a

    defensive mechanism (84).

    Apoptosis can be activated through two pathways, the intrinsic and the extrinsic

    pathways which interlinked with one another with molecules from one pathway

    influencing the other (85). A more unconventional pathway involves T-cells

    activation of perforin-granzyme for triggering cell death. These pathways are

    known as the caspase-dependent pathway as it involves the endoprotease

    caspase responsible for the hydrolysis of peptide bonds at aspartic acid residues

    during cell disassembly into apoptotic bodies (86).

  • 20

    The extrinsic pathway involves binding and activation of TNF superfamily of

    cell death receptor like Fas and tumor necrosis factor (TNF)-1 by its respective

    death ligand, the Fas ligand (FasL) and TNF-2 on the plasma membrane

    resulting in trimerization and death effector domain (DED) clustering of

    receptor in addition to recruitment of adapter proteins like Fas-associated death

    domain (FADD) or TNFR-associated death domain (TRADD) (87) which in

    turns recruits procaspase-8 monomeric protein to form death inducing signaling

    complex (DISC) (88). The self-cleavage of oligermerized procaspase-8 within

    DISC activates caspase-8. Caspase-8 activation then drives various downstream

    procaspases which varies according to cell types. Type I cells which consists of

    several lymphoid cell lines are able to directly activate downstream procaspases

    such as procaspase-3 as caspase-8 is sufficiently activated. In other cell types,

    known as type II cells, the weakly activated caspase-8 are unable to directly

    activate procaspase-3 but able to activate mitochondrion-mediated pathway

    through the truncation activation of the proapoptotic protein Bid into tBid

    which causes the release of apoptotic molecules such as cytochrome c and

    apoptosis-inducing factor from the mitrochondria (89). Procaspase-8 can also

    be activated independent of neither FADD interaction nor DISC formation

    through cytochrome c-dependent pathway where the release of cytochrome c

    triggers the activation of caspase-6 which in turns activates procaspase-8 (90).

  • 21

    A lesser known procaspase-10 mediated extrinsic pathway is similar to that of

    procaspase-8 and responsible mainly for lymphoid cells apoptosis. In Fas- and

    TNF- death ligand-receptor apoptosis, caspase-10 is shown to function

    independently from caspase-8 from the occurrence of apoptosis in capsase-10-

    overexpressed but caspase-8 deficit cells (91). Caspase-10 is also found to

    cleave substrate differently than of caspase-8 (92) which may indicates each

    possesses a unique role in initiation of apoptosis. Caspase-10 was also much

    more frequently under-expressed than caspase-8 in several carcinoma cell lines

    which may suggest a role of caspase-10 in cancer onset (93).

    The highly conserved rudimentary caspase-2 dependent extrinsic pathway also

    functions similarly to that of caspase-8. Likewise, following the binding and

    activation of death ligand and corresponding receptors from apoptotic stimuli

    (94), the recruitment of Rip-associated protein death domain (RAIDD) binds

    and oligomerizes with p53-induced protein with a death domain (PIDD) to

    form PIDDosome complex (95) followed by self-cleavage activation of

    procaspase-2 monomers. It is speculated that the active caspase-2 then cleaves

    the proapoptotic Bid causing membrane permeabilization of mitochondria

    leading to release of proapoptotic proteins (96) which is further elaborated

    below.

  • 22

    The intrinsic pathway or the mitochondrion-mediated pathway is activated in

    the presence of non-receptor stimuli such as cellular stress or absence of certain

    biochemicals leads to mitochondrial inner membrane changes resulting in

    mitochondrial permeability transition (MPT) pore opening allowing the release

    of the pro-apoptotic proteins cytochrome c, Smac/DIABLO and HtrA2/Omi

    serine proteases (97). The release of cytochrome c activates cytosolic caspase-6

    and apoptotic protease activation factor (Apaf)-1 and procaspase-9 leads to the

    formation of apoptosome (98) which in turns activates procaspase-3 and 7. A

    positive feedback pathway occurs between the activated caspase-3 and

    procaspase-9. Caspase activity is also enhanced through the inhibition of

    inhibitor of apoptosis proteins by Smac/DIABLO and HtrA2/Omi (99). In late

    apoptosis, apoptosis inducing factor (AIF), endonuclease G and caspase-

    activated DNAse (CAD) are released from the mitochondria. All of which

    translocates to nucleus causing fragmentation of DNA where AIF and

    endonuclease G performs in a caspase-independent fashion (100) but a

    cleavage by caspase-3 is required for the activation of CAD (101). The

    apoptotic events described are tightly regulated by the Bcl-2 protein family

    (102) which in turn regulated by the tumor suppressor protein p53 (103).

  • 23

    Caspase-2, -8, -9 and -10 is known as the initiator apoptotic caspases, as they

    activate several apoptosis executioner caspases such as caspase-3, -6 and -7

    (104). The executioner caspases, upon activation cleaves several vital proteins

    such as DNA repair proteins such as poly ADP ribose polymerase (PARP),

    DNA-dependent protein kinase (DNA-PK) and U1-70kD and subsequently

    leads to DNA degradation, lamin A and fodrin which are found in nuclear and

    cytosolic skeleton causing chromatins condensation and nuclear membrane

    decomposition and eventually results in the formation of apoptotic body (105).

    2.4.1 Apoptosis and cancer drug-discovery

    In cancer drug discovery, restoration of the apoptotic pathway is an effective

    method in treating cancer. This is because tumor cells are under constant stress

    and marked for removal but sustained due to aberration in apoptotic pathways.

    In tumor cells, apoptosis is evaded through manipulation of the Fas-mediated

    apoptosis. Down-regulation of Fas expression or deregulation of key

    components in Fas-mediated apoptotic pathway (106) is a common tumor

    hallmark in several tumor cell lines.

  • 24

    Additionally, some carcinomas such as brain (107) and ovarian (108) cancers

    were found to overexpressed FasL. This seemingly counter-intuitive way is

    actually a self-defensive mechanism employed by tumor cells to induce

    immune privilege in tumor site not unlike specialized organs such as the brain,

    testes and eyes. FasL expression in tumor cells allows the crosslinking of Fas

    receptor expressed on the surface of tumor invading cytotoxic T-cells that

    subsequently leads to apoptosis of the T-cells (109). The simultaneous down-

    regulation and up-regulation of Fas and FasL in tumor cells prevents apoptosis

    and invoking immune response.

    A drug-induced apoptotic mode of death is much more preferred over necrosis

    as apoptosis does not induce inflammatory response. At a necrotic site,

    leukocytes consisting of neutrophils will infiltrate the site rapidly which is

    followed subsequently by monocytes accumulation (110) which will cause

    further destruction of normal tissues surrounding the necrotic site and

    subsequent fibrosis. In an in vivo condition, apoptotic cells maintain membrane

    integrity and do not release proinflammatory cytokines. Before the cells

    disintegrate, adjacent phagocytes engulf the cells thus preventing the lyses of

    apoptotic cells. Furthermore, production of inflammatory meditators IL-10 or

    TGF- that inhibits inflammation by macrophages can be stimulated by

    apoptotic cells (111,112). However, this may not always be the case for

    apoptotic cells as there are reports of stimulated apoptotic cells causing intense

    inflammation in mice (113).

  • 25

    The unpredictable nature of apoptosis can be attributed to the clearance rate by

    phagocytes as over time, if apoptotic cells are unable to be ingested by

    phagocytes in time, they undergoes secondary necrosis where the membrane

    became macromolecules permeable (114) and thus causing the release of

    intracellular contents with part of it being proinflammatory cytokines which

    triggers host inflammatory responses. Secondary process will also occurs in

    vitro where under the absence of phagocytes, apoptotic cells will ultimately

    swell and lyses, a phenomenon that may occur with phenoxodiol treated cells at

    longer time.

    2.5 Cell cycle

    The most basic nature of a cell is growth and proliferation which is an

    important process in tissue and organ development and also repair and replace

    cells loss due to injury. When cells undergo division, two consecutive processes

    ultimately happen, which are the replication of DNA and chromosomal

    segregration into daughter cells that can be subdivided further in various stages

    collectively known as the cell cycle. A cell cycle is consists of four different

    stages, G1, S, G2 and M which morphologically can be divided into interphase

    (G1, S, G2) and mitosis (M) phase that consist of prophase, metaphase,

    anaphase and telophase.

  • 26

    In G1 phase, the cells prepares for DNA synthesis through increased production

    of mRNA and proteins involved in DNA replication. . It is found that some

    cells like Xenopus embryos (115) and cancer cells (116) are able to bypass G2

    phase completely and enters mitotic phase after the replication of DNA. In the

    M phase, chromosomal condensation occurs by condensing the replicated

    DNA, packaged in elongated chromosomal form into a more compacted form

    for segregation. Subsequently, breakdown of the nuclear envelope leads to

    attachment of sister chromatids to the microtubules of mitotic spindles where it

    will be aligned at the equator during metaphase. In anaphase, separation of

    sister chromatids to opposite pole of spindle occurs where decondensation and

    intact nuclei is reformed. Finally, cytokinesis occurs where the cells are divided

    through cytoplasmic division (117). An additional phase, known as G0 phase is

    used to describe stagnant cells that are not actively dividing but with potential

    for division (115) which consist of the majority of non-proliferating cells in our

    body.

  • 27

    2.5.1 Cell cycle regulation

    Cell cycle phase transition is regulated by a family of serine/threonine protein

    kinases known as the cyclin-dependent kinases (CDK) which is activated at

    various specific points within the cell cycle. There are nine identified CDKs

    and five are activated in cell cycle. CDK2, 4 and 6 is activated during G1 phase,

    CDK2 in S phase and CDK1 that is activated in both G2 and M phase. All

    CDKs is activated by CDK7 in combination with cyclin H known as CDK

    activating kinase. The full activation of CDK activity besides binding of cyclin

    requires phosphorylation at threonine and tyrosine residue by CDK activation

    kinase which induces conformational changes that enhances cyclin binding

    (118).The role of the remaining CDK in cell cycle progression is yet to be

    determined (119). The activation of CDKs requires their interaction with cyclin

    to phosphorylate the downstream proteins (120) that allows the progression of

    cell cycle.

    As CDK is regulated by cyclin, the level of CDK remains constant in contrast

    to cyclin level which allows periodic activation of CDK (121). The activation

    of CDK at different cell cycle phases requires different cyclins as summarized

    in Table 2.1 below. Cyclin D family (Cyclin D1-3) is involved in binding to

    CDK4 and 6 and the CDK-Cyclin D complex is involved in cell cycle

    progression into G1 phase (122).

  • 28

    Expression of cyclin D is not consecutive but rather driven by growth factor

    stimulation (123), unlike other cyclins which is expressed periodically. Cyclin

    E is another cyclin involved in G1 associates with CDK2 which allows

    progression from G1 into S phase (124). Cyclin A forms an essential complex

    with CDK2 in S phase progression (125) and with CDK1 to promote entry into

    mitosis phase. Further regulation in the mitosis phase is done by CDK1-cyclin

    B complex (126). Out of the sixteen identified cyclin proteins, not all are

    involved in cell cycle regulation (127,128). Some cyclins are involved in

    ubiquitination meditated proteolysis at end of each cycle phase (129).

    CDK Cyclin Cell cycle phase

    involved

    CDK4 D1, D2, D3 G1 CDK6 D1, D2. D3 G1

    CDK2 E G1/S transition

    CDK2 A S

    CDK1 A G2/M transition

    CDK1 B Mitosis

    CDK7 H All (As CDK activating

    kinase)

    Table 2.1: Summary of the CDK and cyclin pair involved in the regulation of

    cell cycle phases.

  • 29

    Cell cycle quality control are in place namely restriction points and checkpoints

    to ensure the correct cell cycle progression which upon blocking of early cell

    cycle events such as inhibition of DNA synthesis, later events such as mitosis

    and cytokinesis will be halted.

    Cell cycle in eukaryotic cells is safeguarded at three checkpoints which is at the

    boundary between G1 and S phase, G2 and mitosis phase and metaphase and

    anaphase. Should the condition for cell division is unmet, cell cycle progression

    will be halted at these checkpoints. These checkpoints are made out of

    accelerators and brakes that control progression of cell cycle. Surveillance

    mechanisms are in place to detect conditions and send inhibitory signals should

    the condition is sensed to be unfavorable which are essential for cell

    survivability under hostile environment.

    The G1 checkpoint in mammalian cells is known as the restriction point.

    Restriction point can be summarized as the point of no return where when the

    cell passes this point, it became committed towards cell cycle progression

    and doesnt require stimulation from proliferation stimulants (130). Cyclin D

    will form a complex with G1 phase CDKs and inhibits Rb that is involved in

    negative regulation of cyclin A and E which are involved in synthesis of DNA

    plus accumulation of cyclin B, through binding and inactivation of E2F

    transcription factor and inhibits ribosomal RNA gene transcription leading to

    cell growth inhibition (131).

  • 30

    As mentioned previously, should a cell pass the restriction point (for

    mammalian cells) or START (for budding yeast cells) it will be committed to

    the cell cycle. The irreversible transition is due to positive feedback in the

    CDK-cyclin cell cycle control system. Mammalian cells that does not

    undergoes cell division remains in the G1 or G0 phase as the CDKs and cyclin

    are kept inoperative through three means: suppression of cyclin genes

    transcription by Rb protein, rapid degradation of cyclin by APCCDH1 and

    inhibition by p27 all of which can be inactivated by the CDK-cyclin complex

    phosphorylation. The balance between the antagonists and CDK-cyclin creates

    two irreversible states in the cell cycle: the G1 state and S-G2-M state. During

    the restriction point period, the G1 CDK-cyclin complex (CDK4 and 6 with

    cyclin D) removes the antagonist through phosphorylation of Rb and p27 hence

    tipping the scales in favour of an irreversible transition to the S-G2-M state. The

    state of commitment is irreversible as upon passing the restriction point, cyclin

    D are not required for cell commitment as S and M phase CDK-cyclin complex

    will maintain the inhibition of their antagonists until the end of mitosis where

    all S and M phase cyclins are lost and thus removing the inhibition on

    antagonists which sees the cells maintained in G1 phase (132).

    The activity of CDK1-cyclin B complex or M phase-promoting factor (MPF) is

    essential for cell cycle progression into mitosis phase. Complete undamaged

    DNA replication is required before chromosomal condensation and subsequent

    nuclear division can occur in mitosis.

  • 31

    Should DNA damage is detected, the cell will be arrested in between S and G2

    phase by inactivation of MPF through Wee1 phosphorylation of tyrosine and

    threonine residues within the catalytic site into preMPF thus allowing time for

    DNA repair. The transition from G2 into M phase is continued by Cdc25C

    dephosphorylation of preMPF into MPF. Hence, the G2 checkpoint is guarded

    by positive feedback of MPF which inhibits Wee1 and activates Cdc25C

    concurrently. The transition from G2 to M phase is blocked by DNA damage

    through activation of checkpoint kinase (Chk) 1 and/or 2 which phosphorylates

    Cdc25C that subsequently causes binding to protein 14-3-3 in the cytoskeleton

    and sequestered away in the cytosol thus prevents the conversion of preMPF to

    MPF. Additionally, Chk2 phosphorylation of Cdc25C reduces its catalytic

    activity (133). In incomplete DNA replication or DNA damage, persistent

    presence of single stranded DNA is detected by ataxia telangiectasia and Rad3-

    related protein (ATR) in junction with ATRIP and leads to activation of Chk1

    by phosphorylation. The sequestration of Cdc25 and p53 stabilization by

    activated Chk1 leads to arrest between the S and G2 phase, activation of DNA

    repair enzymes or apoptotic signaling.

  • 32

    At the end of metaphase, all chromosomes should be attached by its

    kinetochores to the bipolar mitotic spindle before sister chromatids separation

    in anaphase can happen. This event is safeguarded by spindle or metaphase

    checkpoint. If presence of free kinetochores is detected such as through

    treatment with microtubule depolymerizing drugs such as nocodazole,

    vinblastine or podophyllotoxin, mitosis is blocked (134).

    Early events in mitosis such as the breaking down of nuclear envelope,

    followed by assembly of spindle fibres and chromosomal alignment are

    promoted by the CDK1-cyclin B complex or MPF but inhibit later mitotic

    events. The activation of Cdc20-APC complex is also promoted by MPF action

    which initiates the separation of sister chromatids in anaphase through the

    destruction of securin or precocious dissociation of sister chromatids (Pds) 1 in

    budding yeast cells. Securin inhibits the protease separase or Esp1 in yeast cells

    throughout most part of cell cycle which upon removal of securin triggers

    separase activation leading to degradation of the cohesin proteins holding the

    sister chromatids together, hence leading to the first stage of anaphase. The

    Cdc20-APC complex also targets and degrades the cyclin B component in the

    MPF complex in a negative feedback fashion, hence as the cell cycle

    progresses, the activities of MPF and Cdc20-APC complex will sequentially

    rises and fall and eventually quenched at the end of mitosis with the loss of

    cyclin B and the cells reenters G1 phase.

  • 33

    The spindle checkpoint blocks progression of mitosis as free kinetochores will

    activate Mad2 protein which binds and inhibits Cdc20 thus preventing from

    degrading securin and mitotic cyclin and causes arrest in metaphase. Post

    mitosis, the daughter cells reenters the G1 state.

    Figure 2.2: A summary of the mammalian cell cycle with its respective

    checkpoints. Arrow in red represents inhibition and arrow in green represents

    stimulation while arrow in black indicates the approximate time point within

    the cell cycle that is regulated by its respective cyclin-CDK complex.

  • 34

    2.6 Ceramide

    As mentioned previously, the core mechanism of phenoxodiol-induced

    apoptotic cell death is due to the accumulation of ceramide. Ceramides consists

    of a family of lipid molecules. Composed of portion of sphingosine and fatty

    acid, these sphingolipids is a major structural element found in biomembranes

    which together with phosphocholine or phosphoethanolamine forms

    sphingomyelin, an important lipid in lipid bilayer. It was later found that the

    role of ceramide extended beyond structural roles by exhibiting a diverse effect

    on cellular signaling and cell function regulation, one of such is the induction

    of signaling cascade that potentiates apoptosis. Generation of ceramide is

    through three intrinsic pathways de novo synthesis pathway, hydrolysis of

    sphingomyelin and salvage pathway which is summarized in Figure 2.3.

    Figure 2.3: Ceramide production and metabolism pathway.

  • 35

    2.6.1 Ceramide and apoptosis

    2.6.1.1 Ceramide and the extrinsic pathway

    The hydrophobicity of ceramide ensures ceramide is always partitioned within

    the bilayer membrane at its site of production and exerting its function from

    within. Known as the ceramide-enriched membrane platforms (135), these

    regions serves to cluster the death receptors, TRAILR2 and CD95 and upon

    activation amplify downstream apoptotic signaling events through facilitation

    of DISC complex formation (136). TRAIL and TNF-induced apoptosis

    associates with activation of neutral and acid sphingomyelinase which leads to

    increased ceramide formation (137). Additionally, in several tumor types such

    as glioblastoma (138)and prostate cancer (139), the down regulation of FLICE

    inhibitory proteins through inactivation of Akt pathway, removes caspase-8

    inhibition and subsequently promotes apoptosis.

  • 36

    2.6.1.2 Ceramide and the intrinsic pathway

    As mentioned previously, the extrinsic and intrinsic apoptotic pathway are not

    mutually exclusive of one another, cross over can happen from extrinsic to

    intrinsic pathway via FLIP inhibition, caspase-8 and truncated-BID activation.

    The proapoptotic nature of ceramide is largely attributed to the orchestration of

    a myriad of downstream signaling pathway that eventually causes the release of

    pro-apoptotic proteins from the mitochondria a la the intrinsic pathway.

    However, to do that, it must first reach the mitochondria which are hindered by

    its hydrophobic nature of ceramide. To overcome, upon generation within the

    membrane bilayer, ceramide platforms are formed that infold into the cytosol

    and fuses with mitochondria. With that, a commute pathway is formed that

    allows the direct transfer of ceramide from plasma membrane to mitochondria

    leading to accumulation of ceramide at mitochondria and subsequent apoptosis

    (140). Other methods that causes mitochondrial accumulation of ceramide

    includes the production of ceramide via de novo or the salvage pathway by

    mitochondria-associated membrane (141) and the localization of key ceramide

    production enzymes such as ceramide synthase, neutral sphingomyelinase and

    ceramidase in mitochondria for in situ production of ceramide (142-145).

  • 37

    The key event of ceramide apoptotic signaling is the induction of mitochondrial

    outer membrane permeabilization (MOMP) from ceramide channel formation.

    The level of mitochondria accumulated ceramide is shown to directly correlate

    with MOMP (146). The induction of MOMP allows the leakage of

    mitochondria apoptotic proteins such as cytochrome c and intermembraneous

    proteins that have a molecular mass lesser than approximately 60kDa (147).

    Ceramide alone is insufficient to induce MOMP but rather together with the

    proapoptotic Bcl-2 protein, Bax in synergistically causes the permeabilization

    of mitochondria outer membrane (148) through the formation of ceramide-rich

    macrodomains essential for BAX insertion, oligomerization and pore formation

    (149).

    Additionally, ceramide also causes a transient increased in pH intracellularly

    leading to essential conformational changes in BAX (150) and allowing BAX

    translocation from the cytosolic 14-3-3 proteins to mitochondria via JUN N-

    terminal kinase activation which is a downstream process from p38 MAPK

    activation and Akt down regulation of ceramide (151). The collective activation

    of protein phosphatase 2A (PP2A) and the endolysosome protease cathespin D

    plus the inactivation of Akt by ceramide all together contributes to the

    activation of glycogen synthase kinase 3 (152,153) which also induces MOMP

    through the activation of caspase-2 and caspase-8 which leads to the cleavage

    of BID to form tBID that translocate to mitochondria (154).

  • 38

    In addition to the activation of protein phosphatase 2A, caspase-2 activation

    and mitochondria apoptosis is also induced by ceramide through the down

    regulation of BCL-2, a pro-survival mitochondrial protein that blocks

    apoptosis, overloading of calcium ions and apoptosis receptors (155). Another

    effect of ceramide on mitochondrial function is the activation of protein kinase

    C which likewise leads to release of cytochrome c and activation of caspase-9

    (156).

    2.6.1.3 Ceramide induced apoptotic signals

    The downstream apoptotic cascade orchestrated by ceramide is due to the

    induced apoptotic signals in upstream pathways. Akt, a serine/threonine-

    specific protein kinase is one of the major pathways being down regulated by

    ceramide (157). Akt pathway is closely associated with tumorigenesis and

    frequently altered in most cancer types and indicates poor prognosis and

    chemoresistance (158). Activation of Akt occurs through growth factors

    binding to a plasma membrane tyrosine kinase receptor which activates PI3K

    that converts phosphatidylinositol-4,5-biphosphate (PIP2) to

    phosphatidylinositol-3,4,5-trisphosphate (PIP3). Cytosolic Akt is then

    translocated to the plasma membrane in an event triggered by PIP3 where it

    will be activated via phosphorylation by phosphoinositide-dependent protein

    kinase 1 and mTOR complex 2 (159).

  • 39

    Ceramide inhibits Akt pathway via three methods. The atypical PKC activity

    is found to be stimulated by the active ceramide form, C6 ceramide which

    causes increased association of PKC with Akt (160), in addition, the binding

    of PIP3 to Akt PH domain is stifled by PKC through phosphorylation on PH

    domain Thr34 which effectively blocks Akt translocation to the plasma

    membrane to be activated (161). As mentioned, another direct downstream

    signal of ceramide is the activation of PP2A (157). In various cancer types, the

    activation of PP2A correlates with inhibited Akt signaling (162). The activation

    of PP2A can occur directly and indirectly by ceramide where the association

    between inhibitor 2 of PP2A and PP2A is reduced which resulted in indirect

    PP2A activation (163). Likewise, Akt phosphorylation can be reduced by the

    activation of p38 from ceramide action as observed in HL-60 cells (151).

    The MAP kinases p38 and JNK that is involved in cell growth and survivability

    is also found to be regulated by both exogenous (164) and endogenous

    ceramide (73). Both of these MAP kinases are activated by ceramide through

    upregulated transcriptional expression of thioredoxin-interacting protein which

    in-turn diminishes thioredoxin activity thus removing the inhibition of

    apoptosis signal regulating kinase 1 leading to downstream activation of both

    p38 and JNK (164).

  • 40

    Activation of p53 is also being promoted by ceramide action (165). The

    activation of PP2A also leads to inhibition of the anti-apoptotic Bcl-2

    phosphorylation and subsequent increased binding between p53 and Bcl-2

    therefore inhibits Bcl-2 and thus leading to apoptosis (166). The accumulation

    of p53 also leads to increased pro-apoptotic Bax level and the decreased in the

    anti-apoptotic Bcl-2 level in neuroblastoma cells, suggesting the regulation of

    Bax/Bcl-2 ratio by p53 in ceramide-induced apoptosis as one the many

    apoptotic mechanism of ceramide (167).

    Finally, ceramide is shown to downregulates prosurvival IAP survivin involved

    in cell proliferation and division, metastasis and angiogenesis in tumor cells

    (168). The expression of survivin at the transcriptional level is inhibited by

    ceramide. The downregulation of survivin synergistically enhanced cell death

    from the intrinsic apoptosis pathway and increased p53 and Bax all of which is

    meditated by ceramide action.

  • 41

    2.6.2 Ceramide and cell cycle

    As shown in Figure 2.1, ceramide regulation of cell cycle involves the

    modulation of cell cycle inhibitors. During the cell cycle G1 to S phase

    transition, complexes are formed between cyclin D and E with CDK2, 4 and 6

    to phosphorylate Rb. Ceramide is found to upregulate p21 leading to activation

    of Rb through decreased expression of cyclin E and D1 and CDK 2 and 7

    activity (169) then inactivates it through degradation in a negative feedback

    fashion to reverse the cell cycle checkpoint arrest (170).

    The mechanism of cell cycle arrest can be partly attributed to activation of

    peroxisome proliferator-activated receptor- (PPAR) (169) to which a reversal

    in CDK7 suppression is found when treated with PPAR antagonist. PPAR

    family of membranous receptor proteins functioning as transcriptional factors

    in regulating genes involved in cell differentiation and metabolism. The

    activation of PPAR causes growth arrest in various cancer cell types (171).

    Exogenously added ceramide is found to stimulate PPAR which subsequently

    forms a heterodimer with retinoid X receptor to bind and activate specific

    regions of DNA. Expression of the CDK inhibitor, p21 is also found to be

    upregulated by PPAR (172) which as a consequence sees the accumulation of

    dephosphorylated Rb and CDK2 association with p21 (173). In addition, p21

    activity is also found to be regulated by ceramide through p53 induction

    leading to downstream activation of p21 (174).

  • 42

    The upregulation of protein phosphatase activity by ceramide leads to CDK2

    inhibition which is reversible upon treated with protein phosphatase antagonists

    (175) as PP2A meditates p27 expression via Akt dependent and independent

    pathways. Finally, the inhibition of Akt pathway by ceramide also

    synergistically enhance p27 action through stabilization of the CDK inhibitor

    which all together contributes to the cell cycle arrest in G1 phase meditated by

    ceramide action.

    Accumulation of ceramide also leads to G2 arrest as seen in rhabdomyosarcoma

    cells from the increased expression of p21 and downregulation of cyclin D

    (176). Ceramide is found to inhibit MDM2 which binds and meditates

    proteosomal degradation of p21. MDM2 also regulates p53 in a negative

    fashion, leading to decreased p21 levels. Consistent with these findings,

    overexpression of MDM2 diminishes G2 arrest meditated by ceramide

    upregulation of p21. Suppression of survivin expression associated with

    transition between G2 and M phase is also found to be meditated by ceramide

    which similarly leads to arrest in G2 phase (177,178).

  • 43

    3 MATERIALS AND METHODS

    3.1 Materials

    Name Brand Product ID

    Annexin V-FITC early

    apoptosis detection kit

    CELL SIGNALING

    TECHNOLOGY INC, USA

    6592

    Cycloheximide ACROS ORGANICS, USA 35742-0010

    Dimethyl sulfoxide (DMSO) SIGMA-ALDRICH, USA 472301

    Dulbeccos Modified Eagle Medium (DMEM)

    SIGMA-ALDRICH, USA D7777

    Ethylenediaminetetraacetic

    acid (EDTA)

    SIGMA-ALDRICH, USA E9617-1-0250

    Fetal bovine serum (FBS) SIGMA-ALDRICH, USA F7524

    Hydrochloric acid ACROS ORGANICS, USA 12463-0010

    Penicillin-streptomycin

    antibiotic

    GIBCO, LIFE

    TECHNOLOGIES, USA

    15140-122

    Phenoxodiol SIGMA-ALDRICH, USA D7446

    Phosphate buffer saline (PBS) AMRESCO, USA E404

    Propidium iodide SIGMA-ALDRICH, USA 81845

    RNAse A SIGMA-ALDRICH, USA R6148

    Sodium azide SIGMA-ALDRICH, USA S2002

    Sodium bicarbonate AMRESCO, USA

    E404

    Sodium hydroxide SIGMA-ALDRICH, USA 221465

    Thiazolyl Blue Tetrazolium

    Blue (MTT)

    SIGMA-ALDRICH, USA M5655

    Triton X-100 SIGMA-ALDRICH, USA X100

    Tryphan blue solution 0.4% SIGMA-ALDRICH, USA T8154

    Trypsin-EDTA solution 10X SIGMA-ALDRICH, USA 59418C

    Table 3.1: List of chemicals used

  • 44

    Name Brand Product ID

    6-well plates JET BIOFIL, USA TCP011006

    96-well plates THERMOSCIENTIFIC,

    USA

    163320

    Bottle-top vacuum filter SIGMA-ALDRICH, USA 430049

    Centrifuge tubes (15 mL) GREINER BIO-ONE, USA 188271

    Centrifuge tubes (50 mL) THERMOLINE

    SCIENTIFIC, AUSTRALIA

    TL32103

    Culture flask (25cm2) THERMOSCIENTIFIC,

    USA

    EW-01932-41

    Microcentrifuge tubes AXYGEN SCIENTIFIC,

    USA

    MCT-150-C

    Serological pipettes (5 mL) JET BIOFIL, USA GSP010005

    Serological pipettes (10 mL) JET BIOFIL, USA GSP010010

    Sterile petri dishes THERMOLINE

    SCIENTIFIC, AUSTRALIA

    CCD009015

    Pipette tips GREINER BIO-ONE,

    BELGIUM

    771290 (0.5-20L)

    739261 (10-200L)

    877270 (200-1000L)

    Table 3.2: List of consumables used

  • 45

    Name Brand Product ID

    Autoclave system HIRAYAMA, JP HV-50

    Centrifuge EPPENDORF,

    GERMANY

    5810

    Class II biosafety

    cabinet

    ESCO

    TECHNOLOGIES INC,

    USA

    SC2-4A1

    CO2 incubator NEW BRUNSWICK

    SCIENTIFIC.

    GERMANY

    170-200

    Flow cytometer BD BIOSCIENCE, USA BDFACSCALIBUR

    Fluorescence

    microscope

    NIKON

    CORPORATION,

    JAPAN

    ECLIPSE-TI

    Hemocytometer GRALE HDS, AUS 4514

    Inverted light

    microscope

    LEICA

    MICROSYSTEMS,

    USA

    DM13000B

    Microcentrifuge EPPENDORF,

    GERMANY

    MINISPIN

    pH meter HANNA

    INSTRUMENTS, USA

    HI 110

    Pipettes EPPENDORF,

    GERMANY

    RESEARCHPLUS

    Refrigerated centrifuge EPPENDORF,

    GERMANY

    5804R

    Ultrapure water system EMD MILIPORE,

    CANADA

    SYNSVR0WW

    Vortex FINEPCR, KOREA FINEVORTEX

    Water bath MEMMERT,

    GERMANY

    WMB10

    Table 3.3: List of apparatuses used

  • 46

    3.2 Methods

    3.2.1 Preparation of culture media and solutions

    3.2.1.1 DMEM

    Thirteen point five gram of DMEM in powder form, containing 4500 mg/L

    glucose and L-glutamine and 110 mg/L sodium pyruvate was weighted out and

    together with 3.7 g of sodium bicarbonate were dissolved using ultrapure water

    to make approximate 800 mL of media. pH of media was adjusted to between

    7.1 to 7.3 using a pre-calibrated pH meter. Media was subsequently top up with

    ultrapure water to make 1 L volume. Under aseptic condition, media was filter

    sterilized with an aid of a vacuum pump into sterile pre-autoclaved bottles. 10

    mL of filtered media was aliquot out into a sterile petri dish and incubated for 3

    days in a CO2 incubator at 37oC and 5% CO2 to validate sterility by noting

    media appearance such as cloudiness, colour changes or presence of microbes

    under microscopic observation or naked eyes. Only sterility verified media will

    be used for cell culturing and experiments. Prepared DMEM media was stored

    in 4oC temperature.

  • 47

    3.2.1.2 PBS

    One tablet of PBS containing 10mM phosphate buffer, 137mM sodium

    chloride, and 2.7mM potassium chloride was dissolved in 100 mL of ultrapure

    water before being autoclaved at 121oC for 15 minutes. Prepared PBS was

    stored in 4oC temperature.

    3.2.1.3 Trypsin-EDTA

    One part of 10X Trypsin-EDTA solution containing 0.5% trypsin and 0.2%

    EDTA was diluted with nine part of sterile PBS to make 1X Trypsin-EDTA

    solution. Prepared Tryspin-EDTA solution was stored in -20oC temperature.

    3.2.1.4 Phenoxodiol and Cycloheximide

    Analytical grade phenoxodiol and cycloheximide was weighted and dissolved

    in DMSO to make a stock concentration of 10 mg/mL which was further

    diluted pre-usage in experiments.

    3.2.1.5 MTT solution

    MTT powder was weighted and dissolved in sterile PBS to make a stock

    concentration of 5 mg/mL in dark.

  • 48

    3.2.2 Culture media for cell lines

    The initial cultures for HepG2 (Homo sapiens hepatocellular carcinoma), HT-

    29 (Homo sapiens colorectal adenocarcinoma), CNE1 (Homo sapiens highly

    differentiated nasopharyngeal carcinoma) and C6 (Rattus norvegicus glioma)

    cell lines used in this study were obtained from from IMUs Institute for

    Research Development and Innovation (IRDI) cell bank (Kuala Lumpur,

    Malaysia) and were cultured in vitro using sterilized high glucose DMEM

    supplemented with 3.7 g/L sodium bicarbonate, 10% FBS and 1% Pen-Strep

    antibiotic solution containing 100 units/mL penicillin and 100 units/mL

    streptomycin hereby designated as complete DMEM.

    The basal medium DMEM is a modified version of Basal Medium Eagle

    (BME) with a four-fold increased concentration of amino acids and vitamins.

    The high glucose content with 4500 mg/L is shown to improve cultivations of

    various cell lines (179). The supplementation of FBS is required as to provide

    the essential proteins and growth factors for optimum cell growth (180) while

    sodium bicarbonate acts as a buffering system in the presence of artificial CO2

    for pH maintenance (179). The use of antibiotic is to suppress growth of

    microbial contaminants (181).

  • 49

    3.2.3 Maintaining and sub-culturing of cells

    All cell lines were seeded and cultured in 25cm2 cell culture flask and

    incubated in a CO2 incubator producing an artificial humidified environment

    with 5% CO2 at 37oC. The cultures were maintained at exponential phase by

    ensuring a cell confluence of less than 90%. Cultures having 80% and above

    confluence will have its extra cells discarded where in brief, old media is

    discarded and the cells were washed with 1 mL of sterile PBS by gently

    swirling the flask as to prevent air bubbles formation. PBS was subsequently

    discarded and 1 mL of 1X Trypsin-EDTA solution was added to flask and

    incubated in CO2 incubator for approximately 5 minutes at 37oC and 5% CO2.

    The flask was tapped gently to further ease detachment of cells and observed

    under an inverted light microscope to verify cells detachment. Approximately

    500 L of detached cells was discarded and the remaining was resuspended in

    10 mL of fresh complete DMEM. The flasks were then further incubated for

    three to four days at 5% CO2 at 37oC. Old flasks will be reused up to a month

    before sub-culturing to a new flask where similarly, cells were collected by

    trypsinization method and resuspended in 1 mL of complete DMEM before

    transferring to new flask. The final volume of the flask was brought up to

    approximately 10 mL by adding 9 mL of fresh culturing media. All procedures

    was performed aseptically in a Class II Biosafety Cabinet and all media and

    solution used were pre-warmed to 37oC in a water bath before used.

  • 50

    3.2.4 Cell seeding

    Culture was first allowed to achieve a confluence level around 80-90%.

    Following that, the cells were seeded into a sterile 96-well or 6-well plate. The

    old media were discarded and the cells were washed with 1 mL of sterile PBS.

    PBS was then discarded and replaced with 1 mL of 1X Trypsin-EDTA solution

    before incubating in a humidified CO2 incubator for approximately 5 minutes at

    37oC and 5% CO2. The flask was tapped gently to detach cells and

    subseque