identification of genes and potential pathways involved in ... · identification of genes and...

221
Identification of Genes and Potential Pathways Involved in Familial Ovarian Cancer By Kelly Kai Yin Seto A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy, Graduate Department of Molecular and Medical Genetics University of Toronto © Copyright by Kelly Kai Yin Seto (2011)

Upload: others

Post on 14-Mar-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Identification of Genes and Potential Pathways Involved in

Familial Ovarian Cancer

By

Kelly Kai Yin Seto

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy,

Graduate Department of Molecular and Medical Genetics

University of Toronto

© Copyright by Kelly Kai Yin Seto (2011)

Page 2: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Identification of Genes and Potential Pathways Involved in

Familial Ovarian Cancer

Degree of Doctor of Philosophy, 2011

Kelly Kai Yin Seto

Department of Molecular Genetics

University of Toronto

ABSTRACT

One of the most important risk factors in ovarian cancer is family history, and two

well-studied tumour suppressor genes BRCA1 and BRCA2 have already been identified in

“high-risk” families. However, alterations of other genes may also be important for

ovarian cancer pathogenesis in individuals with family history of breast/ovarian cancer.

In this thesis, I compared the gene expression profiles of tumours from patients

with strong and weak family history of breast and/or ovarian cancer to identify genes that

may be significant in the subset of patients with ovarian cancer predisposition. Based on

this comparison, two genes of interest were selected for further investigations:

hCDC4/FBXW7 (F-box and WD repeat domain containing 7) and PRKCZ (protein kinase

C zeta).

Through mutational analyses I identified one nucleotide alteration within exon 7

of hCDC4; however, overall I found that hCDC4 mutation is a rare event in ovarian

tumours. Additional epigenetics analyses revealed that promoter methylation is not a

significant mechanism responsible for repression of hCDC4 expression in ovarian cancer.

Nevertheless, the variable expression of hCDC4 proteins observed in ovarian tumour

ii

Page 3: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

tissues by immunohistochemical staining of tissue microarrays suggests that hCDC4

deregulation may potentially be important in a subset of ovarian cancers.

Additionally, I observed that expression levels of PRKCZ are higher in ovarian

tumours from patients with strong family history compared to patients with weak family

history. PRKCZ has previously been shown to be involved in a variety of cellular

processes; however its role in ovarian cancer remained elusive. To further understand the

role of PRKCZ in ovarian tumourigenesis, including cell viability, cell migration, as well

as relevant downstream signaling pathways, I performed functional assays using an in

vitro ovarian cancer model. I observed that PRKCZ increases proliferation of the

SKOV3 ovarian cancer cell line and participates in EGF-induced chemotaxis.

Furthermore, I identified IGF1R (insulin-like growth factor 1 receptor) and ITGB3

(integrin beta 3) as downstream effectors of PRKCZ as expression of these genes is

significantly altered when PRKCZ is over-expressed. Given their previously identified

associations with familial ovarian cancer, the IGF1 and ITGB3 signaling pathways may

therefore represent a possible link between PRKCZ and this disease.

iii

Page 4: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

ACKNOWLEDGEMENTS

First and foremost I would like to thank my supervisor, Dr. Irene Andrulis, for

giving me the opportunity to work on this meaningful project, and for your continuous

guidance during my graduate career. I would also like to thank my committee members,

Dr. Johanna Rommens and Dr. Jeff Wrana. Your intellectual input and enthusiasm for

my project were much appreciated.

I could not have been able to complete my thesis without the help and advice of

my collaborators. Thank you to Dr. Barry Rosen, Dr. Joan Murphy, Dr. Patricia Shaw

and Heather Begley for providing samples for my study, and Gordon Glendon for his

help for assisting in classification of familial ovarian cancer cases. Thank you to Dr.

Shelley Bull, Dr. Dushanthi Pinnaduwage, and Sarah Colby for their help with statistical

analyses of microarray data. I would also like to thank all members of the Toronto

Ovarian Research Network for discussions on my project, and a special thank you goes

Dr. Ted Brown, Alicia Tone and Katherine Sodek for all their helpful advice.

I am very grateful for the support from my “lab family”. Nalan, thank you for

being so generous with your time – from training me on my first microarray experiment,

to the many hours of discussions in the years that followed. Kolja, Chris, Sherry, Andras,

Anita, and Teresa – it was amazing to have such an intelligent group of Ph.D. students

that I could look up to; your mentorship is much appreciated. Yan, it was such a pleasure

to work with you, and I admire your inherent desire for learning. Lucie, your love for

science is inspiring, and I look forward to many more photography outings with you.

Mona, thanks for hiding chocolates in my desk to prop me up during those down days.

To all Andrulis lab members, past and present: Sasha, Eduard, Monica, Salvador,

iv

Page 5: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Catherine, Andrew, Irene, Andreas, Atta, Kristine, Sandrine, and our numerous talented

undergraduate students – thank you for all your support and friendships. Sean, Duygu,

and Winnie, your constant words of encouragement, optimism, and friendships mean a

great deal to me.

I would also like to take this opportunity to thank all my friends, from childhood

buddies to friends I have made in the department of Molecular Genetics and at the

Samuel Lunenfeld Research Institute (SLRI) – you definitely have made my graduate

experience fun and memorable. Thank you to my formal labmates and friends from

McMaster University who played a big part in my decision to pursue my graduate

studies. A huge thank you goes to SciHigh, the wonderful science outreach program

based at the SLRI. Seeing the look of excitement on kids’ faces everytime they look at

GFP-mice or banana DNA in eppendorf tubes reminds me of why I love science in the

first place.

Last but not least, I would like to thank my parents and brother for their patience

and support through this journey. Words cannot express how lucky I feel to have you.

v

Page 6: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

TABLE OF CONTENTS ABSTRACT ....................................................................................................................... ii ACKNOWLEDGEMENTS............................................................................................... iv TABLE OF CONTENTS .................................................................................................. vi LIST OF TABLES.............................................................................................................. x LIST OF FIGURES ........................................................................................................... xi LIST OF ABBREVIATIONS ......................................................................................... xiv

CHAPTER 1

Introduction & Literature Review.................................................................................. 1 1.1 Principles of Molecular Cancer Genetics ..................................................................... 2

1.1.1 Gatekeepers............................................................................................................ 3 1.1.1.1 Tumour Suppressor Genes.............................................................................. 5 1.1.1.2 Oncogenes....................................................................................................... 7

1.1.2 Caretakers .............................................................................................................. 7 1.1.3 Landscapers ........................................................................................................... 8

1.2 Ovarian Cancer ............................................................................................................. 9 1.2.1 Model of Ovarian Carcinogenesis ....................................................................... 11 1.2.2 Familial Ovarian Cancer...................................................................................... 15

1.2.2.1 Hereditary Site-Specific Ovarian Cancer and Hereditary Breast-Ovarian Cancer Syndrome (HBOCS) .................................................................................... 16 1.2.2.2 Hereditary Nonpolyposis Colorectal Cancer (HNPCC) ............................... 17

1.2.3 Molecular Pathology and Genetics of Ovarian Cancer ....................................... 17 1.2.3.1 p53 ................................................................................................................ 18 1.2.3.2 Wnt-Signaling Pathway................................................................................ 18 1.2.3.3 PI3K/Akt Signaling Pathway........................................................................ 19 1.2.3.4 MAPK Signaling Pathway............................................................................ 20 1.2.3.5 Cell Cycle Genes .......................................................................................... 21 1.2.3.6 Epidermal Growth Factor Family Receptors................................................ 23 1.2.3.7 Estrogen Receptors ....................................................................................... 24

1.2.4 BRCA1 and BRCA2............................................................................................ 25 1.2.4.1 BRCA1 and BRCA2 in Ovarian Cancer ...................................................... 26 1.2.4.2 Mutations of BRCA1 and BRCA2 in Ovarian Cancer................................. 27 1.2.4.3 Specific BRCA1 and BRCA2 mutations in closed populations................... 29 1.2.4.4 Modifiers of BRCA1 and BRCA2................................................................ 30

1.3 Ovarian Cancer Genome-Wide Association Studies.................................................. 31 1.3.1 Array Comparative Genomic Hybridization of Ovarian Cancer......................... 32 1.3.2 Single Nucleotide Polymorphism Array Analysis of Ovarian Cancer ................ 34 1.3.3 Gene Expression Profiling of Ovarian Cancer .................................................... 37

1.4 Hypothesis .................................................................................................................. 41 1.5 Rationale and Objectives ............................................................................................ 41

vi

Page 7: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

CHAPTER 2

Gene Expression Profiling of Familial Ovarian Cancer ............................................. 43 2.1 Introduction ................................................................................................................ 44 2.2 Materials and Methods ............................................................................................... 45

2.2.1 Ovarian Cancer Specimens.................................................................................. 45 2.2.2 Family History Classification.............................................................................. 45 2.2.3 RNA Isolation and Reverse Transcription........................................................... 47 2.2.4 cDNA Expression Microarrays............................................................................ 47 2.2.5 Microarray Data Analysis.................................................................................... 49

2.2.5.1 Pre-processing and normalization of expression data .................................. 49 2.2.5.2 Microarray Statistical Analysis..................................................................... 49

2.2.6 Quantitative Real-time RT-PCR.......................................................................... 50 2.2.7 Integration of Array Data to Interaction Networks.............................................. 52

2.3 Results ........................................................................................................................ 53 2.3.1 Identification of Genes Distinguishing Strong and Weak Familial Ovarian Cancers ......................................................................................................................... 53

2.3.1.1 Supervised-Class Comparison...................................................................... 53 2.3.1.2 Candidate Gene Approach............................................................................ 58

2.3.2 Validation of Differentially Expressed Genes..................................................... 58 2.3.3 Molecular Network Analyses .............................................................................. 61

2.4 Discussion................................................................................................................... 68

CHAPTER 3 hCDC4 in Familial Ovarian Cancer.............................................................................. 75 3.1 Introduction ................................................................................................................ 76 3.2 Materials & Methods .................................................................................................. 79

3.2.1 Ovarian tumour samples, and RNA, DNA Extraction ........................................ 79 3.2.2 Protein Truncation Test ....................................................................................... 79 3.2.3 Single Strand Conformation Polymorphism (SSCP) and Manual Sequencing... 80 3.2.4 DNA Methylation-Specific PCR ......................................................................... 82 3.2.5 Loss of Heterozygosity (LOH) Analysis of hCDC4............................................ 83 3.2.6 Immunohistochemical (IHC) Staining of Ovarian Tissue Microarrays .............. 84 3.2.7 Quantitative Real-time PCR for CCNE1 ............................................................. 85

3.3 Results ........................................................................................................................ 86 3.3.1 hCDC4 Sequence Alteration Detection by PTT Analysis................................... 86 3.3.2 SSCP and Sequencing of hCDC4 ........................................................................ 86 3.3.3 hCDC4 Promoter Methylation Analysis.............................................................. 90 3.3.4 Loss of Heterozygosity Analysis of hCDC4 ....................................................... 90 3.3.5 hCDC4 Protein Expression in Ovarian Cancer ................................................... 94 3.3.6 Gene Expression of CCNE1 in Familial Ovarian Cancer.................................... 98

3.4 Discussion................................................................................................................. 100

vii

Page 8: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

CHAPTER 4 Characterization of PRKCZ in Ovarian Cancer....................................................... 106 4.1 Introduction .............................................................................................................. 107 4.2 Materials and Methods ............................................................................................. 109

4.2.1 Cell Culture........................................................................................................ 109 4.2.2 PRKCZ Expression Vector & Generation of Stable Clones ............................. 109 4.2.3 Quantitative Real-Time PCR............................................................................. 110 4.2.4 Western Blotting................................................................................................ 110 4.2.5 MTT Cell Viability Assays................................................................................ 111 4.2.6 TUNEL Assays.................................................................................................. 112 4.2.7 BrdU Proliferation Assay .................................................................................. 112 4.2.8 Matrigel Transwell Assays ................................................................................ 113 4.2.9 Scratch Wound Healing and Pericentrin Orientation Assays ............................ 113 4.2.10 Phagokinetic Track Assays.............................................................................. 114 4.2.11 siRNA Transfections ....................................................................................... 115 4.2.12 Ingenuity Pathway Analyses............................................................................ 115 4.2.13 Statistical Analyses.......................................................................................... 115

4.3 Results ...................................................................................................................... 116 4.3.1 Generation of PRKCZ Stable Ovarian Cancer Cell lines.................................. 116 4.3.2. Cell Viability in PRKCZ-Expressing Cells ...................................................... 125 4.3.3 PRKCZ and ovarian cancer cell migration and invasion................................... 128 4.3.4 Identification of Potential Downstream Effectors of PRKCZ........................... 137

4.3.4.1 IGF1R and ITGB3 as Potential Downstream Effectors of PRKCZ ........... 137 4.3.4.2 TIMP-1 as a Potential Downstream Effector in ITGB3 and IGF1 Signaling................................................................................................................................ 146 4.3.4.3 Effects of IGF and ITGB3 Signaling on Cell Migration/Invasion in SKOV3 Cells ........................................................................................................................ 150

4.4 Discussion................................................................................................................. 156 CHAPTER 5 Conclusions and Future Directions............................................................................. 166 5.1 Summary and Implications of Thesis Findings ........................................................ 167

5.1.1 Expression Profiling of Familial Ovarian Cancer ............................................. 167 5.1.2 hCDC4 and Ovarian Cancer .............................................................................. 169 5.1.3 PRKCZ and Ovarian Cancer ............................................................................. 170

5.2 Future Directions ...................................................................................................... 173 5.2.1 High-Throughput Analyses of Familial Ovarian Cancer................................... 173

5.2.1.1 Gene Set Analysis of Expression Microarrays Data................................... 173 5.2.1.2 Genomic Signature of Familial Ovarian Cancer ........................................ 182

5.2.2 hCDC4 and Ovarian Cancer .............................................................................. 182 5.2.3 PRKCZ and Ovarian Cancer ............................................................................. 183

viii

Page 9: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Appendix…………………………………………………………………………….…187 Bibliography.................................................................................................................. 189

ix

Page 10: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

LIST OF TABLES Table 1-1. Characteristics of Type I and Type II ovarian tumours..................................13 Table 2-1. Characterization of the subjects in the strong versus weak family history groups................................................................................................................................46 Table 2-2. Thirteen cell lines composing common reference pool used for microarray experiments........................................................................................................................47 Table 2-3. Top 100 differentially expressed genes between strong and weak familial ovarian tumours, as ranked by SAM.................................................................................55 Table 2-4. Top functions of networks as identified by Ingenuity Pathway Analysis.......67 Table 3-1. Primer sets for hCDC4 SSCP and manual sequencing analyses.....................81 Table 3-2. Primer sequences for methylation-specific PCR.............................................82 Table 3-3. Polymorphic markers used for LOH analysis of hCDC4................................83 Table 3-4. Histological scores of hCDC4 immunhistochemical staining on ovarian tissue microarray..........................................................................................................................97 Table 5-1. Gene Set Analysis (GSA) of familial ovarian cancer expression microarray data...................................................................................................................................175 Table 5-2. Top functions of networks belonging to significant gene sets as identified by Gene Set Analysis (GSA) and Ingenuity Pathway Analysis (IPA).................................176

x

Page 11: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

LIST OF FIGURES Figure 1-1. Hallmarks of Cancer…………………………………………………………4 Figure 1-2. Knudson’s two-hit hypothesis……………………………………….....……6 Figure 1-3. Dualistic model of ovarian serous carcinoma development………….…….14 Figure 1-4. Gene structure of BRCA1 and BRCA2……………………………………...28 Figure 1-5. Chromosomal aberrations in ovarian tumours...............................................35 Figure 2-1. Subarray of a representative 19K cDNA microarray.....................................48 Figure 2-2. Expression of housekeeping genes in ovarian tumour samples as measured by gene expression microarrays.........................................................................................51 Figure 2-3 Heatmap illustrating differential gene expression patterns in strong and weak familial ovarian cancer groups...........................................................................................54 Figure 2-4. Identification of hCDC4 as a differentially expressed gene between strong and weak familial ovarian tumours by candidate gene approach......................................59 Figure 2-5. Real-time PCR validation of cDNA microarray expression analysis............60 Figure 2-6. Graphical representations of the molecular relationships between genes identified from familial ovarian microarray analysis using Ingenuity Pathway Analysis.............................................................................................................................62 Figure 3-1. Pathway of hCDC4-mediated degradation of cyclin E..................................77 Figure 3-2. hCDC4 protein truncation assay....................................................................87 Figure 3-3. Genetic analysis of hCDC4 with SSCP.........................................................88 Figure 3-4. hCDC4 gene sequence alteration found in exon 7.........................................89 Figure 3-5. Potential methylation sties with the hCDC4 promoter..................................91 Figure 3-6. Evaulation of hCDC4 promoter methylation by methylation-specific PCR....................................................................................................................................92 Figure 3-7. Loss of heterozygosity (LOH) analysis of hCDC4 in four cases of ovarian cancer.................................................................................................................................93 Figure 3-8. hCDC4 IHC staining optimization.................................................................95

xi

Page 12: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Figure 3-9. Immunohistochemical staining of ovarian tissue microarray with hCDC4 antibody.............................................................................................................................96 Figure 3-10. CCNE1 gene expression in familial ovarian cancer.....................................99 Figure 4-1. Endogenous gene and protein levels of PRKCZ in selected ovarian cancer cell lines...........................................................................................................................117 Figure 4-2. Expression of PRKCZ clones in HEY ovarian cancer cell line...................119 Figure 4-3. Expression of PRKCZ clones in SKOV3 ovarian cancer cell line..............121 Figure 4-4. Expression of PRKCZ clones in OVCAR3 ovarian cancer cell line...........123 Figure 4-5. PRKCZ increases cell viability in SKOV3 cells but not HEY and OVCAR3 cells..................................................................................................................................126 Figure 4-6. PRKCZ enhances proliferation of SKOV3 ovarian cancer cells.................127 Figure 4-7. PRKCZ has no effect on apoptosis in SKOV3 cells....................................127 Figure 4-8. Migration of ovarian cancer cells................................................................130 Figure 4-9. Effect of PRKCZ gene knockdown on SKOV3 parental cells migration as observed by wound healing assay....................................................................................131 Figure 4-10. Disorganized cell movement of HEY cells over-expressing PRKCZ........133 Figure 4-11. Measurement of cell polarity of HEY by pericentrin orientation assay.....135 Figure 4-12. Quantitation of ovarian cancer cell motility...............................................136 Figure 4-13. Identification of potential interactors of PRKCZ........................................138 Figure 4.14. Transcript and protein expression of IGF1R in PRKCZ-expressing SKOV3 cells..................................................................................................................................139 Figure 4.15. Transcript and protein expression of IGF1R in PRKCZ-expressing OVCAR3 cells........................... .....................................................................................141 Figure 4.16. Transcript and protein expression of ITGB3 in PRKCZ-expressing SKOV3 cells..................................................................................................................................142 Figure 4.17. Transcript and protein expression of IGF1R in PRKCZ-expressing OVCAR3 cells.................................................................................................................143

xii

Page 13: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Figure 4-18. Knockdown of IGF1R rescues gene expression of ITGB3 in PRKCZ-expressing cells................................................................................................................144 Figure 4-19. IGF1 increases ITGB3 transcript expression in PRKCZ-expressing SKOV3 cells..................................................................................................................................145 Figure 4-20. IGF1 stimulation decreases IGF1R gene expression in SKOV3 cells........147 Figure 4-21.TIMP-1 gene expression decreases in PRKCZ-expressing ovarian cancer cells..................................................................................................................................148 Figure 4-22. TIMP-1 gene expression regulation is independent of ITGB3 gene expression in SKOV3 cells..............................................................................................149 Figure 4-23. Effect of IGF1 on TIMP-1 transcript expression in SKOV3 cells..................................................................................................................................152 Figure 4-24. Effect of IGF1 signaling on SKOV3 migration as observed by wound healing assay....................................................................................................................153 Figure 4-25. Effects of IGF1 and EGF on migration of SKOV3 as determined by transwell migration assay................................................................................................154 Figure 4-26. Effect of ITGB3 on SKOV3 parental cells migration as observed by wound healing assay....................................................................................................................155 Figure 4-27. Proposed model of ITGB3 transcriptional regulation through IGF1 signaling in PRKCZ-expressing SKOV3 cells................................................................................164 Figure 5-1. Potential Significance of HNF4A in familial ovarian cancer as suggested by Gene Set Analysis (GSA) of gene expression microarray data.......................................180

xiii

Page 14: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

LIST OF ABBREVIATIONS aCGH Array comparative genomic hybridization ACHE Acetylcholinesterase ADH4 Alcohol dehydrogenase 4 (class II), pi polypeptide APC Adenomatous polyposis coli AKT v-akt murine thymoma viral oncogene homolog AR Androgen receptor ATM Ataxia telangiectasia mutated AXIN Axin BACH2 Basic leucine zipper transcription factor 2 BCL2 B-cell CLL/lymphoma 2 BIRC3 Baculoviral IAP repeat-containing 3 BRAF v-Raf murine sarcoma viral oncogene homolog B1 BRC BRCA1 Breast cancer 1, early onset BRCA2 Breast cancer 2, early onset BRCT BRCA1 C Terminus BrdU Bromodeoxyuridine CA-125 Cancer antigen-125 CASP8 Caspase 8 CCND1 Cyclin D1 CCNE1 Cyclin E1 hCDC4/FBXW7 F-box and WD repeat domain containing 7 CDK Cyclin-dependent kinases CDKN1B Cyclin-dependent kinase inhibitor 1B (p27, Kip1) CDKN2A Cyclin-dependent kinase inhibitor 2A CGH Comparative genomic hybridization CIMBA Consortium of Investigators of Modifiers of BRCA1/2 CTNNB1 Catenin (cadherin-associated protein), beta 1 CUL1 Cullin 1 CYP11A1 Cytochrome P450, family 11, subfamily A, polypeptide 1 DAPI 4',6-diamidino-2-phenylindole DCC Deleted in colorectal carcinoma EGF Epidermal growth factor EGFR/ERBB1 Epidermal growth factor receptor EMT Epithelial-mesenchymal transition EPHX1 Epoxide hydrolase 1, microsomal (xenobiotic) ER Estrogen receptor ERK Elk-related tyrosine kinase EST Expressed sequence tag EVI1 Ecotropic viral integration site-1 FAT4 FAT tumor suppressor homolog 4 FBS Fetal bovine serum FISH Fluorescence in situ hybridization FGF Fibroblast growth factor

xiv

Page 15: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

FTE Fallopian tube epithelial cells GFP Green fluorescent protein GSK3β Glycogen synthase kinase 3 beta GTP Guanosine triphosphate H3 Histone 3 HBOCS Hereditary breast-ovarian cancer syndrome HER2/ERBB2 v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 HGF Hepatocyte growth factor HNF4A Hepatocyte nuclear factor 4, alpha HNPCC Hereditary non-polyposis colorectal cancer HPRT1 Hypoxanthine phosphoribosyltransferase 1 HR Homologous recombination IGF1 Insulin-like growth factor 1 IGF1R Insulin-like growth factor 1 receptor IHC Immunohistochemistry IPA Ingenuity Pathway Analysis ITGB3 Integrin beta 3 c-JUN Jun proto-oncogene KRAS v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog LBR Lamin B receptor LOH Loss of heterozygosity LPA2 Lysophosphatidic acid receptor 2 LRMP Lymphoid-restricted membrane protein MAGI3 Membrane associated guanylate kinase MAL Mal, T-cell differentiation protein MAPK Mitogen-activated protein kinase MCM5 Minichromosome maintenance complex component 5 MDM2 Mdm2 p53 binding protein homolog MET Met proto-oncogene (hepatocyte growth factor receptor) hMLH1 MutL homolog 1, colon cancer, nonpolyposis type 2 MMP Matrix metallopeptidase hMSH2 MutS homolog 2, colon cancer, nonpolyposis type 1 hMSH6 MutS homolog 6 MRPL19 Mitochondrial ribosomal protein L19 MSP Methylation-specific PCR MTOC Microtubule organization centre mTOR Mechanistic target of rapamycin (serine/threonine kinase) MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide MTUS1 Mitochondrial tumour suppressor 1 MYC v-Myc myelocytomatosis viral oncogene homolog NCOR1 Nuclear receptor co-repressor 1 NEO1 Neogenin 1 NF-κB Nuclear factor kappa B NQO1 NAD(P)H dehydrogenase, quinone 1 OCCR Ovarian cancer cluster region OSE Ovarian surface epithelial cells

xv

Page 16: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

OVCAS Ovarian cancer cells P53 (TP53) Tumour protein 53 PAK1 p21 protein (Cdc42/Rac)-activated kinase 1 PARK2 Parkin 2 PDCD4 Programmed cell death 4 PI3K PI3Kinases PIK3AP1 Phosphoinositide-3-kinase adaptor protein 1 PIK3CA Phosphoinositide-3-kinase, catalytic, alpha polypeptide PIP3 Phosphoinositide-3,4,5-trisphosphate hPMS1 Postmeiotic segregation increased 1 hPMS2 Postmeiotic segregation increased 2 PMT Photomultiplier tube PRKCI Protein kinase C iota PRKCZ Protein kinase C zeta PTEN Phosphatase and tensin homolog PTT Protein truncation test RAD51 RAD51 homolog (RecA homolog, E. coli) RB1 Retinoblastoma 1 RBAK RB-associated KRAB zinc finger RBX1 Ring-box 1, E3 ubiquitin protein ligase RHOA Ras homolog gene family, member A RT-PCR Reverse transcriptase polymerase chain reaction RUNX1T1 Runt-related transcription factor 1 SCF Skp, Cullin, F-box containing complex siRNA Small interfering RNA SIRT3 Sirtuin 3 SKP1 S-phase kinase-associated protein 1 SKY Spectral karyotyping SNP Single nucleotide polymorphism SRC v-Src sarcoma SSCP Single strand conformation polymorphism TBP TATA box binding protein TGFβ Transforming growth factor beta TGFβ1 Transforming growth factor beta 1 TMA Tissue microarray TMR Tetramethylrhodamine TUNEL Terminal deoxynucleotidyl transferase dUTP nick end labeling UHN University Health Network, Toronto, ON UPD Uniparental disomy UPS Ubiquitin-proteosome system VNTR Variable number of tandem repeat VTN Vitronectin XPA Xeroderma pigmentosum, complementation group A YWHAZ Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase

activation protein, zeta polypeptide

xvi

Page 17: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

CHAPTER 1

Introduction & Literature Review

1

Page 18: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

2

1.1 Principles of Molecular Cancer Genetics Major advances have been made over the years in our understanding of different

human diseases, including cancer; however, the underlying mechanisms responsible for

the development of these diseases remain elusive due to the complexity of genetic and

environmental interactions that are involved.

Mutations in cancer cells consist of a wide range of genetic alterations, including

large-scale DNA copy number alterations, chromosomal translocations, amplifications

and deletions, as well as more subtle changes in nucleotide sequences such as point

mutations at positions that are critical for protein functions (1). Additionally, heritable

epigenetic aberrations such as DNA methylation, histone modification, nucleosome

repositioning, and posttranscriptional gene regulation by micro-RNAs may also alter

gene expressions in cells, leading to inappropriate silencing or activation of cancer-

associated genes, and these can occur at various phases of cancer development (1, 2).

Indeed, accumulated evidence suggests that both genetic and epigenetic modifications are

important contributors that can lead to deregulation of molecular pathways responsible

for carcinogenesis.

Genetic aberrations in cancers can be acquired somatically or inherited from one

or both of the parents through the germline, followed by additional acquisition of somatic

mutations. These types of cancer are thus termed “sporadic” or “hereditary”,

respectively. Since the first rate-limiting step of mutation acquisition is bypassed in

hereditary cancers, it often results in multiple cancer types, and the age of onset for these

cancer patients is generally earlier than their sporadic counterparts.

Page 19: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

3

As described by Hanahan and Weinberg in their classic review of hallmarks of

cancer, there are six features necessary for a cell to develop a cancer phenotype: self-

sufficiency in growth signals, evasion of apoptosis, insensitivity to growth-inhibitory

signals, limitless replicative potential, sustained angiogenesis, and tissue invasion and

metastasis (3). And in their more recent review, they described two additional hallmarks

of cancer: reprogramming of energy metabolism and evading immune destruction (4)

(Figure 1-1). The identification of genes involved in each of these processes is important

in gaining a better understanding of cancer development and progression. Thus far, the

most well-described cancer progression model is found in colon cancer, whereby

mutations in APC, KRAS, DCC, and p53 are associated with a defined series of stages

from normal colonic mucosa to colorectal carcinoma (5, 6). However, clarification of

this type of cancer progression model is still required for other cancer types. Regardless,

the group of cancer susceptibility genes that play significant roles in tumour initiation and

progression can be generally classified into three main categories: gatekeepers,

caretakers, and landscapers (7).

1.1.1 Gatekeepers

Gatekeepers are genes that can directly regulate tumour cell expansion and

comprise tumour suppressor genes and oncogenes (8, 9). Mutations or deregulations of

these genes can drive the neoplastic process by stimulating cell growth or reducing cell

death.

Page 20: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

4

Figure 1-1. Hallmarks of Cancer. Esential alterations in cell physiology important in malignant growth of cancer cells (Adapted from Hanahan and Weinberg, 2000, 2001, ref (3, 4)).

Evading apoptosis Self-sufficiency in

growth signals

Insensitivity to

anti-growth signals

Tissue invasion &

metastasis Sustained

angiogenesis

Evading immune

destruction Limitless replicative

potential

Reprogramming of

energy metabolism

Page 21: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

5

1.1.1.1 Tumour Suppressor Genes Tumour suppressor genes are involved in the regulation of cellular growth and

differentiation. Most tumour suppressors act in a recessive manner, meaning that loss or

inactivation of both copies of a gene is required for cellular transformation, a theory

known as “Knudson’s two-hit hypothesis” (10) (Figure 1-2). This concept first emerged

when Knudson performed a statistical analysis of retinoblastoma incidence in children

and observed that the inherited form of retinoblastoma occurs earlier than its sporadic

counterpart (9). This can be explained by the fact that the initiation of a tumour involves

two-rate limiting steps, of which two losses or inactivations of tumour suppressor genes

are required, and that in inherited forms of cancers, the first loss is already present in the

germline. However, recent studies have also suggested that some tumour suppressor

genes may still be able to confer a growth advantage upon a cell when only one allele is

inactivated, a condition known as haploinsufficiency (11).

Loss or inactivation of a tumour suppressor gene can occur through different

mechanisms, including point mutation, deletion, mitotic recombination, and

chromosomal loss (12). Additionally, epigenetic alterations can also decrease the

expression of these genes without alteration of their underlying DNA sequence, through

cancer-specific CpG island hypermethylation, in combination with repressive histone tail

modification (13).

Page 22: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

6

Figure 1-2 Knudson's two-hit hypothesis. Normal individuals have two normal copies of a tumour suppressor gene, thus two independent mutational events “hits” are required for a cell to initiate cancer. However, individuals with inherited germline mutation already have the first “hit” in every cell, thus only one additional mutation is required to initiate cancer. (Adapted from Richards, 2001, ref (14)).

Normal allele

Inherited germline mutation

Deletion Normal Inherited mutation

Page 23: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

7

1.1.1.2 Oncogenes

Proto-oncogenes encode proteins that are normally involved in the stimulation of

cell division; however, when these genes are genetically altered, they are capable of

causing cellular transformation and uncontrolled proliferation (15). Oncogenes (proto-

oncogenes with gain-of-function mutations) are considered to be dominant transforming

genes because changes in only one allele of the gene are usually sufficient to confer a

selective growth advantage on the cell. These genes are frequently activated by gain of

function mutations or fusions with other genes, or are abnormally over-expressed due to

gene amplification, increased promoter activity, or protein stabilization (16). Many

oncogenes have been identified and they all typically act through three biochemical

mechanisms: protein phosphorylation (e.g. Raf kinase), transcription regulation (e.g.

Myc), and signal transmission (e.g. Ras) (17-19).

1.1.2 Caretakers

Caretaker genes (or stability genes) function in maintaining genomic integrity of

the cell by regulating DNA repair, chromosome segregation, and cell cycle checkpoints

(8). Some well-characterized caretaker genes include hMLH1, hMSH2, ATM, XPA,

BRCA1 and BRCA2, all of which play important roles in DNA repair (20). When

deregulated, these genes can indirectly promote neoplastic transformation by contributing

to an accumulation of mutations in oncogenes and tumour suppressor genes (8, 21). The

importance of these genes is affirmed in that their mutations can lead to a variety of

cancer-prone chromosomal instability disorders, including ataxia telangiectasia,

Page 24: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

8

Nijmegen breakage syndrome, hereditary nonpolyposis colorectal cancer, Fanconi

anemia, Li-Fraumeni, and hereditary breast/ovarian cancer (21).

Caretaker genes are also labelled as tumour suppressor genes because inactivation

of both of their alleles is required for a pathological phenotype to manifest. However, it

is interesting to note that while the risk of cancer in families with inherited mutations of

caretaker genes is less than the risk in families with inherited defects in a gatekeeper

gene, the most common forms of hereditary cancer dispositions, such as colon and breast

cancers, are caused by inherited mutations of caretaker genes rather than gatekeepers (8,

20).

1.1.3 Landscapers

Landscapers are genes that do not directly regulate cellular growth, but are instead

tumour modifiers that can provide an abnormal stromal environment to increase a cell’s

ability to transform (22). As such, landscaper genes can be classified as tumour

suppressor genes but they act on the tumour microenvironment rather than the tumour

itself. Stromal-epithelial interactions are important in the regulation of tissue

homeostasis, and disruptions of these interactions may lead to tumour formation, by

increasing proliferation and transdifferentiation of fibroblasts, infiltration and activation

of inflammatory cells, induction of angiogenesis and altered deposition and degradation

of the extracellular matrix (23).

It is controversially speculated that genetic alterations in either the epithelial or

the stromal cells can lead to altered stromal-epithelial interactions to promote

tumourigenesis, and there are evidence suggesting that inherited or acquired genetic

Page 25: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

9

alterations in stromal cells may give rise to cancer. For example, loss of heterozygosity

(LOH) in stromal cells has been described in various types of cancer, including breast,

ovarian, colon, lung, and head and neck carcinomas (24). Mutations of tumour suppressor

genes (eg. p53, PTEN) and oncogenes (eg. EGFR) have also been reported in the stromal

cells of breast carcinomas, and the frequency of these genetic changes differ between

sporadic and hereditary tumours, implying that these changes are specific to tumour

subtypes (25-27). In addition to LOH and mutations, epigenetic alterations such as DNA

methylation have also been found in tumour-associated stromal fibroblasts from

neoplastic human breast and prostate carcinomas (24). However, as mentioned above,

this remains controversial and the potential mechanisms responsible for these alterations

will require further investigations.

As mentioned earlier, genetic mutations are the driving force for cancer

development, by promoting tumour initiation and progression. At the cellular level, it is

a multistep process in which mutations can lead to a selection of cells that have acquired

advanced proliferation, survival, and metastatic potentials. Both mutational inactivation

of stability genes and tumour suppressor genes, and mutational activation of proto-

oncogenes to oncogenes, as well as additional intrinsic and extrinsic genetic changes, are

important for these acquisitions.

1.2 Ovarian Cancer

Ovarian cancer is the fifth most common form of cancer in women and it is the

leading cause of death in patients with gynaecologic malignancies in North America (28).

Page 26: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

10

In Canada alone, approximately 2500 new cases of ovarian cancer and 1750 deaths due to

this disease were estimated for 2009 (29). The lack of any obvious symptoms that would

indicate an early stage of this disease is responsible for this high mortality rate; in fact,

approximately 70% of the patients are diagnosed in an advanced stage after the cancer

has metastasized beyond the ovaries (30, 31).

The lack of effective screening methods also contributes to the late diagnosis of

ovarian cancer. Measurement of serum cancer antigen-125 (CA-125) is the primary

screening method used for ovarian cancer; however, CA-125 concentration does not have

the sensitivity nor specificity required for accurate detection, since its level is typically

low in early stages of ovarian cancer and it can also be elevated in other gynaecological

conditions, such as endometriosis, pregnancy, adenomyosis and polycystic ovarian

syndrome (32). To increase detection accuracy, transvaginal ultrasonography (TVS) is

often used in parallel with a CA-125 serum test; however, the specificity achieved using

the combination of these two methods remains sub-optimal (33). After being diagnosed,

ovarian cancer patients are often treated with aggressive surgery followed by

combination chemotherapy, but despite high initial response rates to treatment, survival

of patients stands at just 45% at 5 years, with most of the patients eventually relapsing

and succumbing to this disease (31, 34).

There are three main types of ovarian tumours: epithelial, germ cell, and stromal

tumours. Epithelial tumours are derived from cells that cover the surface of the ovary,

germ cell tumours are derived from cells that produce the ova, and stromal tumours arise

from the connective tissues that hold the ovaries together (35, 36). Of all of these tumour

types, epithelial tumours are the predominant type, accounting for 90% of total cases of

Page 27: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

11

ovarian cancer (35). Epithelial ovarian cancer is further subdivided into different

histological types, including serous, mucinous, endometrioid, and clear cell; of these

subtypes, serous carcinomas constitute the majority of ovarian carcinomas (35). Each of

these subtypes has unique morphology, as well as biological and genetic backgrounds,

and these differences imply that patients with different subtypes of ovarian cancer can

have different responses to the same treatment, as well as different prognosis of the

disease (37). Therefore, it is important to understand the molecular pathogenesis for

each type of ovarian carcinoma in order to develop effective screening methods and

treatment options for each of the patients.

1.2.1 Model of Ovarian Carcinogenesis

In a carcinogenesis model proposed by Shih and Kurmen (2004), epithelial

ovarian cancers are categorized into Type I and Type II tumours corresponding to two

main pathways of ovarian tumourigenesis (38) (Table 1-1). Type I tumours arise in a

stepwise manner from borderline tumours, and are composed of low-grade serous

carcinomas, mucinous, endometrioid, and clear cell carcinomas; Type II tumours, on the

other hand, arise de novo and include high-grade serous carcinoma, malignant mixed

mesodermal tumours (carcinosarcomas), and undifferentiated carcinomas (38).

The tumourigenic pathway of Type I tumours resembles the adenoma-carcinoma

progression in colorectal cancer in that these tumours tend to evolve slowly and are

associated with distinct molecular changes during tumour development, whereas Type II

tumours are highly aggressive, and are often characterized by genomic instability, as

shown by genome-wide changes in DNA copy number (33, 38, 39). These two types of

Page 28: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

12

tumours have distinct morphological and molecular signatures; Type I tumours often

contain mutations in BRAF, KRAS, PTEN and CTNNB1 and Type II tumours frequently

harbour p53 mutations (38). It should be noted, however, that while the majority of high-

grade serous carcinomas – the most prevalent form of ovarian cancer – arise

independently from low-grade tumours, there are rare cases of high-grade carcinomas

that have progressed from atypical proliferative serous (borderline) tumours (40) (Figure

1-3).

Page 29: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

13

Table 1-1. Characteristics of Type I and Type II ovarian tumours. Type I and Type II ovarian cancers have distinct morphological and molecular characteristics. Type I tumours evolve slowly and are associated with distinct molecular changes while most of Type II tumours arise rapidly and are highly aggressive.

Type I Type II

Tumour Type - micropapillary serous carcinoma

- mucinous

- endometrioid

- clear cell

- serous carcinomas

- malignant mixed mesodermal tumours (carcinosarcomas)

- undifferentiated carcinomas

Mutations - KRAS

- BRAF

- PTEN

- β-catenin (CTNNB1)

p53

Chromosomal Instability

gradual increase high

Confined to ovary yes no

Page 30: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

14

Figure 1-3. Dualistic model of ovarian serous carcinoma development. Ovarian serous carcinomas, the most common type of ovarian cancer, are proposed to arise through either Type I or Type II pathway. Low-grade tumours develop in a stepwise manner and are often associated with KRAS or BRAF mutations. High-grade tumours develop directly from the ovarian surface epithelium or inclusion cysts without distinct intermediate morphological stages. High-grade serous carcinomas frequently harbour p53 mutations, and are associated with chromosomal instability. (Adapted from Shih and Kurmen, 2004, ref (38)).

?

Page 31: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

15

1.2.2 Familial Ovarian Cancer

There are a number of well established risk factors associated with ovarian

cancer. Factors that lower the number of lifetime ovulations in an individual have been

shown to decrease her risk of developing ovarian cancer; these factors include the use of

oral contraceptive pills, multiparity, breast-feeding, and oophorectomy (41, 42).

Likewise, there are factors that have been shown to increase the risk of developing

ovarian cancer, including old age, early menarche, late menopause, high dietary fat

intake, and use of estrogen-replacement therapy (43). However, the single most

important risk factor for ovarian cancer is family history (44). In fact, it is estimated that

familial ovarian cancer accounts for 5-15% of the total cases of ovarian cancer (44). The

risk of developing ovarian cancer is 1.6% of women in the general population; however,

if a woman has one or two first-degree relative(s) with ovarian cancer, her risk increases

to 4% and 7%, respectively (45). Notably, the age of onset for ovarian cancer patients

with a family history is earlier than those with no family history, with an average age of

53.5 and 60.8, respectively (46).

Three main hereditary syndromes that predispose to ovarian cancer, including

hereditary site-specific ovarian cancer, hereditary breast-ovarian cancer syndrome

(HBOCS), both of which are due to mutations in the tumour suppressor genes BRCA1

and BRCA2, and hereditary non-polyposis colorectal cancer (HNPCC; Lynch Syndrome),

which is mainly due to mutations in DNA mismatch repair genes such as hMSH2,

hMLH1, hMSH6, and PMS2 (47, 48). Another group of minor familial syndromes also

predispose individuals to ovarian cancer, accounting for <1% of total cases, including

Page 32: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

16

Gorlin’s syndrome, osteochondromatosis or Ollier’s syndrome, and the Peutz-Jeghers

syndrome (47, 49-51).

1.2.2.1 Hereditary Site-Specific Ovarian Cancer and Hereditary Breast-Ovarian

Cancer Syndrome (HBOCS)

Site-specific ovarian cancer syndrome is identified in families in which two or

more first- or first- and second-degree relatives are affected by epithelial-type ovarian

cancer, without being affected by breast cancer (45). While it has previously been

speculated that site-specific ovarian cancer represents a unique syndrome, it has been

reported that it may in fact be a variant of HBOCS with a high prevalence of ovarian

cancer, since no susceptibility gene has been identified specifically for ovarian cancer

(52).

As its name implies, hereditary breast-ovarian cancer syndrome is identified in

families in which both breast and ovarian cancer are common. This syndrome is

characterized by early-onset breast cancer, ovarian cancer at any age, bilateral breast

cancer, breast and ovarian cancer in the same individual, or male breast cancer (45).

Genetic predisposition is suggested by early onset, as well as multiple cases of these

cancers within the same family. Thus far, the two well-studied high penetrance

susceptibility genes found to be associated with ovarian cancer are BRCA1 and BRCA2,

both of which lead to autosomal dominant inheritance of susceptibility (45). The

importance of these two genes in ovarian cancer will be discussed further in this chapter.

Page 33: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

17

1.2.2.2 Hereditary Nonpolyposis Colorectal Cancer (HNPCC)

Familial ovarian cancer may also occur in individuals with hereditary

nonpolyposis colon cancer syndrome, which is characterized by an autosomal dominant

inheritance of colonic cancer in the absence of colonic polyposis (53). In addition to

ovarian cancer, individuals within HNPCC families also have higher risk for other

cancers such as endometrial, uro-genital, pancreatic and biliary tract cancers (45). Over

70% of the mutation carriers among HNPCC families have mutations within the DNA

mismatch repair (MMR) hMLH1 and hMSH2 genes. Mutations in hPMS1, hPMS2 and

hMSH6 are not as prevalent, but also have high penetrance (54-56). All of these MMR

genes are important for the repair of nucleotide mismatch during DNA replication to

prevent propagation of potentially harmful mutations (57). The cumulative risk for

colorectal and ovarian cancer for carriers of MMR gene mutations from HNPCC families

is estimated to be 80% and 12%, respectively (52).

1.2.3 Molecular Pathology and Genetics of Ovarian Cancer

Ovarian cancer is a heterogeneous disease with different histological grades and

subtypes, each encompassing a distinct, though not necessarily unique, set of molecular

genetic attributes. Over the years, extensive research has been conducted to identify

these genetic aberrations in order to improve our understanding of ovarian cancer

pathogenesis, ovarian tumour classification, as well as to develop personalized therapies

that target specific defects in the tumour cells of patients.

Page 34: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

18

1.2.3.1 p53

One of the genes most consistently associated with ovarian pathogenesis is p53, a

well-defined tumour suppressor gene. p53 encodes a DNA-binding protein that responds

to external cues and insults and is responsible for the transcriptional regulation of genes

involved in cell cycle control, DNA repair, and apoptosis of damaged cells (58).

Mutations that lead to loss of p53 function result in failure to activate responses, thus

leading to unrepaired genetic damage and increased chromosomal instability (59). It is

the most commonly mutated gene in human cancer and its mutations are observed in

more than 50% of high-grade ovarian serous carcinomas, but p53 mutation is a rare event

in low-grade tumours, which supports the idea that different pathogenic pathways are

responsible for high- and low-grade serous carcinomas (58, 60). It has also been

suggested that p53 mutation is an early event in the development of high-grade

carcinomas, as mutation of this gene has been observed in normal-appearing epithelium

and dysplastic epithelium within inclusion cysts next to the tumour that has the same

mutation (58).

1.2.3.2 Wnt-Signalling Pathway

Missense mutations of CTNNB1, which codes for β-catenin and maps to

chromosome 3p21, are observed in approximately 30% of endometrioid adenocarcinomas

(61). β-catenin is an important player within the canonical Wnt signalling pathway

known to be involved in various cellular processes, including regulation of cell fate,

proliferation, motility, and survival; indeed, constitutive activation of Wnt signalling is

often observed in endometrioid ovarian cancer (39, 62). Mutations of CTNNB1 often

Page 35: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

19

result in alterations in its protein residues such that it is no longer subjected to

phosphorylation by its upstream regulator GSK3β (glycogen synthase kinase 3 beta), thus

leading to stabilization of β-catenin (39). This stabilization allows for an accumulation of

β-catenin to constitutively activate the transcription of downstream target genes that are

important for neoplastic transformation and tumour progression (39). In addition to

CTNNB1, other defects within the Wnt pathway are also observed in ovarian

endometrioid tumours, including mutations of APC, AXIN1 and AXIN2, which all encode

components of a protein complex involved in the regulation of β-catenin (63).

1.2.3.3 PI3K/Akt Signalling Pathway

The PTEN tumour suppressor gene is also commonly mutated in endometrioid

carcinomas, with a frequency of 30-80% (64). Moreover, it is located on chromosome

10q23, a region that is lost in 20-30% of ovarian cancers of this specific subtype (64).

PTEN is one of the key regulators within the PI3K/Akt signalling pathway, as it can

dephosphorylate the plasma membrane lipid second messenger phosphoinositide-3,4,5-

trisphosphate (PIP3) generated by PI3Kinases (PI3K) to PIP2, leading to inhibition of

this signalling cascade (39). Inactivating mutations of PTEN can therefore enhance the

activation of PI3K/Akt pathway, leading to uncontrolled cell cycle progression, cell

survival, cell motility, and angiogenesis (39). Activating mutations are also observed in

PIK3CA, a potential ovarian oncogene that encodes the p110α catalytic subunit of PI3K

(39). While mutations of PIK3CA are only found in approximately 2% of ovarian serous

carcinomas, it is a more common event in endometrioid and clear cell subtypes, with a

frequency of approximately 20% (65). Interestingly, amplifications of PIK3CA (>7

Page 36: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

20

fold) are observed across all histological subtypes (24.5%) and are inversely correlated

with gene mutations (65). Additionally, amplification of PIK3CA has also been

associated with chemotherapy resistance in ovarian cancer patients (66).

The PIK3CA downstream target gene AKT2 has also been examined as a potential

ovarian cancer oncogene. It encodes a serine-threonine kinase that is able to

phosphorylate a variety of proteins, including Ezrin, a protein with roles in cell adhesion,

migration, and organization (67). High-level amplification (>3 fold) of AKT2, but not its

related genes AKT1 and AKT3, has been observed in high-grade ovarian serous

carcinoma; this specific amplification suggests an important role of AKT2 in ovarian

carcinogenesis (48, 68). Amplification/over-expression of this gene has been shown to

have a statistically significant association with higher grade tumours and poorer survival

(69). Interestingly, AKT2 over-expression and a loss of PTEN expression function

synergistically to promote metastasis in colorectal cancer (70). While this has yet to be

examined, it is possible that the same may be observed in ovarian cancer, since loss of

PTEN expression has also been found in a subset of ovarian tumours, as mentioned

above.

1.2.3.4 MAPK Signalling Pathway

The RAS family of G proteins belongs to several signalling pathways, including

the well-studied MAPK (mitogen-activated protein kinase) signalling cascade. Their

function involves coupling membrane receptor kinases to intracellular signalling cascades

through their GTPase activity, and they are critical players in the regulation of cellular

proliferation (71). Point mutations within codons 12, 13, or 61 of KRAS, which result in

Page 37: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

21

a constitutively activated protein, are important for ovarian carcinogenesis (72). While

KRAS mutations are rare in invasive serous epithelial ovarian cancers, they do occur

frequently in mucinous ovarian cancers (50%), and are common events in serous

borderline ovarian tumours (33%) and low-grade serous carcinomas (35%) (58, 73).

Activating mutations of BRAF within codon 599 are also often observed in

borderline and low grade serous tumours, with similar frequencies as KRAS mutations

(58). BRAF is a RAF family protein, and is activated by RAS to stimulate the MAPK

signalling cascade. Interestingly, KRAS and BRAF mutations are not found in the same

tumour, which may be explained by their closely related functions in the pathway (58).

Also, the observed differences in KRAS and BRAF mutation status between histological

subtypes indicate that the MAPK pathway may play a major role in pathogenesis in

certain but not all types of ovarian cancer.

1.2.3.5 Cell Cycle Genes

Correct control of the cell cycle is critical in the regulation of cell proliferation,

and it requires proper expression of various regulatory proteins, including cyclins and

cyclin-dependent kinases (CDK). Cyclins function in activating CDKs, which

subsequently phosphorylate and activate key proteins to allow cell cycle progression. In

human cancers, CCND1, which encodes cyclin D1, is most frequently over-expressed.

(74). Cyclin D1 forms a complex with CDK4/6, which can then phosphorylate and

activate the retinoblastoma protein (RB), leading to the release of E2F, triggering G1 cell

cycle progression (75) . While amplification of CCND1 is rarely observed in ovarian

cancer, it is found to be over-expressed in a subset of ovarian tumours, mostly in low-

Page 38: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

22

grade serous carcinomas and mucinous tumours, and is associated with decreased

survival of patients (58, 76, 77). Over-expression of CDK4 has also been observed in

approximately 15% of ovarian cancers and is reported to be correlated with increased

expression of CCND1; similarly to CCND1, gene amplification of CDK4 is a rarely

observed (78).

p16 acts as a negative regulator of the cell cycle by inhibiting the kinase activity

of the cyclin D-CDK4 complex (75). Loss of p16 mRNA is observed in ovarian cancer,

mostly in serous, mucinous, and endometrioid carcinomas (79). It has been proposed that

the decrease in p16 gene expression is due to hypermethylation at its 5’-CpG island, since

mutation and deletion of this gene are uncommon (79). It is interesting to note that the

lack of p16 expression occurs more frequently in ovarian tumours lacking p53 mutations,

suggesting that p53 inactivation may not be as important in ovarian tumour development

when another G1 cell cycle regulatory gene has already been inactivated (78).

The cyclin E-CDK2 complex is important for the progression of G1-S phase of

the cell cycle, and over-expression of cyclin E, which is encoded by the gene CCNE1,

has been found in various human cancers, including ovarian cancer (75). Evidence

suggests that this increase in gene expression is partially due to gene amplification and

that CCNE1 expression is correlated with CDK2 expression (80). CCNE1 over-

expression may be involved in malignant progression of epithelial ovarian cancer, as its

expression is highest among malignant tumours (~70%), compared with borderline

(~48%) and benign tumours (~9%) (81). Furthermore, cyclin E is negatively regulated

by p21 and p27, both of which are expressed at low levels in high-grade serous ovarian

Page 39: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

23

carcinomas (82). Notably, the potential of cyclin E as a prognostic marker has also been

suggested, since its over-expression is associated with poor disease outcome (75).

1.2.3.6 Epidermal Growth Factor Family Receptors

In the presence of ligand, epidermal growth factor family receptors can form

homodimers and heterodimers to initiate intracellular signalling pathways that are

important for cell proliferation and tumour growth (58). EGFR1/ERBB1 is a member of

the type I tyrosine kinase receptor family HER (i.e., ERBB) and is expressed in normal

ovarian surface epithelium (83). While this gene is rarely mutated or amplified, it is

often over-expressed in ovarian cancer, presenting in 35-70% of all cases, and is

associated with poor prognosis, as well as with drug resistance (82-84).

Another HER family member, HER2/neu (c-ERBB2), is amplified in ~6-18% of

ovarian tumours, and its increased copy number is associated with poor prognosis (84-

88). Additionally, HER2 over-expression is found in 20-30% of ovarian cancers (48).

Specifically, it is frequently observed in serous ovarian carcinomas, and tumours

associated with advanced stages, late age at diagnosis, and differentiation (89). HER2

over-expression can lead to its dimerization, even without the presence of a ligand, which

leads to constitutive activation of its signalling pathway (90). Moreover, it was found

that HER2 is over-expressed in 40% of HBOCS, demonstrating its importance within the

subset of ovarian cancer patients with a family history of breast and/or ovarian cancer

(48).

Page 40: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

24

1.2.3.7 Estrogen Receptors

Estrogens are involved in normal cellular processes such as growth,

differentiation, and physiology of the reproductive system; however, they can also

participate in the progression of hormone-dependent cancers, including ovarian cancer

(91). Estrogen receptor alpha (ER-α) is a member of the superfamily of nuclear receptors

that transduce hormone signals. It is a ligand-activated transcription factor that can

affect normal or cancer cells upon stimulation with estrogens (92). Amplification of the

ESR1 gene is a rare event in ovarian cancer, accounting for only 2.1% of the cases;

however, ESR1 has been found to be over-expressed in 25-86% of ovarian cancer cases

(93). Estrogen receptor beta (ER-β), on the other hand, is under-expressed in a subset of

ovarian cancers, with malignant tumours having the lowest expression (94). This

opposite expression of ER-α and ER-β observed in ovarian cancers may be correlated

with their roles in regulation of cyclin D1, since ER-α stimulates the gene expression of

cyclin D1, whereas ER- β suppresses its expression (95).

As stated earlier, ovarian cancer is a heterogeneous disease with various types of

genetic changes. And while the above-mentioned genetic alterations have all been shown

to play significant roles in the pathology of ovarian cancer, the most well characterized

genetic risk factors genes associated with ovarian cancer are evidently BRCA1 and

BRCA2.

Page 41: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

25

1.2.4 BRCA1 and BRCA2

In the early 1990’s, Hall and colleagues identified a linkage between chromosome

17q and site-specific breast cancer arising in young women in certain cancer prone

families in an unusually large proportion compared to the general population (96).

Shortly after, Narod et al. showed that this same genetic locus was linked to HBOCS

(97). The BRCA1 gene was subsequently cloned by Miki and colleagues in 1994,

followed by the identification of another breast and ovarian cancer susceptibility gene

appropriately named BRCA2, by Wooster et al. (98, 99).

Both BRCA1 and BRCA2 are expressed in a variety of tissues, particularly

during S and G2 phases of the cell cycle (100). They play major roles in DNA repair

mechanisms, by participating in homologous recombination (HR) in the presence of

double-strand breaks, transcription-coupled repair during oxidative damage, and possibly

in non-homologous end joining (100). Additionally, they are involved in the control of

cell cycle checkpoints, protein ubiquitination, and chromatin remodelling (101).

BRCA1 is located on chromosome 17q21 and it comprises 24 exons encoding a

protein of 1863 amino acids (98). BRCA1 contains a highly conserved N-terminal RING

domain and a tandem of two BRCT (BRCA1 C Terminus) domains at its carboxyl

terminus, which are important for protein ubiquitylation and phosphorylated-protein

binding, respectively (102, 103). These domains are very important for its protein

function, as demonstrated by the observations that these regions are often targets of

clinically important mutations (102).

BRCA2 is located on chromosome 13q12-13 and is made up of 27 exons encoding

a protein of 3418 amino acids (30). BRCA2 contains eight BRC repeats that are involved

Page 42: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

26

in protein binding, including recombinase RAD51, to function in DNA double strand

repair (104).

1.2.4.1 BRCA1 and BRCA2 in Ovarian Cancer

Approximately 90% of all the cases of hereditary ovarian cancer are due to

mutations within the BRCA1 or BRCA2 genes (34). Indeed, for those who have inherited

mutations within BRCA1 or BRCA2, their lifetime risks for developing ovarian cancer are

20-40% and 15-25%, respectively, compared to a lifetime risk of 1.6% in the general

population (45, 105).

While one defective copy of either BRCA1 or BRCA2 in the germline is enough to

increase cancer predisposition of an individual, it is often observed that the second allele

is also lost in tumour cells isolated from predisposed patients (106). This “second hit”

often occurs through LOH. In fact, BRCA1 and BRCA2 have reported LOH frequencies

of 80% and 70%, respectively, in mutation carriers in both breast and ovarian tumours

(107, 108). While BRCA promoter hypermethylation occurs in sporadic breast and

ovarian cancer, it is a rare event in mutation carriers (109, 110). In addition to LOH and

promoter methylation, other epigenetic or transcription silencing of BRCA1 and BRCA2,

such as chromatin-mediated repression, and other yet unknown environmental factor(s)

may affect BRCA expression at the gene level.

Page 43: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

27

1.2.4.2 Mutations of BRCA1 and BRCA2 in Ovarian Cancer

The type of mutations within BRCA1 and BRCA2 varies, with the majority of

them being small insertions or deletions resulting in frameshift, nonsense mutations or

splice site alterations, all of which may cause truncation of the protein (30). Missense

mutations resulting in dysfunctional proteins have also been reported, along with

mutations that involve non-synonymous coding changes or in-frame deletions (30). In

recent years, with the introduction of high-throughput DNA-based techniques, it has also

been discovered that large genomic deletions and rearrangements can occur; however,

large genomic alterations are far less common in BRCA2 compared to BRCA1 (111-114).

It has been reported that up to 75% of hereditary ovarian cancer families have a

mutated BRCA1 (115), and while BRCA2 mutations are found within 35% of hereditary

breast cancers, they confer a lower risk in ovarian cancer, accounting for 10-20% of

hereditary ovarian cancers (105). Despite the high frequency of BRCA gene mutations in

breast and ovarian cancer, there are no specific mutational hotspots, as mutations are

distributed throughout the whole gene (45). However, there are reports that showed an

association between the sites of mutation and ovarian cancer risk. In BRCA1, mutations

within nucleotides 2401 and 4190 (named “high risk region for ovarian cancer”) were

shown to result in increased risk of ovarian cancer while decreasing the risk of breast

cancer (30) (Figure 1-4). Likewise, mutations between nucleotides 4075 and 6503 of

BRCA2, termed “Ovarian Cancer Cluster Region (OCCR)”, also contribute to an increase

in ovarian cancer risk (116-118).

Page 44: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

28

Figure 1-4. Gene structure of BRCA1 and BRCA2. Coloured boxes represent exons

with corresponding exon number below each box. Common founder mutations are

indicated, as well as mutational regions associated with increased risk of ovarian cancer

(Adapted from Russo et al., 2009, ref (45))

BRCA1

BRCA2

High risk region for ovarian cancer

Ovarian Cancer Cluster Region (OCCR)

2401 4190

4075 6503

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27

185delAG ins6kb 5382inC

999del5 617delT

Page 45: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

29

A catalogue of BRCA1 and BRCA2 mutations can be found at the Breast-Cancer

Information Core (BIC) database (http://research.nhgri.nih.gov/bic/), an international

collaboration hosted by the NIH National Human Genome Research Institute. As of

2009, there were approximately 12,000 carriers of a BRCA1 mutation or unclassified

variant and approximately 11,000 BRCA2 carriers recorded (30).

1.2.4.3 Specific BRCA1 and BRCA2 mutations in closed populations

Highly penetrant germline BRCA mutations are rare, with an approximate

frequency of 1 in 500 individuals in most population. However, for certain relatively

closed and geographically confined populations, the frequency of BRCA mutations

increases dramatically. The Ashkenazi Jewish population in North America and Israel,

for example, has a BRCA mutation frequency of 1 in 40 (119). Numerous genetic studies

have focused on this population and specific “founder mutations” have been identified

within these individuals, including 183delAG and 5382inC in the BRCA1 gene and

6174delT in the BRCA2 gene (48). These three mutations alone account for 98-99% of

identified mutations in this population and screening for these mutations has become a

common clinical practice for individuals with Ashkenazi Jewish background (30).

Other groups have also implemented specific genetic screenings to identify

founder mutations that are prevalent in their respective populations in individuals with a

family history of breast and/or ovarian cancer. For example, most of the cases of familial

breast and/or ovarian cancer within the Icelandic population are due to a 999del5

mutation within the BRCA2 gene, while Eastern European countries such as Russia,

Poland, and Hungary have a high frequency of 5382inC BRCA1 mutation as well as a few

Page 46: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

30

other common mutations (30). In North America, French Canadian founder mutations

C4446T within BRCA1, as well as G6085T and 8765delAG within BRCA2 have also

been identified (120, 121). The identification of these founder mutations has facilitated

the development of effective genetic screening programs that allow identification of high

risk individuals for appropriate surveillance and informed treatment strategies.

1.2.4.4 Modifiers of BRCA1 and BRCA2

It is likely that both genetic and environmental factors can affect the penetrance of

BRCA gene mutations for ovarian cancer. Indeed, previous studies have attempted to

identify such factors. For example, in a study conducted within the Ashkenazi Jewish

population, a functional single nucleotide polymorphism in the promoter of the MDM2

gene was associated with an increased risk of breast and/or ovarian cancer among BRCA

carriers diagnosed with either one of these cancers before or at the age of 51 (122). And

in a Polish population study, it was found that the Leu33Pro polymorphism within the

ITGB3 gene increased the risk of BRCA1-associated ovarian cancer but not breast cancer

(123); however, subsequent genotyping of the ITGB3 gene in 9998 BRCA1 and 5544

BRCA2 carriers from 34 studies from CIMBA (Consortium of Investigators of Modifiers

of BRCA1/2) showed that when the Polish population is excluded from analysis, the

Leu33Pro polymorphism is no longer associated with the increased risk of ovarian cancer

(124), suggesting that this ITGB3 polymorphism may only be important in a subset of

BRCA1 carriers. Interestingly, a recent genome-wide association study conducted by

Ramus et al. identified 9p22.2 as a novel ovarian cancer susceptibility locus, as a rare

allele of this locus (rs3814113) was found to be associated with a decreased ovarian

Page 47: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

31

cancer risk in both BRCA1 and BRCA2 carriers (125). This is a significant finding as it is

the first confirmed common genetic variant that has been associated with reduced ovarian

cancer risk for either BRCA1 or BRCA2 carriers. However, mechanisms of how this SNP

may alter the risk of ovarian cancer in BRCA carriers remain to be investigated.

In terms of non-genetic/environmental factors, oral contraceptives, as well as

tubal ligation appear to significantly lower the risk of ovarian cancer in women with

BRCA germline mutations, as they do in the general population (126, 127). However,

while the discovery of genetic and environmental modifiers of BRCA1 and BRCA2 has

begun to emerge in recent years, the number of identified modifiers is still limited.

It appears that the development of ovarian cancer is a result of a multi-step

process involving an accumulation of genetic alterations. For people with family history

of ovarian cancer, these alterations could be inherited. In addition to high-penetrance

susceptibility genes BRCA1 and BRCA2, it is plausible that changes in low-penetrance

genes can increase an individual’s risk for ovarian cancer. Therefore, the identification

of these risk-altering genes may be useful in assessing patients with a family history of

breast and/or ovarian cancer.

1.3 Ovarian Cancer Genome-Wide Association Studies

The development of genomic technologies such as array comparative genome

hybridization (aCGH), single nucleotide polymorphism (SNP) arrays and microarray

expression profiling has allowed the elucidation of many important genetic events that

occur in cancer development. Their ability to simultaneously measure thousands of

genes not only allows researchers to identify individual genes but also to identify

Page 48: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

32

biological pathways that may be important in cancer development, including ovarian

cancer.

1.3.1 Array Comparative Genomic Hybridization of Ovarian Cancer

In addition to mutations at the nucleotide level, altering DNA copy number can

also affect gene expression and function of a cell. Certain DNA amplifications allow

cancer cells to increase expression of critical genes such as oncogenes involved in growth

regulation and genes responsible for drug resistance, while DNA deletions can cause a

decrease in expression of tumour suppressor genes. Researchers have employed various

cytogenetic methods such as fluorescence in situ hybridization (FISH), spectral

karyotyping (SKY), and conventional comparative genomic hybridization (cCGH) to

identify such aberrations in various diseases, including ovarian cancer; however, high

throughput array CGH (aCGH) has been used widely in recent years due its ability to

refine copy number alterations at a much higher resolution.

A study conducted by Lambros and colleagues identified regions of gains and

losses in 23 different ovarian cancer cell lines using aCGH, including those regions that

have been previously reported using other conventional methods, such as loss of

chromosome 4 or 4q, loss of 18q, and gain of 20 or 20q (128). Additional genomic

changes were detected in the study, including two regions of amplification on

chromosome 11q13 containing the cyclin D1 gene and candidate oncogene PAK1, as well

as amplification of 11q22 near the progesterone receptor gene, and a locus containing a

cluster of matrix metalloproteinase genes (128).

Page 49: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

33

This approach can be applied to ovarian tumours in addition to cell lines. For

example, characteristic patterns of copy number changes between histological subtypes

of ovarian tumours were identified using aCGH (Figure 1-5) (129). Every histological

subtype exhibited multiple copy number gains; however, it was found that their regions

of amplification differ, and that serous carcinomas have the largest number of alterations

(129). The authors of this study suggested that this observation is consistent with the

hypothesis that borderline ovarian tumours can progress to serous carcinomas, through

additional accumulation of genetic alterations (129). Differential patterns of recurrent

copy number alterations were also identified in sporadic and BRCA1-mutated ovarian

tumours by aCGH. As revealed by Leunen and colleagues, BRCA1 ovarian tumours

exhibited a greater number of losses, and that deleted regions are longer than those found

in the sporadic tumours, with a median length of 5.2 Mb vs. 0.2 Mb, respectively,

indicating that this major loss of genetic material in BRCA1 patients contributes to the

genetic instability of the tumours (130).

The application of aCGH also has diagnostic potential in clinical settings. For

example, because the genetic profiles of ovarian and endometrial tumours are presumably

different from each other, aCGH has been suggested to be used as a complementary tool

in distinguishing metastases from these two types of tumours to improve accurate

diagnosis (131). Additionally, regions associated with chemotherapy resistance in early-

stage epithelial ovarian cancer and late stage ovarian serous carcinomas have also been

identified using this method (132, 133), suggesting that aCGH may be used as a

screening tool, as the genetic profiles of the tumours may reveal whether or not the

patients can benefit from various therapeutic regimens.

Page 50: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

34

1.3.2 Single Nucleotide Polymorphism Array Analysis of Ovarian Cancer

The more recent development of single nucleotide polymorphism (SNP) arrays

has revealed additional information regarding the various types of chromosomal

aberrations that may occur in cancer. In addition to increased resolution, SNP arrays

differ from aCGH in that they can also detect the allele ratio of a single DNA sample,

hence allowing them to detect copy neutral loss of heterozygosity events such as

uniparental disomy (UPD), a genetic alteration resulting from deletion of one allele and

duplication of the second allele.

In fact, SNP arrays have been shown to be a useful tool in the identification of

such genetic alterations in ovarian cancer. For example, in their ovarian cancer SNP

array analysis, Walsh and colleagues have shown that BRCA-associated ovarian tumours

exhibit a greater frequency of amplification and LOH compared to sporadic ovarian

tumours, and that this increase in LOH is mostly due to UPD rather than deletion (134).

This observation of increased chromosomal instability in BRCA-associated tumours

further confirms the role of BRCA proteins in maintaining genomic integrity of a cell

(134). A more recent study conducted by Yoshihara et al. also showed that germline

copy number variation differs between BRCA1-associated ovarian tumours compared to

sporadic tumours (135). The authors showed that while BRCA1-associated tumours have

a higher number of deleted segments compared to sporadic tumours, the degree of

amplifications are lower in BRCA1-associated tumours when compared to sporadic

tumours (135).

Page 51: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

35

Figure 1-5. Chromosomal Aberrations in Ovarian Tumours. An example ideogram illustrating chromosomal changes in ovarian cancer as detected by conventional and array CGH (lines and dots, respectively). Red lines, chromosomal loss; green lines, chromosomal gains. (adapted from Mayr et al., 2006).

Page 52: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

36

High-resolution SNP arrays have also been used in the identification of novel

candidate tumour suppressor genes in ovarian cancer. By comparing 106 primary

ovarian tumours of various histological subtypes with matching normal DNA, Gorringe

et al. were able identify different LOH frequencies in different ovarian tumour subtypes

(136). This study confirmed previously identified homozygous deletions such as

CDKN2A (cyclin-dependent kinase inhibitor 2A), RB1 (retinoblastoma 1), and PTEN

(phosphatase and tensin homolog), and it allowed the identification of novel candidate

tumour suppressor genes near minimal regions of LOH on chromosomes 17, 13, 8p, 5q,

and X (136).

SNP analyses have been used to identify genes and their respective biological

processes that may be significant in ovarian cancer initiation and development. For

example, the importance of certain SNP alleles within xenobiotic metabolizing genes has

been recently examined in ovarian cancer (137). In the study, it was observed that

specific SNP alleles within the EPHX1 and NQO1 genes are associated with increased

serous ovarian cancer risk while a SNP within the ADH4 gene is associated with a

decreased risk, suggesting the importance of processing of pro-carcinogens in the

development of this disease (137). Another study comprising 19 study groups

participating in CIMBA revealed an association between the CASP8 D302H

polymorphism and decreased risk in breast and ovarian cancer in BRCA1 mutation

carriers but not BRCA2 mutation carriers, illustrating the importance of alterations within

certain apoptosis-associated genes in a subset of ovarian cancer patients (138).

The application of SNP arrays in clinical settings has also been explored in a

study that examined the accuracy and efficiency of this method in the screening of

Page 53: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

37

genetic variants associated with disease predisposition (139). Specifically, Monaco et al.

demonstrated that BRCA1-specific SNP arrays had both the sensitivity and specificity

needed for the detection of point mutations, insertions or deletions of any length, of

known and unknown variants that are associated with this gene (139), thus suggesting

that high-throughput SNP analysis may provide a powerful means to identify individuals

who may be predisposed to breast and/or ovarian cancer in a time and cost efficient way,

as compared to high-throughput sequencing.

1.3.3 Gene Expression Profiling of Ovarian Cancer

Numerous studies employing microarrays to determine gene-expression profiles

of ovarian carcinomas or ovarian cancer cell lines have been reported (140). Many of

these studies were conducted to identify diagnostic markers that may potentially be used

in clinical settings to improve early detection of ovarian cancer, since most patients

(>60%) are diagnosed at advanced stages of the disease (32). These were accomplished

by comparing the gene expression signatures of ovarian cancers with normal ovarian

epithelium. For example, a study profiling the gene expression patterns of primary

cultures of ovarian cancer specimens and primary cultures of normal ovarian epithelia

identified IL-8 and FGF-2 as potential serum-based diagnostic markers that can be used

along with CA-125, a commonly used marker (141).

Microarrays have also been applied to identify predictive/prognostic markers for

disease outcomes. By examining the molecular signatures of early-stage and advanced

ovarian cancer tumours with different outcomes, researchers are able to correlate gene

expression levels with survival of the patients. For example, the expression of MAL,

Page 54: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

38

which encodes a T-cell differentiation protein, has consistently been found to be

correlated with poor prognosis for ovarian cancer; indeed, its potential as a prognostic

indicator was further validated by detection of MAL protein by immunohistochemistry in

which high protein expression was associated with shorter survival in ovarian cancer

patients (142). In addition, due to the fact that many of the ovarian cancer patients

experience relapse within 6 months of chemotherapy (143), expression profiling has been

used to identify genes that may confer drug-resistance in ovarian cancer cells. By

comparing the expression signatures of paired ovarian tumours prior to and following

adjuvant chemotherapy, L’Espérance and colleagues identified a list of 121 genes that

were commonly up-regulated and a list of 54 genes that were down-regulated in post-

treatment tumours (144). These types of analyses have the potential to lead to assays to

help physicians in determining the appropriate treatment for patients according to their

ovarian cancer expression profiles.

Furthermore, to understand the biological mechanisms of ovarian cancer, studies

have been conducted to identify signalling pathways that may contribute to ovarian

carcinogenesis. In fact, alterations of different signalling pathways such as TGFβ, Myc,

Src, Rb/E2F, and β-catenin have been identified through ovarian cancer expression

microarray analyses (145, 146). Moreover, transcriptional profiling has been shown to be

able to distinguish between ovarian carcinomas of different tumour grades, thus giving

evidence that different pathways participate in the development of these tumours (82).

The site of origin of ovarian cancer remains controversial. The presence of occult

serous carcinomas in the fallopian tubes of BRCA mutation carriers undergoing

prophylactic surgery as observed from several studies has suggested the idea of distal

Page 55: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

39

fallopian tube epithelium as the source of putative precursors of tubal and ovarian serous

carcinomas (147). To further investigate this hypothesis, Tone et al. compared the gene

expression profiles of high-grade tubal and ovarian serous carcinomas, and non-

malignant fallopian tube epithelial cells from individuals with and without BRCA

mutations (FTEb and FTEn, respectively) (148). Similar global gene expression patterns

were observed between tubal and ovarian specimens, suggesting that fallopian tubal

epithelium is the cell of origin for both of these tumours (148), or that they arise fro cells

that share similar features. Interestingly, despite their histological similarity, the

expression profiles of FTEb differ from FTEn; in fact, FTEb exhibited gene expression

patterns similar to those of serous carcinomas, especially those from FTEb obtained

during the luteal phase of ovarian cycle, implicating that the changes in gene expression

found in BRCA mutation carriers in certain hormonal environments are likely to

contribute to an increased risk of malignant transformation (148).

Motamed-Khorasani and colleagues also sought to examine the early molecular

processes that may be involved in the development of BRCA-associated ovarian cancer,

by comparing the expression profiles of ovarian surface epithelial (OSE) cell cultures

derived from BRCA carriers and malignant ovarian cancer cells (OVCAS), to OSE

derived from control patients, based on previous observations that there is a loss of

coordinated androgen regulation in ovarian cancer cells and in non-malignant epithelial

cells derived from women who are carriers of BRCA1 or BRCA2 mutations (149, 150).

Upon continuous exposure to androgen, a total of 17 differentially expressed genes were

identified in OSE from BRCA carriers and OVCAS when compared to control OSE

(149). In particular, BACH2 (basic leucine zipper transcription factor 2) and ACHE

Page 56: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

40

(acetylcholinesterase) were found to be up-regulated in OSE from BRCA carriers as

compared to controls, and increased gene expression correlated with increased protein

expression in ovarian tumours, as shown by their immunohistochemical analysis from the

same study (149). Their observations therefore support the idea that altered androgen

response in BRCA mutation carriers could be involved in ovarian cancer susceptibility.

Given that published gene expression microarray datasets are publicly available to

the scientific community, researchers are able to gather information on ovarian cancer by

combining multiple studies from different laboratories, which may eliminate bias of

results due to different laboratory techniques, as well as increasing sample size. Indeed,

various research groups have sought to identify the differential gene expression patterns

in ovarian cancer by pooling data generated by different studies, in order to discover

biological pathways involved in ovarian tumourigenesis, as well as novel molecular

markers for better diagnosis and prognosis for the disease (151-154).

The above-mentioned expression microarray studies have begun to shed some

light on the cellular processes associated with hereditary-linked ovarian cancer.

However, much remains to be revealed regarding the various biological pathways that

may be involved in ovarian cancer progression in individuals with predispositions for the

disease. Therefore, the goal of my study is to examine and compare the expression

profiles of tumours from ovarian cancer patients with strong and weak family history of

breast and/or ovarian cancer, in order to identify biological pathways that may be

important in the progression of familial ovarian cancer.

Page 57: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

41

1.4 Hypothesis The hypothesis of the work presented in this thesis is that in addition to

deregulated BRCA1 and BRCA2, alterations in other signalling pathways are also

important for the progression of ovarian cancer in the subgroups of patients with different

family history of breast and/or ovarian cancer. Specifically, I hypothesize that ovarian

tumours from patients with a strong family history of breast and/or ovarian cancer have

molecular alterations that may be responsible for the earlier age of onset observed in this

group of patients compared to those with weak family history.

1.5 Rationale and Objectives Despite recent advances in the understanding of its pathology, ovarian cancer

remains one of the most lethal gynaecological cancers. While the roles of BRCA1 and

BRCA2 in ovarian cancer have been well studied, it is likely that polygenic expression

alterations, which can lead to deregulation of important signalling pathways, are also

responsible for ovarian cancer pathogenesis in the subset of patients with familial

predispositions. Moreover, alteration of certain signalling pathways may be responsible

for the earlier age of onset and tumour progression as seen in patients with strong family

history.

A microarray study conducted by Jazaeri et al. revealed that the expression

profiles of sporadic ovarian tumours do share similarities between the expression profiles

of either BRCA1- or BRCA2-related tumours, implying that common genetic pathways

are involved in sporadic and familial cases of ovarian cancer (155). However, when

compared to each other, distinct expression signatures of BRCA1- and BRCA2-related

Page 58: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

42

tumours were observed, suggesting that a diverse group of signalling pathways are

responsible for the development of ovarian cancer within familial cases of ovarian cancer

(155). Therefore, the examination and comparison of gene expression patterns among

patients with strong and weak family histories of breast and/or ovarian cancer will

identify candidate genes, as well as potential pathways that may be responsible for the

progression of familial ovarian cancer. Further investigation of these genes will provide

novel insights into the mechanisms involved, and this information may be useful for the

development of therapeutic tools.

In this thesis, a series of studies was conducted to investigate the hypothesis that

alterations in various signalling pathways are responsible for ovarian cancer progression

in patients with family history of breast and/or ovarian cancers.

The objectives of this thesis are:

i) To examine the molecular profiles of ovarian tumours from patients with family

history of breast and/or ovarian cancer, with the aim of identifying signalling pathways

that may be important in ovarian cancer pathogenesis in the group of patients with early

age of onset.

ii) To investigate the various mechanisms of hCDC alterations that may be involved in

familial ovarian cancer.

iii) To understand the role of PRKCZ in ovarian cancer progression by in vitro

biochemical and functional assays, and to identify its potential links to familial ovarian

cancer, by examining its relation to IGF1R and ITGB3, genes previously suggested to be

involved in BRCA-related breast and ovarian cancers.

Page 59: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

CHAPTER 2

Gene Expression Profiling of Familial Ovarian Cancer

The work presented in this chapter was performed by KS with the exception of the

microarray statistical analyses, which was performed in collaboration with biostatisticians

Dr. Shelley Bull, Dr. Dushanthi Pinnaduwage, and Sarah Colby.

43

Page 60: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

44

2.1 Introduction

As discussed in Chapter 1, the development of microarray technology has guided

the discovery of genes associated with various diseases, including ovarian cancer.

However, despite the plethora of ovarian cancer microarray studies, the global patterns

that distinguish groups of subjects with familial ovarian cancer have yet to be determined.

Additionally, while previous microarray studies have focused and compared the

expression profiles of BRCA1-linked and BRCA2-linked breast and ovarian tumours

(155, 156), it is possible that gene expression alterations in familial ovarian cancer

patients can occur independently of these two susceptibility genes, or that specific

changes in gene expression can affect both the BRCA1 and BRCA2 pathways.

In an effort to identify gene alterations that may play roles in familial ovarian

cancer progression, I utilized cDNA microarrays to examine the expression profiles of

ovarian cancer in patients with a strong or weak family history of breast and/or ovarian

cancer. In collaboration with biostatisticians Dr. Bull and Dr. Pinnaduwage, we

identified a subset of genes that were differentially expressed between the two subject

groups. Most of the functions of these genes have previously been shown to be important

in cancer development and progression, including roles in apoptosis, cell migration, cell

adhesion, and cell cycle regulation. I further interpreted the results by bioinformatics

analyses to identify the pathways and key molecules that may play potential roles in

ovarian cancer susceptibility and development.

Page 61: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

45

2.2 Materials and Methods

2.2.1 Ovarian Cancer Specimens

Twenty-seven flash-frozen high-grade serous epithelial ovarian cancer samples

with similar pathological characteristics were obtained from the Toronto Ovarian Tissue

Bank and Database. The tumour specimens were obtained from consenting patients

according to the institutional guidelines of the Research Ethics Board. Specimens were

selected based on two main criteria: patients had not received neo-adjuvant chemotherapy

prior to surgery in order to preserve molecular signature of ovarian tumours and each

sample contained at least 75% of tumour content as assessed by the surface area of

corresponding histology slides by pathologist, with no evidence of necrosis, to ensure

purity and quality of tumours.

2.2.2 Family History Classification

Tumours were ranked and classified under two categories based on patients’

family history data and age of diagnosis. The strength of family history was ranked as

followed: multiple first-degree relatives with ovarian cancer, multiple relatives with

ovarian cancer, single relative with ovarian cancer, multiple relatives with breast cancer,

single relative with breast cancer, and relative(s) with other types of cancer. Nine

tumours were classified as “strong familial” (mean age of 51), and 18 were classified as

“weak familial” (mean age of 62) (Table 2-1). BRCA1 and BRCA2 mutation status were

available for four of the patients (two BRCA1 mutation carriers, and two non-carriers),

but since the classification of tumours was solely based on family history, the mutation

status information was not used for the present study.

Page 62: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

46

Table 2-1. Characterization of the subjects in the strong versus weak family history groups. Classification of serous ovarian tumour samples according to the patients’ family history data. Selection of high grade tumours was based on similar pathological characteristics between the two subject groups. Refer Appendix A1 for detailed family history data.

Strong Familial Weak Familial

Total # of cases

9

18

Family history of ovarian cancer (# of affected relatives)

0 3/9 18/18 1 2/9 2 2/9 3 1/9 4 1/9

Family history of breast cancer (# of affected relatives)

0 3/9 6/18 1 4/9 12/18 2 2/9

Age at diagnosis (yrs) ≤ 55 8/9 5/18 > 55 1/9 13/18

Mean age at diagnosis (yrs)

51 62

Page 63: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

47

2.2.3 RNA Isolation and Reverse Transcription

Total RNA was extracted using Trizol according to the manufacturer’s

instructions (Life Technologies, Frederick, MD, USA). The purity of RNA was

confirmed by A260/A280 ratio of 1.8-2.2, while the integrity of RNA was verified by 1%

agarose gel electrophoresis. Five micrograms of RNA from each tumour sample, as well

as reference RNA composed of 13 pooled cell lines (Table 2-2), were reverse-transcribed

(Superscript III reverse transcriptase, Invitrogen) using anchor oligo dT (AncT) to

generate complementary DNA (cDNA).

Table 2-2. Thirteen cell lines comprising common reference pool used for microarray experiments. The diversity of gene representation by each of these cell lines provides hybridization signal at each probe location on the microarray slide, which is needed for normalization of signal output during analysis process.

Cell Line Description NTERA-2 c1.D1 (CRL-1973) Human testis cancer Hs578T (HTB-126) Human breast carcinoma HepG2 (HB-8065) Human hepatoblastoma Ht1080 (CCL-121) Human fibrosarcoma SW872 (HTB-92) Human liposarcoma T47D (HTB-133) Human breast carcinoma MCF-12A (CRL-10782) Human breast normal Fetal normal muscle 12 week old fetus normal Colo-205 (CCL-222) Human colon cancer MOLT-4 (CRL-1582) Human leukemia RPMI8226 (CCL-155) Human plasmacytoma SKOV-3 (HTB-77) Human ovarian adenocarcinoma SK-MEL-28 (HTB-72) Human melanoma

2.2.4 cDNA Expression Microarrays

Microarray experiments were carried out using arrays that contain ~19,000

characterized and unknown ESTs (University Health Network Microarray Center,

Page 64: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

48

Toronto, Canada, www.microarrays.ca). Tumour and reference cDNAs were

differentially and fluorescently labelled by incorporation of either Cy3-dCTP or Cy5-

dCTP, and were co-hybridized on microarray slides at 37oC for overnight. Arrays were

scanned the next day using the Gene Pix 4000B scanner (Axon Instruments, USA)

following stringent washes. Cy3- and Cy5-labeled cDNA were excited by the green laser

at 532nm and the red laser at 635nm, respectively, and each of the fluorescent spots on

slides were measured and stored as microarray images (Figure 2-1). To minimize the

number of saturated pixels, the PMT (photomultiplier tube) gain for each laser was

adjusted to give Cy3:Cy5 of 1.0. Images were analyzed using GenePix Pro 4.0 Software

(Axon Instruments, USA). Dye swap experiments were performed for each of the

specimens to compensate for dye bias.

Figure 2-1. Subarray of a representative 19K cDNA microarray. Image of a representative subarray of a cDNA expression microarray slide. In this example, cDNA from an ovarian tumour specimen was labelled with Cy5 (red) and cDNA from reference pool was labelled with Cy3 (green). Red spots indicate genes that are more highly expressed in tumour sample, while green spots indicate genes that are less expressed in tumour sample.

Page 65: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

49

2.2.5 Microarray Data Analysis

2.2.5.1 Pre-processing and normalization of expression data

The gene expression data were obtained from the original GenePix image files as

spot intensities by correcting the mean foreground for each spot with the median local

background. The array quality was controlled by requiring arrays to have more than 80%

of spots with spot intensities higher than their local background and more than 75% of

spots with spot intensity higher than 1.2 times their local background in both channels.

Spots with foreground intensity lower than background were treated as missing. A

relative expression value was obtained for each gene as the log base 2 ratio of the

adjusted intensity for the sample channel versus the reference channel. As proposed by

Yang et al. (157), the log2 ratios were normalized by a within-array print-tip loess

adjustment followed by a between-array scale adjustment. Poor quality spots as flagged

by the GenePix image analysis software were excluded from the normalization. Pre-

processing and normalization were carried out using R (http://cran.r-project.org) and the

Bioconductor (http://www.bioconductor.org) package LIMMA (158). There were 15,437

genes retained in the final data set for analysis. All tumours were assessed.

2.2.5.2 Microarray Statistical Analysis

To identify genes that discriminate between strong and weak familial ovarian tumours,

supervised univariate analyses of array-based log2 gene expression were performed for

each gene using the modified Student t-test in the SAM (159) procedure implemented in

R (version 2.10.1, http://cran.r-project.org). P-values were estimated from a set of 500

random sample permutations.

Page 66: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

50

2.2.6 Quantitative Real-time RT-PCR

To validate the gene expression results from the microarray, quantitative real-time

RT-PCR was performed using commercially available Assay-on-Demand probe primer

sets (Applied Biosystems). The standard curve method was employed for quantification

of gene expression. The PCR conditions were as follows: 95oC for ten minutes,

followed by 40 cycles of 95oC for 15 seconds and 59oC for 1 minute. HPRT1

(hypoxanthine phosphoribosyltransferase 1) was chosen as the internal control gene as it

was expressed at similar levels across all tumour samples (Figure 2-2). The difference

between means was tested by using the Student's t test.

Page 67: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

51

Figure 2-2. Expression of housekeeping genes in ovarian tumour samples as measured by gene expression microarrays. In order to determine the appropriate internal control gene for subsequent quantitative real-time RT-PCR experiments, the gene expression of 8 different housekeeping genes were measured and compared among 27 ovarian tumour samples from the present study. MRPL19, HPRT1, YWHAZ, and TBP were shown to have similar expression across tumour samples. HPRT1 was subsequently chosen as the reference gene. MRPL19 PUM1

RPL13A PSMC4

HPRT1 YWHAZ

TBP HMBS

Page 68: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

52

2.2.7 Integration of Array Data to Interaction Networks

Ingenuity Pathway Analysis (Ingenuity® Systems, www.ingenuity.com) was used

to create interaction networks consisting of genes that were significantly differentiated in

the familial ovarian cancer microarray analysis in order to identify and explore the

biological functions that are relevant in the development/progression of ovarian cancer.

In brief, the top significantly differentiated genes from microarray results were mapped

onto Ingenuity Pathway Knowledge Base using gene symbols to generate interaction

networks. The Functional Analysis of a network identified the biological functions that

were most significant to the genes in the network. Fisher’s exact test was used to

calculate a p-value determining the probability that each biological function assigned to

that network is due to chance alone.

Page 69: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

53

2.3 Results

2.3.1 Identification of Genes Distinguishing Strong and Weak Familial Ovarian

Cancers

To identify differentially expressed genes among tumours from patients with

strong and weak family history of breast and/or ovarian cancer, two different approaches

were taken in the analysis: a supervised-class comparison, and a candidate gene

approach.

2.3.1.1 Supervised-Class Comparison

As described, a total of 27 ovarian tumour samples were subjected to gene

expression profiling by a supervised-class comparison approach. Nine of the tumours

represented the group of patients with a strong family history of breast and/or ovarian

cancer and 18 tumours were from patients with a weak family history. The expression

profiles of these two groups were compared and a list of differentiated genes was

generated based on the significance criterion of p<0.01 (Figure 2-3). The most

significant genes identified from the analysis are shown in Table 2-3, along with their

molecular functions and the cellular processes in which they are involved, including

apoptosis/programmed cell death, transcription, protein modification, signal transduction,

cell migration, cell adhesion, and cell cycle regulation.

Page 70: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

54

Figure 2-3. Heatmap illustrating differential gene expression patterns in strong and weak familial ovarian cancer groups (p<0.01). Clustering of top 175 significant genes is shown. Differences with expression levels greater than the mean are coloured in red those below the mean are coloured in green, and no expression differences are in black. Each row represents a single probe set, as identified by GenBank ID, and each column represents individual ovarian tumour samples (blue lines – strong familial tumours; purple lines – weak familial tumours).

Page 71: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

55

Table 2-3. Top 100 differentially expressed genes between strong and weak familial ovarian tumours, as ranked by SAM. Fold difference = strong familial gene expression/weak familial gene expression. *Empty gene ID indicates uncharacterized EST.

GenBank Accession Gene* Molecular Function / Biological Process Cytoband Fold

difference p-value

W60281 ZHX1 Transcription factor activity; negative regulation of transcription, DNA-dependent

8q24.13 1.346 0.00050

AI375078 MAGI3 Kinase activity; apoptosis; intracellular signalling cascade

1p12-p11.2 1.498 0.00057

W31814 PRKCZ Protein binding; protein serine/threonine kinase activity; anti-apoptosis; intracellular signaling cascade

1p36.33-p36.2

2.058 0.00065

AI734230 LOC728676 1q42.13 2.462 0.00084

W38594 TUT1 RNA binding; nucleotide binding; transferase activity 11q12.3 1.877 0.00134

W06830 MCM5 ATPase activity; DNA replication; cell cycle regulation; transcription regulation

22q13.1 1.421 0.00135

AA036944 GMFB Enzyme activator activity; protein kinase inhibitor activity; signal transducer activity; signal transduction

14q22.2 2.710 0.00138

W04817 1.747 0.00178 T70749 0.708 0.00187 BI832845 GNA14 GTPase activity; signal transducer activity; signal

transduction 9q21 1.988 0.00203

T85827 0.392 0.00212 H09018 STX1B Extracellular ligand-gated ion channel activity;

intracellular protein transport; regulation of exocytosis 16p12-p11 0.686 0.00217

R60831 RGMB Identical protein binding; cell adhesion; positive regulation of transcription; signal transduction

5q15 0.559 0.00230

AV725418 0.530 0.00235 N75196 1.436 0.00273 N46715 1.518 0.00283 W03485 SMAD5 Transcriptional activator activity; receptor signaling

protein activity; signal transduction; transcription 5q31 3.257 0.00294

T80520 1.322 0.00309 T95724 SLC25A15 Transporter activity; amino acid metabolic process;

transport; urea cycle 13q14 0.794 0.00309

H19077 DBP RNA polymerase II transcription factor activity; protein dimerization activity; regulation of cell proliferation; regulation of transcription from RNA polymerase II promoter

19q13.2 0.611 0.00311

R51440 0.641 0.00314 R54550 0.578 0.00320 T90266 1.542 0.00321 H71721 1.907 0.00327 BG570119 RSRC2 Inhibitor of cell proliferation 12q24.31 1.411 0.00355 H62960 2.014 0.00374 BQ006563 SIRT3 DNA binding; hydrolase activity; chromatin silencing;

regulation of transcription, DNA-dependent 11p15.5 0.796 0.00375

H61030 REXO2 Hydrolase activity; nucleic acid binding; nucleotide metabolic process

11q23.2 0.645 0.00382

BM469380 ZDHHC6 Metal ion binding; transferase activity 10q25.3 1.886 0.00419 W01536 PDCD4 RNA binding; protein binding; apoptosis; cell aging;

negative regulation of cell cycle progression; negative regulation of transcription

10q24 0.698 0.00424

W00465 TSPYL5 Nucleosome assembly 8q22.1 0.722 0.00435

Page 72: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

56

N92095 0.517 0.00445 R97881 1.412 0.00458 R91589 0.514 0.00462 R17265 HGSNAT Transferase activity 8p11.1 1.516 0.00471

AA203115 SLC22A16 Transporter activity; transport 6q22.1 1.605 0.00472

R54138 GPR162 Receptor activity; G-protein coupled receptor protein signaling pathway; signal transduction

12p13 0.769 0.00477

N77718 1.505 0.00488 AL519368 LOC389833 4 1.669 0.00501

R86730 LBR DNA binding; protein binding; receptor activity 1q42.1 0.608 0.00506 N46724 ARHGEF7 Guanyl-nucleotide exchange factor activity; protein

binding; apoptosis; signal transduction 13q34 1.547 0.00527

H17686 LRRC17 Protein binding; osteoblast differentiation and proliferation

7q22.1 1.910 0.00536

W31507 CHRFAM7A Extracellular ligand-gated ion channel activity; ion transport

15q13.1 1.626 0.00536

N91701 PAPD1 PAP associated domain containing 1 10p12.1 1.562 0.00571 BG616960 VTN Protein binding; cell adhesion; cell-matrix adhesion 17q11 0.717 0.00576 T80023 1.367 0.00577 AA043077 0.642 0.00593 R87552 GNAO1 GTPase activity; receptor signaling protein activity;

signal transduction 16q13 0.742 0.00607

R50922 NLGN4X Protein binding; protein homodimerization activity; cell adhesion; cell-cell junction organization

Xp22.33 0.656 0.00612

H28503 NOTCH2NL Calcium ion binding; Notch signaling pathway; cell differentiation

1q21.2 1.143 0.00613

R06130 FCN1 Calcium ion binding; receptor binding; signal transduction

9q34 0.697 0.00614

H16624 FAT4 Calcium ion binding; cell adhesion 4q28.1 1.689 0.00617

W87412 ARID2 DNA binding; protein binding; chromatin modification; regulation of transcription, DNA-dependent

12q13.11 1.732 0.00656

AI820728 0.559 0.00656 H88063 C1orf9 Multicellular organismal organization 1q24 0.434 0.00669 R24998 0.665 0.00683 N95578 1.403 0.00713 H04765 CPD Metal ion binding; peptidase activity; proteolysis 17p11.1-

q11.2 0.700 0.00721

AA143153 CYP11A1 Cholesterol binding; metal ion binding; cholesterol metabolic process; response to estrogen stimulus

15q23-q24 1.630 0.00727

T83295 HEXA Hydrolase activity; metabolic process 15q23-q24 0.832 0.00743 AA203550 2.423 0.00750 R97190 0.625 0.00763 H08597 PLCXD3 Hydrolase activity; signal transducer activity; lipid

catabolic process 5p13.1 0.596 0.00767

W03856 ABCC6P1 16p12.3 1.516 0.00770

H26209 0.755 0.00772 H66891 0.679 0.00777 N46663 1.420 0.00777 T83443 IGHMBP2 ATP binding, DNA binding; RNA binding;

transcription factor binding; protein binding; DNA recombination; DNA repair; DNA replication; cell

11q13.2-q13.4

0.644 0.00786

Page 73: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

57

death; regulation of transcription R12356 MAOB Protein homodimerization activity; oxidative reduction Xp11.23 1.639 0.00801 BM982143 POLR3H DNA binding; DNA-directed RNA polymerase activity;

transcription 22q11.2-q13.31

1.314 0.00811

AL556801 NCAPD3 Binding; cell cycle; cell division; mitosis 11q25 1.597 0.00839

AA099730 FVT1 Binding; oxidoreductase activity; oxidation reduction; sphingolipid biosynthetic process

18q21.3 1.361 0.00846

T97415 1.499 0.00848 H10128 SHC2 Protein binding; Ras protein signal transduction;

intracellular signaling pathway 19p13.3 0.842 0.00849

R54527 0.670 0.00867 R67303 NEO1 Cadherin binding; receptor activity; transcription

regulation activity; cell adhesion 15q22.3-q23

0.661 0.00877

AA037624 C7orf49 7q33 1.364 0.00889 R59287 SFRS14 RNA binding; RNA splicing; mRNA processing 19p12 1.518 0.00905 H19859 RABAC1 Protein binding 19q13.31 0.683 0.00905 T95778 SH3BP4 Protein binding; endocytosis 2q37.1-

q37.2 0.792 0.00911

AL575239 0.601 0.00924 H51850 0.542 0.00934 AW953237 PITPNC1 Lipid binding; protein binding; lipid transport; signal

transduction 17q24.3 0.723 0.00939

W78914 PILRA Protein binding; receptor activity; signal transduction 7q22.1 2.230 0.00942

H72224 MRPS12 Protein binding; translation 19q13.1-q13.2

0.691 0.00954

BQ017489 ACTN1 Actin binding; integrin binding; protein binding; focal adhesion assembly; regulation of apoptosis

14q24.1-q24.2

1.420 0.00960

BM509122 IGL@ 22q11.1-q11.2

0.403 0.00968

R24238 1.235 0.00968 R73462 ATHL1 Hydrolase activity; carbohydrate metabolic process 11p15.5 0.731 0.00972 T83168 0.479 0.00989 AA134742 BRWD2 Cell cycle progression; signal transduction; apoptosis,

gene regualtion 10q26 2.349 0.00990

W86215 ERF Ligand-regulated transcription factor activity; transcription corepressor activity; cell proliferation

19q13 2.160 0.00992

H29655 RELB DNA binding; protein binding; transcription corepressor activity; regulation of transcription, DNA-dependent

19q13.32 0.624 0.01051

AA126588 0.754 0.01065 AA047000 IL17RC Receptor activity 3p25.3 1.436 0.01070 AW962742 LOC400604 17q21.33 1.357 0.01077

H42572 0.700 0.01085 R83139 SCFV 14 0.436 0.01095 H88020 1.608 0.01096 AA034344 8p21.1 1.539 0.01110

Page 74: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

58

2.3.1.2 Candidate Gene Approach

In addition to the supervised-class comparison, I have chosen a list of genes from

the literature that has previously been implicated in ovarian cancer or genes that are

biologically relevant, and compared each of their microarray gene expression values

between the strong and weak family history ovarian cancer groups. The selected genes

include: MYC, KRAS, PIK3AP1, hCDC4, LRMP, MLH1, TP53, CDKN1B, EVI1,

CCND1, PIK3CA, as well as genes from the TGFβ family. Most of these genes did not

show differential expression between our two subject groups. Interestingly, while

differential expression of hCDC4 was not detected as significant in the supervised-class

comparison analysis at cut-off p-value of 0.01, it was identified as significantly less

expressed in group with strong family history at p=0.05 using the candidate gene

approach (Figure 2-4).

2.3.2 Validation of Differentially Expressed Genes

I selected several genes for gene expression validation using quantitative real-time

RT-PCR with RNA from the same 27 tumour specimens used for my microarray

experiments, based on fold difference of at least 2.0, as well as their potential roles in

cancer based on previous literature, which included PRKCZ, FAT4, and hCDC4. The

gene expression patterns between the two subject groups for these genes as identified by

real-time PCR were in concordance with the microarray results (Figure 2-5). Other genes

such as SMAD5 and NEO1 were chosen for validation, and while similar trends were

observed, the differences did not reach statistical significance.

Page 75: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

59

Figure 2-4. Identification of hCDC4 as a differentially expressed gene between strong and weak familial ovarian tumours by candidate gene approach. Selected genes were chosen from the literature and their gene expressions from microarray results were analyzed. hCDC4 gene expression was shown to be differentially expressed (p =0.05).

Page 76: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

60

Figure 2-5. Real-time PCR validation of differential gene expression between strong and weak family history groups. Expression levels of selected genes FAT4, PRKCZ, and hCDC4 were independently validated using real-time PCR using the same 27 ovarian tumour samples from microarray experiments. Differential gene expression patterns identified from real-time PCR were in concordance with microarray expressions. cDNA microarray expression values were normalized using global scaling approach and real-time PCR expression values were normalized to housekeeping gene HPRT1 (Student’s t test).

PRKCZ mRNA Expression

0

1

2

3

4

5

Microarray RT-PCR

Rel

ativ

e m

RN

A E

xpre

ssio

n

Strong Familial

Weak Familial p=0.01

FAT4 mRNA Expression

0

0.5

1

1.5

2

2.5

Microarray RT-PCR

Rel

ativ

e m

RN

A E

xpre

ssio

n

Strong FamilialWeak Familial p=0.03

hCDC4 mRNA Expression

00.5

11.5

22.5

33.5

44.5

Microrray RT-PCR

Rel

ativ

e m

RN

A E

xpre

ssio

n p=0.05 Strong Familial

Weak Familial

Page 77: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

61

2.3.3 Molecular Network Analyses Dynamic pathway modelling was carried out using Ingenuity Pathway Anaysis

(IPA) software to examine various molecular interactions/pathways associated with

familial ovarian cancer. I mapped the top ranked 300 differentially expressed genes

between the strong and weak ovarian cancer groups onto IPA Knowledge Base, and the

connectivity of the genes through direct physical, transcriptional and enzymatic

interactions was computed and interaction networks were generated. Focus genes

(differentially expressed genes from the present study), the network score (in which a

value greater than 2 corresponds with a probability of <0.01% that the network was

assembled by chance alone), and the cellular functions associated with each of the

identified networks are listed in Table 2-4.

Through IPA, it is confirmed that the genes that are differentially expressed

between strong and weak familial tumours include players that participate in relevant

signalling pathways related to tumourigenesis. These pathways include those involving

MAPK, HNF4A (hepatocyte nuclear factor 4, alpha), histone 3, HGF (hepatocyte growth

factor), and beta-estradiol (Figure 2-6). Some of the cellular functions and diseases

associated with molecular networks include DNA replication, DNA recombination, DNA

repair, cell cycle, lipid metabolism, cell death, and cellulr growth & proliferation, and

cancer.

Page 78: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

62

Figure 2-6. Graphical representations of the molecular relationships between genes identified from familial ovarian microarray analysis using Ingenuity Pathway Analysis. Red nodes represent genes that had higher expression in the strong familial group, green nodes represent genes that had lower expression in the strong familial group, and white nodes represent genes that had similar expression between two groups or were not on the array. Biological relationship between two nodes is represented as an edge (line). All interactions are supported by at least one reference from the literature, from a textbook, or from canonical information stored in the Ingenuity Pathways Knowledge Base. Different shapes of enodes represent the functional class of the gene product as indicated in the figure legend. Interaction networks involving A) players within MAPK signaling pathway, B) HNF4A, C) histone 3, and D) HGF and beta-estradiol.

Page 79: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

63

A)

Page 80: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

64

B)

Page 81: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

65

C)

Page 82: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

66

D)

Page 83: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

67

Table 2-4. Top functions of networks as identified by Ingenuity Pathway Analysis (IPA). Differentially expressed genes from microarray analysis are shown in bold (focus molecules). A score of >3 was considered statistically significant (p<0.001).

Molecules in Network Score Focus Molecules Top Functions

Actin, ADCY, Akt, BIRC3, Caspase, CDC23, Cyclin A, CYP11A1, E3 RING, ERK, ERK1/2, G alpha, G alphai, G-protein beta, GMFB, GNA14, GNAO1, GPC1, hCG, HIRA, IGF1R, ITGB3, Jnk, LBR, MAP3K8, Mapk, NFkB (complex), NPPA, PARK2, PI3K, PIP5K1B, Pkc(s), PRKCZ, Rac, VTN

36 17

DNA Replication, Recombination, and Repair, Infection Mechanism, Endocrine System Development and Function, Lipid Metabolism

ACO2, ACTN1, BMP15, C1ORF9, CABC1, CXXC5, DACH1, DLG4, FBP1, FDX1, FSH, FXN, HDAC8, HEXA, HNF4A, KCNA5, MIR298, NEO1, NLGN4X, PDCD4, RABAC1, RBAK, REXO2, RGMA, RPLP1, SF3B1, SH3BP4, SMAD5, Smad1/5/8, SNRPA, TOB1, TUT1, VPS29, XRCC4, ZDHHC6

28 14

Cell Cycle, Reproductive System Development and Function, RNA Damage and Repair, Cancer

ACHE, beta-estradiol, CKS2, CPD, CTSH, FCN1, G3BP2, GM2A, HGF, IFNG, IGHMBP2, IL6, IRS, IRS1, MYC, NAPSA, NFKBIA, OSBPL1A, PPP1R12C, RDBP, RFC2, SERPINA1, SFRS2, SFRS2IP, SFTPB, SH2B2, SLC22A16, SLC25A15, SMARCA4, SNRNP70, SPHK2, TFDP1, TGFB1, ZFP36L2, ZNF185

26 13

Cell Cycle, Cancer, Infection Mechanism, Lipid Metabolism

ABI3, AKAP, AKAP1, AKAP13, ALDOA, ARHGEF7, ATXN1, C3ORF15, CBFA2T3, DIO1, FAT4, Histone h3, Histone h4, LRCH1, MCM5, Mi2, MTUS1, NCOR1, P38 MAPK, Pka, PRKAC, PRKAR1B, RUNX1T1, SCRIB, SIRT3, TBL1XR1, THAP7, THRAP3, WWC1, WWC2, YWHAZ, ZFP36, ZHX1, ZHX3, ZMYND11

21 11

Cellular Development, Hematological System Development and Function, Hematopoiesis

AHR, CDC2, Ck2, CREM, CSNK2A1, CSNK2B, CTGF, D830050J10RIK, FBL, FGF1, GTF2F1, L1CAM, LRRC17, MDM2, Mg2+, MYCN, NCL, NETO2, NME1, NOP2, ODC1, PTN, RBL2, SNAP25, SPP1, STX4, STX1B, SYT1, SYT4, TBP, TCF4, THRA, TOP1, VAMP2

8 5

Cellular Growth and Proliferation, Cancer, Cell Morphology, Cell Death

Page 84: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

68

2.4 Discussion

Gene expression microarray is a powerful tool for identifying abnormal gene

expression patterns associated with various human diseases. Indeed, a number of

microarray studies have been conducted in the past decade to identify gene expression

alterations in ovarian cancer (140). These studies have identified signature profiles of

ovarian cancer cell lines and ovarian carcinomas, which subsequently led to the

identification of potential prognostic markers, as well as genes involved in ovarian

tumourigenesis and those involved in chemotherapy resistance, such as resistance to

cisplatin (140). While these studies have indeed provided a better understanding of

ovarian cancer, the global gene expression profiling for familial ovarian cancer is lacking.

To address this, I have applied cDNA microarrays and compared the gene expression

patterns in tumour samples from patients with strong versus weak familial backgrounds

of breast and/or ovarian cancer.

We have identified a list of significant genes that are differentially expressed

between ovarian tumours from patients with strong and weak family history of breast

and/or ovarian cancer, on the basis of the analysis of 9 strong-familial and 18 weak-

familial ovarian tumours.

A subset of the genes identified from the array analysis has specifically been

demonstrated to be involved in cancer development in earlier studies. Of these genes,

some showed lower expression within the strong familial group, including the following:

SIRT3 (sirtuin 3), a gene encoding a NAD-dependent histone deacetylase that has been

demonstrated to play role in apoptosis in a variety of human cancer cell lines (160); VTN

(vitronectin), an extracellular matrix protein that plays role in cell adhesion and

Page 85: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

69

spreading, and previously demonstrated to play role in ovarian cancer dissemination

(161); NEO1 (neogenin homolog 1), which encodes a cell surface protein that shares

homology with tumour suppressor candidate gene DCC (deleted in colorectal carcinoma)

and whose expression has been previously observed to be significantly reduced in

prostate tumours compared to normal prostate tissues (162, 163), and PDCD4

(programmed cell death 4), a tumour suppressor gene shown to inhibit proliferation of

neuroendocrine tumour cells, and whose expression is frequently lost in human glioma, in

addition to its involvement in the progression of lung, breast, colon, prostate carcinomas,

and most notably shown to be able to inhibit the malignant phenotype of ovarian cancer

in vitro and in vivo (164-167).

Likewise, some genes exhibit higher levels of expression in the strong familial

patient group compared with the weak familial group. Such genes include MAGI-3

(membrane associated guanylate kinase), whose gene product interacts with LPA2 to

facilitate LPA2-mediated activation of ERK and RhoA, potentially leading to an increase

in gene transcription and cell survival (149); cell cycle regulation gene MCM5

(minichromosome maintenance complex component 5), which has been described to be

over-expressed in cervical and esophageal cancer (168, 169); as well as CYP11A1

(cytochrome P450, family 11, subfamily A, polypeptide 1), which encodes a member of

the cytochrome P450 superfamily of enzymes involved in steroidogenesis and its genetic

variation has been suggested to be able to influence risk of various malignancies,

including endometrial and prostate cancers (170, 171).

Additionally, the gene PRKCZ (protein kinase C zeta) also exhibited a higher

level of expression in the strong familial group. This gene encodes a serine/threonine

Page 86: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

70

kinase that has previously been implicated in the regulation of cellular transformation and

carcinogenesis (172). The importance of PRKCZ in tumourigenesis has been shown in

various studies (172-178); however, its role in ovarian cancer had yet to be determined.

Therefore, I have chosen this gene for further investigations. Detailed description of this

gene, as well as the functional studies of PRKCZ that I have performed, will be discussed

in Chapter 4 of this thesis.

In order to interpret my microarray data and to generate hypotheses for future

studies, I have utilized Ingenuity Pathway Analysis (IPA) software for further analyses.

IPA was applied to identify interaction networks among genes that are differentially

expressed between ovarian cancers from strong versus weak family history to gain an

understanding of which functional cellular processes were altered between these two

subject groups.

Through statistical analyses performed by IPA, several potentially important

interaction networks were identified. The top functions and diseases associated with

these networks include DNA replication, recombination and repair, cancer, cell

morphology, cell death, cell cycle, as well as cellular growth and proliferation. Other

cellular processes such as lipid metabolism, endocrine, reproductive, and haematological

system development were also identified.

By examining these networks, I was able to identify signalling molecules and

pathways that were altered in patients with a strong family history of breast and/or

ovarian cancer. For example, it was observed that the expressions of genes directly or

indirectly associated with the mitogen-activated protein kinase (MAPK) pathway were

altered between these patient groups. These genes include VTN (vitronectin), IGF1R

Page 87: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

71

(insulin-like growth factor 1 receptor), ITGB3 (integrin beta 3), LBR (lamin B receptor),

BIRC3 (baculoviral IAP repeat-containing 3), and PARK2 (parkin 2). Some of these

genes have previously been associated with ovarian cancer (179-184); however,

additional functional studies are required to determine if these genes are able to cooperate

with each other to enhance ovarian cancer development. A protein network that

functions in cell cycle and cell death was also found from our IPA analysis. This

particular network centers around HNF4A, a member of the nuclear receptor superfamily

that has been demonstrated to be a useful marker for histological and cytological

diagnosis of ovarian mucinous tumours (185). Its role in other ovarian tumour types,

including serous carcinomas, however, requires further investigation. In addition to

HNF4A, other molecules such as tumour suppressor PDCD4 (programmed cell death 4),

TGFβ1 interactor SMAD5 (SMAD family member 5), and tumour suppressor

retinoblastoma 1 interactor RBAK (RB-associated KRAB zinc finger) were also present

in this interaction network, and may be directly or indirectly regulated by HNF4A. Their

exact regulation mechanisms and their roles in familial ovarian tumourigenesis remain to

be examined. Another network encompassing histone H3 was observed in which gene

expression of histone H3 interactors such as NCOR1 (nuclear receptor co-repressor 1),

MTUS1 (mitochondrial tumour suppressor 1), MCM5 (minichromosome maintenance

complex component 5), and RUNX1T1 (runt-related transcription factor 1) were found to

be differentially expressed between the two groups. Since histone H3 modification has

been previously described in ovarian cancer (186, 187), it may be relevant to examine

how these interactors can affect the activity of histone H3, or vice versa, in familial

ovarian cancer.

Page 88: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

72

Lastly, an interaction network we observed involves multiple molecules including

MYC, TGFβ1 (transforming growth factor, beta 1), HGF, and beta-estradiol. While

differential expression levels of these specific genes were not identified in our study, it is

interesting to note that their common direct/indirect interactors are shown to be

differentially expressed between the two groups, thus linking these important signalling

pathways. In particular, the presence of HGF and beta-estradiol within this interaction

network is a notable finding.

HGF is a pleiotropic factor that, along with its receptor tyrosine kinase Met, can

stimulate multiple biological responses, including epithelial morphogenesis (188).

Moreover, it has been observed that HGF and Met are both differentially regulated in

normal human ovarian surface epithelium cultures derived from women with and without

a family history of ovarian cancer, and was suggested that co-expression of these two

genes may enhance susceptibility to ovarian carcinogenesis in women with hereditary

ovarian cancer syndromes by their ability to activate the PI3K and MAPK signalling

pathways, which may lead to metaplastic changes of the ovarian surface epithelium

(188). Based on this previous observation, and results from our interaction analysis, I

hypothesize that players associated with the HGF pathway may be important in ovarian

cancer in the subset of patients with a strong family history, by altering and sustaining the

expression of the genes involved in ovarian tumourigenic transformation. Therefore it

may be worthwhile to further examine the regulatory mechanisms within the HGF-axis in

relation to familial ovarian cancer development.

The presence of beta-estradiol related interactors found within this interaction

network was also of interest given that recent studies have found an association between

Page 89: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

73

certain genetic polymorphisms within the steroid hormone pathway and an increase in

breast and ovarian cancer risks (189). How these specific polymorphisms can affect the

expression of these genes, or how these genes may directly or indirectly be altering the

steroid hormone pathway thus leading to ovarian carcinogenesis, however, still requires

further investigation. Nevertheless, the importance of beta-estradiol in ovarian cancer

development has been demonstrated. It was suggested that this hormone may play a role

in ovarian tumourigenesis by up-regulating bcl-2 gene and protein expression levels, thus

preventing apoptosis in tumourigenic ovarian surface epithelial cells (190). This is

critical during early stages of ovarian cancer development, as this advantage in cell

survival may permit these cells to accumulate mutations at a greater than normal rate,

thereby accelerating the overall rate by which these cells can transform from a pre-

malignant to malignant state. In another study, the role of beta-estradiol in the

reinforcement of invasion in epithelial ovarian cancer cell lines was examined (191). It

was found that ovarian cancer cells induced by beta-estradiol can lead to an increase in

MMP-2 expression and a decrease in E-cadherin expression, and that these changes were

associated with an increased expression of Snail, a transcription factor shown to be

involved in epithelial-mesenchymal transitions (EMT) of cancer cells, which is important

in invasion and metastasis (191). Taken together, it has been suggested that the

regulation in beta-estradiol expression during both early and late stages may be critical

during ovarian carcinogenesis.

Based on these sub-networks, it would be interesting to examine whether the

different players that control the levels of HGF and beta-estradiol are differentially

expressed or activated in patients with different family history of breast and/or ovarian

Page 90: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

74

cancer, and how these changes in expression or activity may affect downstream targets

that can affect ovarian cancer development, such as those players involved in

tumourigenic transformation, cell survival and EMT. It may also be possible that

BRCA1 and/or BRCA2 are participants in these pathways directly or indirectly; however,

further studies are required to determine if this is the case.

In conclusion, the microarray results from this Chapter provided the genetic

profiles of familial ovarian cancer, and while larger sets of families with familial breast

and/or ovarian cancer are required to further validate and to increase robustness of our

results, my findings have provided some promising candidate genes that may potentially

be important in familial ovarian tumourigenesis. Potential biological pathways involved

in this disease are further identified through bioinformatics analyses and polygenic

alterations within these pathways are likely to be involved in increasing the susceptibility

of ovarian cancer in individuals with strong family history of breast and/or ovarian

cancer. However, the detailed mechanisms of how and which of these genetic

alterations are involved, and whether these alterations have different effects in BRCA1 or

BRCA2 mutation carriers, remain to be investigated. Therefore, additional functional

studies of the identified genes will be required to provide information that may be

valuable in understanding the relevant mechanisms involved in the development of

familial ovarian cancer.

Page 91: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

CHAPTER 3

hCDC4 in Familial Ovarian Cancer

The work presented in this Chapter was performed by KS with the exception of the

immunohistochemistry on ovarian tissue microarray (TMA), which was performed in

collaboration with Dr. Patricia Shaw, with assistance from laboratory technician Kelvin

So. Scoring of TMA was assisted by Dr. Alicia Tone.

75

Page 92: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

76

3.1 Introduction

The ubiquitin-proteosome system (UPS) is an important regulator of cellular

homeostasis, as it is involved in the destruction of regulatory proteins within the

eukaryotic cell (192). Some of the cellular processes which are UPS-dependent include

differentiation, proliferation, DNA damage repair and apoptosis (192, 193). UPS

functions through an enzymatic cascade consisting of ubiquitin-activating enzyme E1,

ubiquitin-conjugating enzyme E2, and ubiquitin-protein ligase E3 (192).

Several hundred E3 ligases exist in the human genome and the SCF

(SKP1/CUL1/F-box) ubiquitin ligase complex is among the best studied (192). SCF

complex contains a cullin family scaffolding protein that binds to a catalytic RING finger

(RBX1), which recruits E2 and SKP1 that interact with an F-box protein (194). Besides

the SKP1-binding domain, F-box proteins also contain a substrate-binding domain that

recognizes specific phosphorylations within its target proteins (194). Currently

approximately 69 F-box proteins have been identified in humans, and each of these

proteins can target multiple substrates for degradation (195).

hCDC4 (hAGO/FBXW7) is an example of a F-box protein previously shown to

play a critical role in the regulation of multiple oncoproteins, including cyclin E1 (Figure

3-1), c-Myc, c-Jun, Notch, and mTOR (196-200). hCDC4 possesses three isoforms:

hCDC4-α, β, and γ, each containing a unique N-terminal protein domain that is likely

responsible for cellular localization and tissue expression. All three isoforms share a

common C-terminal region, which consists of 7 WD repeat domains that are responsible

for substrate recognition (193).

Page 93: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

77

Figure 3-1. Pathway of hCDC4-mediated degradation of Cyclin E. The enzymes Cdk2 and cyclin E are important for the G1-S transition of the cell cycle. Upon completion of this transition, cyclin E is ubiquitylated through its interaction with E1 (ubiquitin-activating enzyme), E2 (ubiquitin-conjugating enzyme), and E3 complex (ubiquitin ligase, consisting of Cul1, Skp1, and Rbx1), and degraded through the 26S proteosome. hCDC4 F-box protein contains WD domains that recognize phosphorylated cyclin E and delivers cyclin E to the E3 complex. In tumour cells, mutation of hCDC4 prevents recognition of cyclin E, leading to its aberrant accumulation, thus deregulation of the cell cycle (Adapted from Schwab and Tyers, 2001, ref (201)).

Page 94: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

78

The significance of hCDC4 in cancer development is further made evident by the

observations of mutations within the hCDC4 gene in several human neoplasias, such as

colorectal and endometrial tumours, as well as cancer cell lines, including ovarian cancer

cell lines (202-205). Based on these observations, hCDC4 has been suggested to be a

tumour suppressor and thus its deregulation may be important in cancer development.

Interestingly, as discussed in Chapter 2, my present familial ovarian cancer

microarray analysis revealed that the hCDC4 gene was less expressed in strong familial

group compared to weak familial group (Figures 2-4, Chapter 2).

In this Chapter, I discuss the genetic approaches that I have taken to investigate

the mechanisms that may be responsible for the altered gene expression of hCDC4.

First, I performed protein truncation test, single strand conformation polymorphism

(SSCP) analysis and manual sequencing to determine the mutation status of the coding

regions of hCDC4 of ovarian tumours. I also examined the promoter methylation status

of these tumours by performing methylation-specific PCR (MSP). Additionally, I

sought to examine the loss of heterozygosity (LOH) status of ovarian tumours by

performing LOH analysis. The protein expression of hCDC4 in ovarian tumours was

also examined by immunohistochemistry (IHC) staining of ovarian cancer tissue

microarrays (TMA). Lastly, the gene expression of cyclin E, a downstream target of

hCDC4, was also examined in familial ovarian cancer.

Page 95: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

79

3.2 Materials & Methods

3.2.1 Ovarian tumour samples, and RNA, DNA Extraction

Ovarian tumour sample acquisition and RNA isolation were as described in

Chapter 2 (section 2.2.3). One additional tumour from patient with strong family history

was acquired after microarray analyse, thus a total of 28 ovarian tumour samples were

used for experiments described in this Chapter. Total genomic DNA was isolated from

each sample with QIAamp® DNA mini kit (Qiagen) according to the manufacturer’s

protocol.

3.2.2 Protein Truncation Test

To examine the entire coding sequence of the hCDC4 gene, two pairs of primers

were used to amplify fragments of 1425bp (spanning exons 1-8) and 1431bp (spanning

exons 4-11) containing an overlap of 588 bp. Three microlitres of reverse transcribed

cDNA was utilized in each reaction. The forward and reverse primers flanking exons 1-8

were 5’-

GCTAATACGACTCACTATAGGAACAGACCACCATGATGATGAGCTGGCTTTT

GGAAATGAA-3’ and 5’-ATGCATACAACGCACAGTGG -3’; and the primers

flanking exons 4-11 were 5’-

GCTAATACGACTCACTATAGGAACAGACCACCATGATGATGGAACCCCAGTT

TCAACGAGAC-3’ and 5’-CAACATCCTGCACCACTGAGAACAAGG -3’. The

forward primers included a leader sequence consisting of a bacteriophage T7

transcription promoter plus eukaryotic translation initiation signals. The ATG-initiation

codon was in frame and upstream of the natural translation initiation site. The PCR

Page 96: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

80

reaction was carried out in a 25 μl reaction volume containing 1.6-2mM MgCl2, 0.6 mM

dNTPs, 2.5 U platinum Taq Polymerase (Promega), and 9 pmol of each primer. PCR

parameters were: 94°C denature for 2 minutes, followed by two steps of PCR, 4 cycles of

94°C for 30 seconds, 69°C for 30 seconds and 72°C for 1.5 minute; 35 cycles of 94°C for

30 seconds, 58°C for 30 seconds and 72°C for 1.5 minute. A sample of of the PCR

product (2.4 μl) was then used in the coupled transcription and translation reaction in

accordance with the recommendations of the supplier (TNT Coupled Reticulocyte Lysate

System; Promega). After in vitro transcription/translation incorporation of radioisotope

35S-methionine, the incorporated protein products were electrophoresed on a 12.5% SDS

polyacrylamide gel. The gel was then fixed, dried and subjected to autoradiography for 3-

12h. In addition to hCDC4, the BRCA1 gene with a known truncation mutation was used

as positive control to test the sensitivity of the PTT method.

3.2.3 Single Strand Conformation Polymorphism (SSCP) and Manual Sequencing

All coding exon primers for SSCP were designed individually by using Primer3

input software (Whitehead Institute, Howard Hughes Medical Institute, NIH). Genomic

DNA was used as a template for polymerase chain reaction (PCR) amplification of

fragments containing an exon and its adjacent intronic boundaries. Placenta genomic

DNA was used as control. The sequences of each primer set used to amplify exons are

listed in Table 3-1. Some exons were multiplexed (exons 3 and 7, exons 4 and 9, exons 6

and 11, exons 8 and 10). 50ng of tumour DNA was added to a reaction buffer containing

10mM Tris, pH 8.3, 50mM KCl, 1.8-2.6 mM MgCl2, 0.4mM of dNTPs, 6-9 pmol of each

primer, 1mCi 33P-dATP (10mCi/μL, Amersham, USA), and 2 units of AmpliTaq (Perkin

Page 97: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

81

Elmer, Norwalk, CT). 33P-dATP incorporated PCR products were heat denatured and

electrophoresed on a 37.5% native polyacrylamide gel containing 10% glycerol. Results

were obtained following autoradiography. Sequence alterations which were detected as

electrophoretic mobility shifts on SSCP gels were confirmed and characterized

independently by direct manual sequencing of PCR products using the same SSCP

primers (ThermoSequenase cycle sequencing kit; Amersham Life Science, Arling

Heights, IL).

Table 3-1. Primer sets for hCDC4 SSCP and manual sequencing analyses.

Exon Forward Primer (5’ – 3’) Reverse Primer (5’ – 3’) Product Size (bp)

1 (α) GACGAACTGGAGGCTCTCTG CTCCTCCTCCTCATCCTCCT 311

1 (β) CCCTCGAGTTCTTCTCAGTCA GCAGGCATACACACACAATCA 347

2 TGACTCAAGATTTGATAGTTAGACGA AAACTAAAACACTTTCAGAATCAACTC 217

3 TTTTCCTTTTATCCTTTCTCTCTCTC GCAGCAATTAAGTGAGGCATT 233

4 GCCTGTAATTTGGGACATCTG CAAATAACACCCAATGAAGAATG 231

5 TCAAGTATCTCATCCTGTGGAGAA TTTCAGAATCACTCTGCTTTTCA 283

6 TGGTGAAGGCAATTTACTCTTG AACGGTTTCTGTTACATTGTGC 210

7 CATATTTCTAATCTGCACATCTTTCTT TGACTTTGTGAAGTGTAGGAAGAG 178

8 AAGTAATCATCTTAAGTGTTTTTCCAG CCAACCATGACAAGATTTTCC 235

9 TTTTTCTGTTTCTCCCTCTGC TTCATCAGGAGAGCATTTAAGG 290

10 TCAGTAATTGATAGGAAGAGTATCCA AACAAAACGAAAGGTGAGTAAGAC 298

11 CCAGTAATTAAATTCTTTTGGTTTTTG TGGACAAATTCATCTTTTCTGCT 326

Page 98: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

82

3.2.4 DNA Methylation-Specific PCR

hCDC4 promoter methylation was assessed using the bisulfite PCR method.

Bisulfite modification of DNA was carried using the EpiTect Bisulfite kit (Qiagen)

according to the manufacturer’s protocol to detect cytosine methylation. In brief,

genomic DNA from all ovarian tumour samples was treated with sodium bisulfite to

convert any unmethylated cytosine residues into uracil, giving rise to different DNA

sequences for methylated and unmethylated DNA. Primer pairs for bisulfite sequencing

PCR (Table 3-2) were designed using MethPrimer software (206). Bisulfide-modified

DNA was then subjected to PCR amplification using the following thermal PCR

conditions: initiation step at 94ºC for 4 minutes, 40 cycles of 95ºC for 30 seconds, 50ºC

for 30 seconds and 72ºC for 30 seconds, followed by an extension at 72ºC for 10 minutes.

CpGenome Universal Methylated and Unmethylated DNA (Chemicon, Billerica, MA,

USA) were used as positive and negative controls, respectively. PCR products were

electrophoresed on a 1.5% agarose gel. Methylation was determined by the presence or

absence of a DNA band.

Table 3-2. Primer sequences for methylation-specific PCR.

Primer Forward Primer (5’-3’) Reverse Primer (5’-3’) Product Size (bp)

hCDC4_1m GCGGTAGTTTAGGTTCGATTC CTCTAACGCGCTCTAATAACG 170

hCDC4_1u GGGTGGTAGTTTAGGTTTGATTT CCTCTAACACACTCTAATAACACT 173

hCDC4_2m AGGCGAGAGTTTCGTATAGAGC TCTAACTCCGACTCCGACGTA 248

hCDC4_2u AGGTGAGAGTTTTGTATAGAGTGA ACTCTAACTCCAACTCCAACATA 250

hCDC4_3m GTTGTCGTTTGGTTTAGC GATAC GGGTTGTTGTTTGGTTTAGTGATAT 102

hCDC4_3u GGGTTGTTGTTTGGTTTAGTGATAT ATAAATTAATTCCCTTCCTCCTTCA 107

Page 99: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

83

3.2.5 Loss of Heterozygosity (LOH) Analysis of hCDC4

Genomic DNA from four ovarian tumour samples, paired with DNA from an

adjacent area of normal tissue was available for hCDC4 LOH analysis. Allelic loss was

evaluated using 3 polymorphic markers D4S1548, D4S2934 and D4S3049, which closely

flank the hCDC4 locus on chromosome 4q31.3. Primers sequences used for LOH are

listed in Table 3-3.

Table 3-3. Polymorphic markers used for LOH analysis of hCDC4.

Polymorphic Marker Forward Primer (5’-3’) Reverse Primer (5’-3’)

D4S1548 TGCCATAAACAAGGTGAAAC TTACCCAACTGCTACACCAT

D4S2934 CAAAACAGATCAGGATGTGG TTGCTGTCTTTACAGAGCACC

D4S3049 ATTCAGTTCTCTGCGAATG AGTTCGTGCCACTGTACTC

Additional information about these loci was obtained from UniSTS

(http://www.ncbi.nlm.nih.gov/sites/entrez?db=unists). The locations of primers and the

genes that lie between microsatellite repeat markers were determined by Map Viewer at

the National Center for Biotechnology Information (NCBI) Web site

(http://www.ncbi.nih.gov/Tools/index.html). PCR amplification was performed in a final

volume of 30 μl containing 1 μl (200 ng) of DNA template, 1x High Fidelity PCR Buffer,

2 mM MgSO4, 0.2 mM of each dNTP, 0.3 mM of forward and reverse primers, 1 U of

Platinum Taq DNA polymerase High Fidelity (GIBO BRL, Life Technologies, Canada),

and 0.1 μCi of [33P] dATP (Perkin-Elmer, USA). Thermal conditions were as follow:

initiation denaturation step at 95oC for 2 minutes, 40 cycles of 95oC for 15 seconds, 54oC

(D4S1548, D4S2934) or 55oC (D4S3049) for 15 seconds, and 72oC for 20 seconds,

Page 100: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

84

followed by an extension at 72oC for 2 minutes. A stop solution (95% formamide,

20mM EDTA, 0.05% bromophenol blue, 0.05% xylene cyanol FF) was added to each

reaction, heat denatured and subjected to electrophoresis on a 7% denaturing formamide

gel, which was run at 80W for 3 hours. Results were obtained following

autoradiography.

3.2.6 Immunohistochemical (IHC) Staining of Ovarian Tissue Microarrays

Ovarian tissue microarray slides (TMAs) were obtained from collaborator Dr.

Patricia Shaw and were constructed as previously described (149). The array contained

165 duplicate cores from formalin-fixed, paraffin-embedded tissue blocks from the UHN

Ovarian Tissue Bank. Of the 165 ovarian samples on the TMA, 140 were evaulable by

immunostaining, including 15 samples that were common to my familial ovarian cancer

study (4 cases with strong family history, 11 with weak family history).

For IHC, TMA slides were first subjected to microwave heat-retrieval for 20

minutes with 10 mM citrate buffer (pH 6.0), washed with PBS buffer and blocked with

0.3% hydrogen peroxide and 10% normal serum before one-hour incubation with hCDC4

antibody (GenTex) in room temperature at 1/2000 dilution. Antibody concentration was

optimized using ovarian tissue slides prior to IHC on TMAs. Slides were washed and

incubated with secondary antibody at 1:200 for 30 minutes at room temperature.

Following washes, slides were stained with streptavidin-peroxidase for an additional 30

minutes. Staining of slides was visualized using ImageScope software (Aperio

Technologies). Staining was scored blindly based on percentage of stained cells (0: none;

1: 1-24%; 2: 25-49%; 3: ≥50%), intensity of staining (0: negative; 1: light, 2: medium, 3:

Page 101: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

85

dark), and site of localization (nuclear, cytoplasmic). Total staining score is the sum of

percentage and intensity scores.

3.2.7 Quantitative Real-time PCR for CCNE1

Method for quantitative real-time PCR was as described in Chapter 2 using

Assay-on-Demand probe primers targeting CCNE1 (Applied Biosystems).

Page 102: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

86

3.3 Results

3.3.1 hCDC4 Sequence Alteration Detection by PTT Analysis

To detect mutations that lead to premature translation termination, cDNAs from

28 ovarian tumour samples (10 strong familial, 18 weak familial) were subjected to PTT

analysis. All samples exhibited the same PTT pattern of normal bands with less intense

common bands using either the 1425-bp cDNA fragment spanning exons 1-8, or the

1431-bp cDNA fragment spanning exons 4-11 (Figure 3-2). The breast cancer cell line

T47D with wild-type hCDC4 was used as positive and negative controls, respectively.

3.3.2 SSCP and Sequencing of hCDC4

To investigate DNA sequence variations within the hCDC4 gene, I first analyzed

the genomic DNA from 28 ovarian tumour samples by single-strand conformation

polymorphism analysis. One aberrantly migrated band was identified in primary tumour

OVC345, as shown in Figure 3-3. This altered band corresponded to exon 7 of hCDC4,

which encodes the first of 7 WD domains. Subsequent sequencing analysis revealed a

silent nucleotide substitution (C → T) (Figure 3-4).

Page 103: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

87

Figure 3-2. hCDC4 Protein truncation assay. In vitro transcription/translation reaction results in synthesis of hCDC4 protein fragments. A) hCDC4 fragment #1 (translation of exons 1-8) results in synthesis of a 54.3 kDa protein. Six representative ovarian tumour samples are shown in lanes 2-9. No aberrant proteins are identified. T74D breast cancer cell line, which has wild-type hCDC4, was used as reference control (lane 1). B) hCDC4 fragment #2 (translation of exons 4-11) results in synthesis of a 47.3 kDa protein. Nine representative ovarian tumour samples are shown in lanes 2-10. T47D reference control is shown in lane 1. No aberrant proteins are identified.

47.3 kDa

Ovarian Tumour Samples

1 2 3 4 5 6 7 8 9 10 B)

A)

54.3 kDa

1 2 3 4 5 6 7 8 9

Ovarian Tumour Samples

Page 104: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

88

Figure 3-3. Genetic analysis of hCDC4 with SSCP. An altered banding pattern was observed for one ovarian tumour sample (lane 4, case OVC345) for exon 7 of hCDC4. Placental DNA was used as control (lane 20.)

Ovarian Tumours

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Page 105: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

89

Figure 3-4. hCDC4 gene sequence alteration found in exon 7. A) Manual sequencing of exon 7 of hCDC4 detected a C → T nucleotide change in ovarian tumour sample OVC345. Placental DNA was used as control. B) Subsequent analysis revealed that this specific nucleotide change corresponds to a silent alteration (Asp400→Asp400, amino acid in red). Alternating colours represent protein translation from different exons. A)

A C G T A C G T G A T G A C/T A

G A

T G

A C A A

C

A C

Placenta OVC345 B) 1 MNQELLSVGSKRRRTGGSLRGNPSSSQVDEEQMNRVVEEEQQQQLRQQEEEHTARNGEVV 61 GVEPRPGGQNDSQQGQLEENNNRFISVDEDSSGNQEEQEEDEEHAGEQDEEDEEEEEMDQ 121 ESDDFDQSDDSSREDEHTHTNSVTNSSSIVDLPVHQLSSPFYTKTTKMKRKLDHGSEVRS 181 FSLGKKPCKVSEYTSTTGLVPCSATPTTFGDLRAANGQGQQRRRITSVQPPTGLQEWLKM 241 FQSWSGPEKLLALDELIDSCEPTQVKHMMQVIEPQFQRDFISLLPKELALYVLSFLEPKD 301 LLQAAQTCRYWRILAEDNLLWREKCKEEGIDEPLHIKRRKVIKPGFIHSPWKSAYIRQHR 361 IDTNWRRGELKSPKVLKGHDDHVITCLQFCGNRIVSGSDDNTLKVWSAVTGKCLRTLVGH 421 TGGVWSSQMRDNIIISGSTDRTLKVWNAETGECIHTLYGHTSTVRCMHLHEKRVVSGSRD 481 ATLRVWDIETGQCLHVLMGHVAAVRCVQYDGRRVVSGAYDFMVKVWDPETETCLHTLQGH 541 TNRVYSLQFDGIHVVSGSLDTSIRVWDVETGNCIHTLTGHQSLTSGMELKDNILVSGNAD 601 STVKIWDIKTGQCLQTLQGPNKHQSAVTCLQFNKNFVITSSDDGTVKLWDLKTGEFIRNL 661 VTLESGGSGGVVWRIRASNTKLVCAVGSRNGTEETKLLVLDFDVDMK

Page 106: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

90

3.3.3 hCDC4 Promoter Methylation Analysis

Abnormal hypermethylation within promoter regions can lead to transcriptional

silencing of a gene. To determine the methylation status in the putative promoter and 5’

region in exon 1 of hCDC4, I performed methylation-specific PCR (MSP) in all 28

ovarian tumour samples after sodium bisulfite-modification of genomic DNA. The

predicted CpG islands are shown in Figure 3-5 and a representative result of MSP

reaction is shown in Figure 3-6. For each tumour sample, I performed three separate PCR

reactions comprising of different sets of primers targeting different regions of the putative

promoter region. The sensitivity of MSP method was confirmed with universally

methylated and unmethylated control DNA. No methylation was detected in any of the

28 tumour samples.

3.3.4 Loss of Heterozygosity Analysis of hCDC4

Three different polymorphic markers in close proximity to hCDC4 were used to

evaluate LOH. Of the 28 ovarian tumours used in all of my analyses, four matched

normal samples were available for this part of study. No LOH was observed in these four

paired samples (Figure 3-7).

Page 107: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

91

Figure 3-5. Potential methylation sites within the hCDC4 promoter. A 4260-bp putative promoter region overlapping the translational start site of hCDC4 gene was analyzed via MethPrimer program. This region contains 4 CpG islands, one of which overlaps with the ATG start codon, with each having an observed/expected CpG ratio > 0.60. MethPrimer was also used to select primers for methylation-specific PCR (MSP), with CpG islands as input parameter. Graph depicts GC%, positions of CpG islands, and CpG sites. Red arrow at position 3000-bp indicates ATG start codon of hCDC4. An example of bisulfite PCR primer sets for one CpG island is depicted below. Additional primer sets for other CpG islands were also used for experiments (not shown).

Page 108: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

92

Figure 3-6. Evaluation of hCDC4 promoter methylation by methylation-specific PCR. Genomic DNA from ovarian tumours was treated with bisulfite, and standard PCR was performed using primers specific to methylated (m) an unmethylated (u) DNA to detect methylation within the hCDC4 promoter region. CpGenome Universal Methylated (m-DNA) and Unmethylated DNA (u-DNA) were used as control templates. Five representative tumour samples are shown. OVC29 OVC54 OVC109 OVC161 OVC197 m-DNA u-DNA

m u m u m u m u m u m u m u

Page 109: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

93

Figure 3-7. Loss of heterozygosity (LOH) analysis of hCDC4 in four cases of ovarian cancer. Image below illustrates LOH analysis of DNA from one ovarian tumour sample (lane 3) along with DNA from its adjacent normal tissue (lane 4). No LOH was observed in any of the ovarian samples. Breast cancer cell line T47D (lane 1) and ovarian cancer cell line OVCAR3 (lane 2) were used as method controls. 1 2 3 4

Page 110: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

94

3.3.5 hCDC4 Protein Expression in Ovarian Cancer

In order to examine hCDC4 protein expression in primary ovarian tumours, IHC

staining was conducted on ovarian tissue microarray (TMA), in collaboration with Dr.

Patricia Shaw. Optimization of hCDC4 antibody was performed using formalin-fixed

paraffin embedded (FFPE) normal breast tissue that expresses normal levels of hCDC4

and a FFPE breast cancer cell line SUM149PT mutated for hCDC4 (Figure 3-8).

Positive nuclear staining of hCDC4 was observed for normal breast tissue and negative

staining was observed for SUM149PT.

The TMA slide contains 165 ovarian tumours of various histological subtypes.

Of the 140 evaluable tumours on the array, 15 samples overlapped with those used in the

gene expression microarray study, including four ovarian tumour from patients with

strong family history and 11 tumours from those with weak family history.

Nuclear and cytoplasmic staining was scored based on staining intensity and the

percentage of stained cells (Figure 3-9). hCDC4 protein expression varies among ovarian

tumours; it was observed that ~50% of total ovarian tumours express hCDC4 at low

levels, with histological scores of 0, 1, or 2, while ~23% express at high levels, with

histological scores of 5 or 6 (Table 3-4). As expected, for those cells that express

hCDC4 protein, staining was primarily localized in the nucleus (Figure 3-9E). Due to

the small number of representative familial ovarian tumours on the TMA, however,

differential protein expression between strong and weak familial ovarian tumours was

indeterminate, with histological score of 2.5 and 2.27, respectively (p = 0.44).

Page 111: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

95

Figure 3-8. hCDC4 IHC staining optimization. A) Formalin-fixed, paraffin-embedded (FFPE) normal breast tissue stained for hCDC4 antibody shows positive nuclear staining. B) FFPE breast cancer cell line SUM149PT with hCDC4 mutation shows negative staining. (High power, 40X) Positive hCDC4 staining Negative hCDC4 staining

A B

Page 112: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

96

Figure 3-9. Immunohistochemical staining of ovarian tissue microarray with hCDC4 antibody. Representative cases of ovarian tumour specimens illustrating different IHC intensity scores: A) negative = 0; B) mild = 1; C) moderate = 2; D) high = 3. E) Close up image of D. As expected, hCDC4 protein is predominantly localized in the nucleus (arrow).

A B

C D

E

100 µm 100 µm

100 µm 100 µm 50 µm

Page 113: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

97

Table 3-4. Histological scores of hCDC4 immunohistochemical staining on ovarian tissue microarray. Various levels of hCDC4 protein expression are observed across different histological subtypes of ovarian cancer. Histological score represents sum of stain intensity (0-3) and % of stained cells (0, 1: 1-24%, 2: 25-49%, or 3: 50-100%). *MMMT=Malignant Mixed Mullerian Tumour

Histological Score Histological Subtype 0 1 2 3 4 5 6

Total # of Tumours

Clear Cell 7 (58%) 0 0 0 3 (25%) 0 2 (17%) 12 Endometrioid 2 (17%) 1 (8%) 1 (8%) 1 (8%) 2 (17%) 1 (8%) 4 (33%) 12 Mixed 2 (20%) 0 2 (20%) 2 (20%) 1 (10%) 3 (30%) 0 10 MMMT* 2 (66%) 0 0 0 0 1 (33%) 0 3 Mucinous 4 (33%) 0 2 (17%) 0 3 (25%) 1 (8%) 2 (17%) 12 Serous 32 (40%) 0 11 (14%) 8 (10%) 12 (15%) 11 (14%) 6 (8%) 80 Transitional Cell 1 (50%) 0 0 0 1 (50%) 0 0 2 Undifferentiated 0 0 0 1 (100%) 0 0 0 1 Other/Unknown 3 (38%) 0 1 (20%) 2 (25%) 1 (20%) 1 (20%) 0 8 Total 53 1 17 14 23 18 14 140

Page 114: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

98

3.3.6 Gene Expression of CCNE1 in Familial Ovarian Cancer

As mentioned earlier, hCDC4 is involved in the degradation of cell cycle

regulator protein cyclin E; therefore, an evaluation of cyclin E protein expression in the

same ovarian tumours would be useful in determining if decreased levels of hCDC4

expression correlate with increased cyclin E expression levels. However, due to the lack

of ovarian tumour samples for immunohistochemical analysis, the protein expression

levels of cyclin E could not be determined. Nonetheless, the deregulation of cyclin E at

the transcriptional level has also been previously observed in ovarian cancer (81).

Therefore, I sought to examine CCNE1 gene expression levels in familial ovarian

tumours. Since the CCNE1 gene was not represented on the microarray platform used for

my familial ovarian cancer gene expression profiling analysis (Chapter 2), to assess and

compare the gene expression levels of cyclin E in tumours from ovarian cancer patients

with strong and weak family history, I performed quantitative real-time PCR using

mRNA extracted from ovarian tumour samples. As seen in Figure 3-10, the average gene

expression of CCNE1 observed in the weak familial group was two-fold higher than the

average expression seen in the strong familial group (p < 0.05).

Page 115: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

99

Figure 3-10. CCNE1 gene expression in familial ovarian cancer. The mean gene expression of CCNE1 in familial ovarian tumours was evaluated using quantitative real-time PCR. Tumours from patients with weak family history of breast and/or ovarian cancer (18 samples) expressed a higher level of CCNE1 compared to the strong family history group (10 samples). (n=3, p < 0.05).

0

0.5

1

1.5

2

2.5

3

3.5

4

Strong Familial Weak Familial

Rel

ativ

e C

CN

E1

Exp

ress

ion

Page 116: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

100

3.4 Discussion

As described in Chapter 2, we have identified hCDC4 as a differentially

expressed gene among ovarian cancer patients with strong and weak family histories of

breast and/or ovarian cancer using the data from my microarray analysis of familial

ovarian cancer. More specifically, ovarian tumours from the strong family history group

exhibited lower hCDC4 expression compared to the weak familial group (p < 0.05). This

observation was of great interest as accumulating evidence from recent years has

suggested human F-box protein hCDC4 to be a tumour suppressor. Functionally, it

mediates the ubiquitin-dependent proteolysis of various oncoproteins involved in cell

division and cell fate determination, thus its deregulation can lead to tumourigenesis

(207).

The direct role of hCDC4 in relation to ovarian cancer was first implicated in a

study conducted by Moberg et al., in which hCDC4 mutations (including nonsense,

missense, and frameshift mutations), were detected in various ovarian cancer cell lines

(205). Based on Moberg’s observations, I sought to examine the mutation status of

hCDC4 in familial ovarian tumours.

From my genetic screening of the 28 primary ovarian cancers using SSCP, I

observed one nucleotide change within exon 7 of one ovarian tumour sample that belongs

to the strong familial ovarian cancer group (OVC345). This initial finding was promising

since exon 7 encodes one of the WD domains responsible for substrate recognition.

However, subsequent DNA sequencing revealed a nucleotide change that did not alter the

hCDC4 protein sequence. This specific previously unreported alteration may be a

significant event nevertheless, as previous studies have suggested that synonymous

Page 117: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

101

polymorphisms can affect the timing of cotranslational folding of proteins (208), thus it is

possible that this change may alter the structure of substrate recognition site of hCDC4.

However, this hypothesis remains to be investigated.

Simultaneously to my genetic analyses of hCDC4, other groups have also sought

to examine the mutation status of hCDC4 in human primary ovarian cancers. In a

comprehensive genetic screen of over 1500 human tumours, Akhoondi and colleagues

observed that hCDC4 is mutated in a variety of human malignancies, with an overall

mutation frequency of ~6% (207). However, none of the 32 primary ovarian tumours

from the study exhibited any hCDC4 mutations (207). Similarly, Kwak et al. reported in

their study comprising 111 primary ovarian tumours that mutations of hCDC4 is a rare

event in ovarian cancer (209). More specifically, they observed 2/95 (~2%) sporadic

ovarian cancer cases that harbour mutations, while no mutations were observed in any of

the 16 cases of familial ovarian cancer (209).

Based on the results from my current study, as well as other studies mentioned

directly above, it appears that hCDC4 mutational inactivation is an uncommon event in

primary ovarian tumours. However, other gene regulation mechanisms, such as promoter

hypermethylation, may be responsible for the decreased hCDC4 expression observed in a

subset of familial ovarian tumours, as numerous reports have investigated the importance

of epigenetics in the inactivation of tumour suppressor genes involved in cancer

development, including ovarian cancer (210, 211). To explore this possibility, I

examined the methylation status of the hCDC4 promoter region in familial ovarian

tumours.

Page 118: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

102

While multiple potential methylation sites were identified for the promoter region

of this gene, no methylation was detected in any of the ovarian tumours analyzed, thereby

suggesting that promoter methylation may not be an important mechanism for hCDC4

repression in familial ovarian cancer. Recently, a study regarding the epigenetics of

hCDC4 promoter in glioma cell lines found that the low gene expression of hCDC4-β

(the hCDC4 isoform expressed in the brain) seen in various glioma cell lines was not due

to promoter hypermethylation (212). Interestingly, they also showed that normal

peripheral blood cells, which dominantly express α- and γ-isoforms of hCDC4, exhibited

increased methylation within the hCDC4-β promoter (212). Therefore, while methylation

may not be a significant event in repressing hCDC4 gene expression in cancer cells, it

nevertheless plays a role in controlling its tissue- and isoform-specific gene expression.

The hCDC4 gene is suggested to be a haploinsufficient tumour suppressor gene,

and this stems from the observation that in many primary tumours and derived cell lines,

hCDC4 mutations occur without a concomitant loss or additional mutations in the second

allele (207). Moreover, it has been demonstrated that a loss of a single copy of this gene

is sufficient for tumour development in p53+/- mice (213). Therefore, a loss of a single

copy of hCDC4 in human cancers may occur through gene deletion and this deletion may

explain its decrease in gene expression. Indeed, the hCDC4 gene maps to chromosome

4q31.3, a region that is deleted in approximately 30% of human malignancies, including

ovarian cancer (207, 214). Furthermore, LOH within the hCDC4 locus has previously

been reported in esophageal adenocarcinoma and gastric carcinoma (215, 216). Since

currently there are no reports of hCDC4 LOH in human ovarian cancer, I attempted to

examine the LOH status of the familial ovarian tumours from my study. Four ovarian

Page 119: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

103

cancer cases with matched normal samples were available for LOH analysis, but no LOH

was detected. However, attributable to the very small sample size, the significance of

hCDC4 LOH in familial ovarian cancer remains inconclusive and requires further

investigations using a larger collection of samples.

In an attempt to address whether the differential gene expression observed

between strong and weak familial ovarian tumours correlates with hCDC4 protein

expression, immunohistochemical analysis was performed with ovarian TMA. The

limited number of corresponding familial ovarian tumours represented on the TMA,

however, prevented a robust analysis of this specific comparison. Nonetheless, our IHC

analysis revealed that hCDC4 expression varies among ovarian tumours. Interestingly,

low hCDC4 protein expression was observed in a majority of clear cell tumours,

mucinous tumours, malignant mixed mullerian tumours, as well as serous carcinomas

(subtype from current familial ovarian cancer study). The low expression seen in these

tumours indicates that loss of hCDC4 may be a significant event in the development of

these tumour subtypes. Further correlations on the expression of downstream effectors of

hCDC4 may reveal the specific pathways that are affected in these tumours.

As mentioned earlier, hCDC4 plays a critical role in the proteolytic regulation of

various proteins, including cell cycle regulator cyclin E1 (CCNE1), and the accumulation

of cyclin E1 due to deregulation by hCDC4 has certainly been associated with an increase

in chromosomal instability, an important event that contributes to malignant

transformation and cancer progression (193, 202). Indeed, it has previously been shown

that the cyclin E gene is frequently amplified and over-expressed in ovarian tumours at

Page 120: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

104

both the RNA and protein levels, and that its expression is negatively correlated with

survival, thus implicating its importance in ovarian cancer development (80, 217, 218).

Based on my hCDC4 gene expression results, as well as its characterized role in

cyclin E1 regulation, I hypothesized that in strong familial cases of ovarian cancer which

express a low level of hCDC4, would have an increased protein expression of cyclin E1.

For that reason, I was interested in examining the protein expression of cyclin E1 in a

large collection of ovarian tumour samples, as it would be ideal to be able to examine if

its expression negatively correlates with hCDC4 expression. However, due to the lack of

TMA for cyclin E1 staining, I was unable to perform this particular experiment.

Nevertheless, I sought to examine the gene expression of CCNE1 in my set of ovarian

tumour samples, since ~20% of ovarian cancers have previously been shown to have

increased CCNE1 mRNA (219). My results showed that the mRNA expression of

CCNE1 was two fold higher in the weak familial ovarian group compared to the strong

familial group (p < 0.05). This is an interesting observation, as it suggests that

expression of cyclin E1 in ovarian cancer may be regulated differently at the gene and

protein levels, according to family history status.

Based on the expression data of hCDC4 and CCNE1, it can be speculated that

there may be some yet to be defined heritable factors (eg. SNP alleles) in strong familial

ovarian cancer that play roles in decreasing the gene expression of hCDC4, which may

result in increased steady-state levels of cyclin E1 protein. Additionally, other genetic

alterations such as gene amplification or increased transcriptional activities may be

responsible for increasing CCNE1 gene expression in weak or non-familial ovarian

cancers where hCDC4 expression is not altered. Previous reports have strongly

Page 121: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

105

suggested that proper regulation of cyclin E1 is important in preventing malignant

transformation in ovarian cancer (220-222); however, whether or not heritable genetic

alterations that can affect cyclin E1 activity can predispose individuals to an earlier onset

of ovarian cancer requires further investigation.

The data presented in this Chapter suggested that deregulation of hCDC4 gene

expression may be important in a subset of ovarian cancers in patients with a strong

family history. However, the mechanism(s) responsible for this altered expression

remain elusive. Thus further research exploration using larger sample size of familial

ovarian tumours may reveal additional information regarding the genetic alterations or

regulation of hCDC4.

Page 122: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

CHAPTER 4

Characterization of PRKCZ in Ovarian Cancer

All experiments and analysis presented in this Chapter was performed by KS.

106

Page 123: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

107

4.1 Introduction

As discussed earlier in Chapter 2, my microarray study of familial ovarian cancer

has identified a set of differentially expressed genes between tumours from patients with

strong and weak family history of breast and/or ovarian cancer. Among these genes,

PRKCZ was identified as more highly expressed in patients with strong family history,

and I have chosen this gene for further analyses.

PRKCZ encodes a protein belonging to the atypical subclass of the protein kinase

C family of serine/threonine kinases that has been implicated in the regulation of cellular

transformation and carcinogenesis (172). PRKCZ has previously been observed to be

involved in multiple signal transduction pathways, including activation of the

ERK/MAPK cascade, p70 ribosomal S6 kinase signalling cascade, transcription factor

NF-κB, as well as regulation of cell polarity (175). The regulation of these pathways

may explain some of the mechanisms by which PRKCZ can promote human cancers.

Indeed, the roles of PRKCZ in various cancer types have been examined in recent years.

For example, it was reported that PRKCZ expression level is two fold higher in

glioblastoma cell lines compared with normal astrocytes (178). Subsequent studies

showed that this high level of expression is correlated with increased proliferation of

glioblastoma cells, while reduced expression is correlated with inhibition of migration

and invasion (174, 223). The involvement of activated PRKCZ in epidermal growth

factor (EGF) -induced chemotaxis has also been examined in lung and breast cancer, and

it was shown that PRKCZ is able to elicit a migration response of these cells by acting as

a downstream mediator in the phosphatidylinositol 3-kinase (PI3K)/AKT pathway (176,

177). As mentioned above, PRKCZ participates in cell polarity pathways, and studies

Page 124: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

108

have illustrated that loss of cell polarity, which results in tissue disorganization, may

contribute to cancer development (224). Interestingly, it has been observed that PRKCZ

is mislocalized in a subset of ovarian cancers, and it was suggested that this

mislocalization may reflect a role for apical-basal loosening, thus disrupting cell-cell

adhesion, as well as increasing cell growth (173); however, additional evidence

supporting the role of PRKCZ in ovarian cancer remains limited.

The studies mentioned above clearly suggest the importance of PRKCZ in cancer

progression, thus providing rationale for further analyses. In this Chapter, I describe the

various in vitro functional experiments that I have performed in order to characterize its

role in ovarian cancer, including cell viability, cell migration, as well as downstream

signalling pathways in which it may be participating.

Page 125: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

109

4.2 Materials and Methods 4.2.1 Cell Culture

Ovarian cancer cell lines SKOV3 and OVCAR3 were purchased from American

Type Culture Collection (Manassas, VA). OVCAR3 cells were maintained in RPMI-

1640 medium supplemented with 20% FBS and 0.01 mg/ml bovine insulin. SKOV3

cells were maintained in McCoy’s medium supplemented with 10% FBS. The HEY cell

line was kindly provided by Dr. Theodore Brown (Samuel Lunenfeld Research Institute,

Toronto, ON) and was maintained in MEM-alpha medium containing 10% FBS. All

cells were incubated at 37oC in a humidified atmosphere of 5% CO2 and 95% air.

4.2.2 PRKCZ Expression Vector & Generation of Stable Clones

PCR conditions to amplify human PRKCZ in a 25 µL reaction volume were as

follows: 2.5 µL of 10X Platinum HiFidelity Buffer (Invitrogen), 1.5 µL of 10 mM dNTPs

(Invitrogen), 1.0 µL of 50 mM MgSO4 (Invitrogen), 0.3 µL of 30 µM EcoRI-tagged

forward primer, 0.3 µL of 30 µM SalI-tagged reverse primer, 0.5 µL of Platinum

HiFidelity Taq Polymerase (5U/µL, Invitrogen), 1 µL (50 ng) of pooled cDNA (Table 2-

2), and 17.9 µL of ddH2O. Thermal cycling parameters were as follows: initial

incubation for 2 minutes at 94oC; 40 cycles of 30 seconds at 94oC, 30 seconds at 73oC, 2

minutes at 72oC. PCR products were resolved by 1.0% agarose gel electrophoresis,

visualized under UV, and gel extracted and purified according to the manufacturer’s

protocol (Qiagen). Subsequently, they were transferred to pEGFP-N2 (N-terminal GFP

tag) expression vector (Clontech). Correct PRKCZ sequence within vector was

confirmed by sequencing. Each cell line was transfected with the plasmid vectors

Page 126: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

110

PRKCZ-pEGFP or vector controls, using Fugene ® 6 Transfection Reagent (Roche).

Following transfection, cells were cultured with G418 sulfate (400 μg/ml for HEY, 800

μg/ml for SKOV3, 500 μg/ml for OVCAR3). Surviving colonies were individually

selected and maintained in G418 sulfate-containing medium.

4.2.3 Quantitative Real-Time PCR

Primer pairs for genes of interest were designed individually by using Primer3

input software (Whitehead Institute, Howard Hughes Medical Institute, NIH).

Quantitative real-time RT-PCR was performed on an ABI Prism 7000 Sequence Detector

(Applied Biosystems) using SYBR Green PCR Master Mix (Applied Biosystems). Each

of the 20 µL PCR reactions contained 1 µL (50 ng) of cDNA and 0.45 µM of each of the

primers. The thermal cycles for PCR reaction were as follow: initial denaturation for 10

minutes at 95oC, followed by 40 cycles of 95oC for 15 seconds, and annealing extension

at 60oC for 1 minute. The housekeeping gene HPRT1 was used to normalize gene

expression values. Reference cDNA was used to generate standard curve to quantify

cDNA levels of samples and consisted of a pool of 13 cell lines, as described in Chapter 2

(Table 2-2).

4.2.4 Western Blotting

Cell extracts were prepared as followed. Cells were washed three times with cold

phosphate-buffered saline (PBS), lysed with NETN lysis buffer (20mM Tris-HCl, pH 7.5;

150mM NaCl; 1 mM EDTA, pH 8.0; 0.5% Nonidet P-40; 1 mM PMSF; 1x protease and

phosphatase inhibitors) on ice for 10 minutes and centrifuged for 8 minutes at 12,000 rpm

Page 127: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

111

to separate lysates from cell debris. Protein concentrations were determined with BCA

Protein Assay Kit (Pierce). Equal amounts of protein from different cell lines were

loaded and separated on 8-10% SDS-PAGE. Proteins were transferred to Hybond ECL

nitrocellulose (Amersham) and blotted using anti-PRKCZ, anti-IGF1R, anti-ITGB3

antibodies (Cell Signalling) at 1:1000 dilutions. Secondary conjugates, HRP-Donkey

anti-mouse or HRP-Donkey anti-rabbit (Jacksons Immunochemicals) were incubated for

1 hour at a 1:5000 dilution. Protein bands were visualized by chemiluminescence using

ECL detection system (Amersham).

4.2.5 MTT Cell Viability Assays

When added to viable cells, MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-

diphenyltetrazolium bromide, a yellow tetrazole), is reduced to purple formazan by

activated reductase enzymes. Thus the MTT assay can be applied to assess cell viability

based on changes in absorbance of coloured solution. In brief, cells were seeded in 96-

well plates at a concentration of 1000 cells/well with a final volume of 100 µL of culture

media and were incubated at 37oC, with or without myristoylated pseudosubstrate peptide

(40 µM), a PRKCZ inhibitor. After each incubation period, 10 µL of the MTT labelling

reagent (Roche) were added to each well at a final concentration of 0.5 mg/mL. Cells

were incubated for an additional 4 hour period, followed by addition of 100 µL of

solubilization solution (10% SDS in 0.01 M HCl). Plates were allowed to stand overnight

at 37oC and the spectrophotometrical absorbance of the samples was measured using a

microplate (ELISA) reader at a wavelength of 570 nm with background subtraction at

630 nm. Each assay was performed in triplicates.

Page 128: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

112

4.2.6 TUNEL Assays

Cells were plated and allowed to grow to confluency in 96-well plates, and serum

starved in 0.1% FBS media for 20 hours, then for an additional 4 hours with or without

TRAIL-induction of apoptosis (100ng/ml). Cells were fixed with 4% paraformaldehyde

in PBS for 1 hour, followed by permeabilization with 0.1% Triton X-100/0.1% sodium

citrate on ice for 2 minutes. TUNEL assay was performed with an In Situ Cell Death

Detection Kit (TMR red) as described by the manufacturer (Roche). Reactions were

stopped after one hour, and apoptotic cells were visualized and measured using IN CELL

Analyzer 1000 (GE Healthcare Life Sciences). Each assay was performed in triplicates.

4.2.7 BrdU Proliferation Assay

Approximately 1 x 105 cells were plated onto coverslips within 6-well plates and

allowed to grow to ~60% confluency overnight. On the next day, cells were incubated in

10uM of BrdU for 6 hours, washed with PBS, then fixed with 4% paraformaldehyde,

denatured with 2M HCl, and neutralized with 0.1 M sodium borate. Cells were then

incubated with mouse anti-BrdU (1:200, Dako) for 1 hour, washed, followed by

incubation with Alexa Fluro 647 anti-mouse (Molecular Probes) for an additional 30

minutes, and counterstained with DAPI. BrdU incorporation was observed and counted

in five fields of view per well through microscopy. Each cell line was performed in

triplicates per experiment and the ratio of BrdU-positive cells to total cell number was

calculated.

Page 129: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

113

4.2.8 Matrigel Transwell Assays

In vitro cellular invasion was assayed by determining the ability of cells to invade

through Matrigel (VWR), a synthetic basement membrane, as previously described (177).

In brief, Transwells of 8 µm pore size (Corning) coated with ~30 µg Matrigel (diluted in

ice-cold PBS) were placed in a modified Boyden chamber and ovarian cancer cells (1 x

105 per well) were plated onto each of the transwells in serum-free media. The bottom

chambers were either filled with epidermal growth factor (10ng/ml) in serum-free media,

or serum-containing media (10% FBS), which both acted as a chemoattractant. Cells

were incubated at 37oC and allowed to migrate through the matrigel for 24 hrs. After

incubation, cells were collected from the bottom chamber, as well as the underside of the

Transwell by briefly rinsing transwells in PBS and placing them in fresh wells containing

500 µL trypsin (0.1%). Solutions were microcentrifuged at 8000 g for 5 minutes to

pellet invaded cells. CyQUANT dye (Molecular Probes), which shows strong

fluorescence enhancement when bound to nucleic acids, was used for the quantification

of invaded cells. Fluorescence was measured using a multiwell fluorescence plate reader

with excitation at 485 nm and emission at 530 nm.

4.2.9 Scratch Wound Healing and Pericentrin Orientation Assays

Each cell line (HEY, SKOV3, OVCAR3, and their respective clones) were plated

into 6-well plates and cultured to confluence. Cells were rinsed with PBS and serum

starved overnight in 0.5% FBS media at 37oC and 5% CO2. Next day, three separate

scratches were introduced through the monolayer of cells in each of the wells using

sterile 200 µL plastic pipette tips. Cells were then rinsed gently with PBS to remove

Page 130: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

114

cellular debris and replaced with fresh culture media supplemented with 0.5% FBS. The

wounded cells were allowed to incubate at 37oC and representative fields were

photographed with an inverted-phase microscope at different time intervals. For

pericentrin orientation assay, cells were fixed with 4% paraformaldehyde in PBS four

hours after wound scratch, followed by one hour incubation with blocking solution (0.2%

Triton X-100, 1.5% BSA, 5% serum), and stained with anti-pericentrin and anti-tubulin

antibodies (1/5000 dilutions) for one hour at room temperature and counterstained with

DAPI. Between incubation steps, cells were washed several times with PBS. Cells with

microtubule organizing centre (MTOC; stained with anti-pericentrin) situating within

quadrants facing the wound were scored as positive for polarity (225).

4.2.10 Phagokinetic Track Assays

Single cell motility of HEY, OVCAR3 and SKOV3 were measured according to

Cellomics ® Cell Motility Kit’s protocol (Thermo Scientific), as previously described.

Briefly, 96-well plates were coated with fibronectin (1 μg/well) overnight followed by

fluorescent microspheres the next day. Approximately 500 cells were plated in each well

in 100 μl of serum-free media. Following 16 hours of incubation, at 37oC in a humidified

atmosphere of 5% CO2 and 95% air, wells were fixed with 5.5% formaldehyde and cells

were stained with phalloidin. The area of a phagokinetic track from a single cell was

quantified using Cellomics Arrayscan II microscope and software.

Page 131: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

115

4.2.11 siRNA Transfections

Knockdown of expression of PRKCZ, IGF1R, and ITGB3 in ovarian cancer cell

lines was achieved by transfection of siRNAs (Ambion). siRNAs targeting these genes

was performed with Dharmafect-4 transfection reagent (Dharmacon). In brief, cells were

seeded in 12-well or 6-well plates at densities of 1 x 105 or 2 x 105 cells/well,

respectively. Cells were then treated with siRNA transfection mixtures following the

manufacturer’s protocol. Scrambled siRNA (Ambion) was used as a control. Additional

controls included mock-treated cells that received transfection reagent without siRNA, as

well as untreated cells that received only fresh media. Cells were harvested after 48 or 72

hours for RNA and protein extraction, respectively.

4.2.12 Ingenuity Pathway Analyses

Ingenuity Pathway analyses were performed as described in Chapter 2.

4.2.13 Statistical Analyses

All data were represented as means ± the standard deviation (SD) of the mean.

Statistical calculations were performed with Microsoft Excel analysis tools. Differences

between groups were analyzed by student t-test. P values of < 0.05 were considered

statistically significant.

Page 132: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

116

4.3 Results To characterize the roles of PRKCZ in ovarian cancer, I developed an in vitro

ovarian cancer model with altered expression of PRKCZ by generating stable cell lines

that over-express PRKCZ, as well as gene knockdown by PRKCZ-targeted siRNA,

followed by various functional assays. Three serous ovarian cancer cell lines HEY,

SKOV3 and OVCAR3 were chosen for my initial analyses; however, based on my initial

results, as well as its moderate migratory phenotype (versus extremely motile and non-

motile characteristics of HEY and OVCAR3 cells, respectively), most of the later

experiments focused on the SKOV3 cell line.

4.3.1 Generation of PRKCZ Stable Ovarian Cancer Cell lines

I first examined the endogenous transcript levels of PRKCZ in HEY, SKOV3, and

OVCAR3 ovarian cancer cell lines by performing quantitative RT real-time PCR. Levels

of PRKCZ transcript varied among cell lines, with HEY cells expressing the lowest level

of PRKCZ, followed by SKOV3 and OVCAR3 (Figure 4-1a). Protein levels of PRKCZ

in all three cell lines were very low compared to THP-1 cells, a human acute monocytic

leukemia cell line that expresses high endogenous levels of PRKCZ (Figure 4-1b).

Page 133: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

117

Figure 4-1. Endogenous transcript and protein levels of PRKCZ in selected ovarian cancer cell lines. a) HEY, SKOV3, and OVCAR3 cells have variable gene expression levels of PRKCZ. HEY cell line has the lowest PRKCZ transcript level, while SKOV3 and OVCAR3 express have similar transcript levels. b) PRKCZ protein is expressed poorly among all three cell lines, as determined by western blot analysis. Lysate from each cell line was loaded in duplicates in adjacent lanes. Human acute monocytic leukemia cell line THP-1, which expresses a high level of PRKCZ protein, was used as a positive control. The beta-actin loading control is also shown (lower panel). a)

0

0.5

1

1.5

2

2.5

HEY SKOV3 OVCAR3

Ovarian Cancer Cell Line

Rel

ativ

e E

xpre

ssio

n

b)

HEY SKOV3 OVCAR3 THP-1

PRKCZ

Beta-actin

Page 134: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

118

After PRKCZ transfection and selection of stable clones, the gene and protein

expression of PRKCZ stable clones for each ovarian cancer cell line were verified by

quantitative real-time PCR and western blot analyses using a PRKCZ-antibody (Figures

4-2, 4-3, 4-4). The expected differences in PRKCZ expression levels between non-

transfected or empty-vector transfected and PRKCZ-transfected cells were observed. A

band of ~95 kDa was detected in stable cell lines that over-express PRKCZ, which

corresponded with molecular weight of GFP-tagged PRKCZ protein. Additionally,

fluorescent microscopy was used to detect exogenous expression and revealed a

cytoplasmic localization of GFP-tagged PRKCZ in transfected cell lines (Figures 4-2, 4-

3, 4-4).

Page 135: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

119

Figure 4-2. Expression of PRKCZ clones in HEY ovarian cancer cell line. a) Comparison of PRKCZ transcript and protein expression in HEY parental control (PC), control vector (N2), and a selection of PRKCZ clones by a) quantitative real-time RT-PCR, b) western blot analyses with PRKCZ-antibody, and c) fluorescence microscopy (magnification: 20x). In b), a band of ~95 kDa was detected by western blotting in stable cell lines that over-express PRKCZ, which corresponded with molecular weight of GFP-tagged PRKCZ protein. A faint band of ~67 kDa corresponded to endogenous PRKCZ. Shown are HEY parental cells, HEY vector control cells, and HEY-PRKCZ clones #1, #2 and #3 that were chosen for future functional experiments. a)

0

10

20

30

40

50

60

70

~95 kDa

~67 kDa

HEY HEY N2 HEY PRKCZ #1 HEY PRKCZ #2 HEY PRKCZ #3

Rel

ativ

e E

xpre

ssio

n

PC N2 PRKCZ#1 PRKCZ#2 PRKCZ#3

b) PRKCZ clones PC N2 #1 #2 #3 PRKCZ Beta-actin

Page 136: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

120

c)

HEY HEY-N2 HEY-PRKCZ

GFP-PRKCZ GFP

Page 137: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

121

Figure 4-3. Expression of PRKCZ clones in SKOV3 ovarian cancer cell line. Comparison of PRKCZ transcript and protein expression in SKOV3 parental control (PC), control vector (N2), and a selection of PRKCZ clones by a) quantitative real-time RT-PCR, b) western blot analysis with PRKCZ-antibody, c) fluorescence microscopy (magnification: 20x). In b), a band of ~95 kDa was detected by western blotting in stable cell lines that over-express PRKCZ, which corresponded with molecular weight of GFP-tagged PRKCZ protein. A faint band of ~67 kDa corresponded to endogenous PRKCZ. Shown are SKOV3 parental cells, SKOV3 vector control cells and SKOV3-PRKCZ clones #1, #2, #4, #8 that were chosen for future functional experiments. a)

0

50

100

150

200

250

300

S

~95 kDa

~67 kDa

KOV3 SKOV3 N2 SKOV3 SKOV3 SKOV3 SKOV3PRKCZ#1 PRKCZ#2 PRKCZ#4 PRKCZ#8

Rel

ativ

e E

xpre

ssio

n

PC N2 PRKCZ#1 PRKCZ#2 PRKCZ#4 PRKCZ8

b) PRKCZ clones PC N2 #1 #2 #4 #8

PRKCZ Beta-actin

Page 138: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

122

c) SKOV3 SKOV3-N2 SKOV3-PRKCZ

GFP GFP-PRKCZ

Page 139: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

123

Figure 4-4. Expression of PRKCZ clones in OVCAR3 ovarian cancer cell line. Comparison of PRKCZ transcript and protein expression in OVCAR3 parental cells (PC), control vector (N2), and a selection of PRKCZ clones by a) quantitative real-time RT-PCR, b) western blot analyses with PRKCZ-antibody, and c) fluorescence microscopy (magnification: 20x). In b), a band of ~95 kDa was detected by western blotting in stable cell lines that over-express PRKCZ, which corresponded with molecular weight of GFP-tagged PRKCZ protein. A faint band of ~67 kDa corresponded to endogenous PRKCZ. Shown are OVCAR3 parental cells, OVCAR3 vector control cells and OVCAR3-PRKCZ clones #1, #2, and #3 that were chosen for future functional experiments. a)

0

2

4

6

8

10

12

14

~95 kDa

~67 kDa

OVCAR3 OVCAR3 N2 OVCAR3 OVCAR3 OVCAR3PRKCZ#1 PRKCZ#2 PRKCZ#3

Rel

ativ

e E

xpre

ssio

n

PC N2 PRKCZ#1 PRKCZ#2 PRKCZ#3

b) PRKCZ clones PC N2 #1 #2 #3

PRKCZ

Beta-actin

Page 140: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

124

c) OVCAR3 OVCAR3-N2 OVCAR3-PRKCZ GFP GFP-PRKCZ

Page 141: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

125

4.3.2. Cell Viability in PRKCZ-Expressing Cells

PRKCZ has previously been shown to be involved in cell survival in various cell

types (226-230). To assess whether it has the same effect on ovarian cancer cells, I first

performed MTT cell viability assays. By comparing the growth of parental, empty-vector

control, and PRKCZ-expressing cells, I observed that PRKCZ did not have an effect on

HEY or OVCAR cell lines. On the other hand, it significantly increased the viability of

SKOV3 cells, and this effect was abolished by the addition of myristoylated

pseudosubstrate peptide that targets PRKCZ (Figure 4-5). This result suggests that

PRKCZ can enhance cell survival in a subset of ovarian cancer cells.

I then further investigated whether the increased cell viability seen in SKOV3

cells that over-express PRKCZ was due to an increase in proliferation or a decrease in

apoptosis. BrdU proliferation assay was performed to examine if PRKCZ over-

expression results in increased replication rate in SKOV3 cells. As seen in Figure 4-6,

SKOV3 PRKCZ-clones displayed a higher percentage of cells with BrdU incorporation,

indicating that the rate of proliferation in these cells is higher than parental and empty

vector controls.

Page 142: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

126

Figure 4-5. PRKCZ increases cell viability in SKOV3 cells but not HEY and OVCAR3 cells. a) Increased cell viability was observed in cells over-expressing PRKCZ, as measured by MTT cell viability assay. This effect was abolished by the addition of PRKCZ myristoylated pseudosubstrate (PS). No significant change in cell viability was observed in b) HEY and c) OVCAR3 cells. (n=3) N2 = empty vector N2

0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

2 4 6 8 10 12Day

MT

T A

bsor

banc

e U

nit

OVCAR3

N2

PRKCZ#1

PRKCZ#2

PRKCZ#3

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1 2 3 4 5 6

Day

MT

T A

bsor

banc

e U

nit

SKOV3

N2

PRKCZ#1

PRKCZ#4

SKOV3 (+PS)

N2 (+PS)

PRKCZ#1 (+PS)

PRKCZ#4 (+PS)

a)

*p<0.05 *

* b)

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1 2 3 4

MT

T A

bsor

banc

e U

nit

Day

HEY

N2

PRKCZ#1

PRKCZ#2

c)

Page 143: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

127

Figure 4-6. PRKCZ enhances proliferation of SKOV3 ovarian cancer cells. BrdU incorporation assay was performed to measure prolifartion rate of SKOV3 cells. PRKCZ-transfected cells showed an increase in BrdU-positive cells, indicating increased cell growth compared to controls (*p<0.01). This effect was reversed by the addition of PRKCZ myristyolated pseudosubstrate (PS). PC = parental control; N2 = empty vector control N2. (n=3)

0.00

0.05

0.10

0.15

0.20

0.25

0.30

RKCZ#1 PRKCZ#4 PRKCZ#8

TM

R:D

api C

ount

EV P

s

No TrailTrail (100ng/ml)

N2

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

l N2 PRKCZ#1 PRKCZ#4

Brd

U P

ositi

ve C

ells

(%)

* No PS *+PS

Parenta PC Figure 4-7. PRKCZ has no effect on apoptosis in SKOV3 cells. Apoptotic-related DNA fragmentation of SKOV3 cells with and without Trail treatment (100ng/ml) was analyzed with In Situ Cell Death Detection Kit. No significant difference was seen between empty-vector control and PRKCZ clones. (n=2) N2 = empty vector control N2.

Page 144: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

128

Additionally, I sought to examine the effect of PRKCZ on apoptosis in SKOV3

cells by performing TUNEL assays. Since cells from the MTT assays were not induced

by any apoptotic agents, I performed TUNEL assays in non-treated cells to examine if

there is a difference in non-induced apoptosis between PRKCZ-expressing and control

cells. Additionally, I was interested in examining if PRKCZ can protect ovarian cancer

cells against induced apoptosis. To achieve this, cells were treated with TRAIL

(100ng/ml) for 4 hours, and apoptotic-induced DNA fragmentation was detected. As

seen in Figure 4-7, while a significant difference in cell death was observed between non-

induced and induced cells, there was no difference in apoptosis between control and

PRKCZ-expressing cells in either case. Based on these results, it can be speculated that

the increased cell viability seen in PRKCZ-expressing SKOV3 cells is most likely due to

an increase in cell proliferation.

4.3.3 PRKCZ and ovarian cancer cell migration and invasion

The ability of cancer cells to migrate is one of the key processes in cancer

progression. In order to examine whether PRKCZ can increase the migratory properties

of ovarian cancer cells, I undertook various experimental approaches. Microscopic

observations revealed that the morphology of each of these cell lines is associated with

their respective migration properties. HEY cells are fibroblast-like cells with filopodia

that tend to undergo single-cell migration and are highly motile; while SKOV3 cells are

also fibroblast-like, they are less motile compared to HEY. OVCAR3 cells, on the other

hand, have cobblestone-like structures, they migrate as a sheet and are the least motile.

Page 145: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

129

No invasive differences were observed between parental and PRKCZ-expressing

HEY and SKOV3 cells in transwell migration assays. This specific assay was also

performed for OVCAR3 cells, but due to its low rate of migration, results were

inconclusive. Additional scratch wound healing migration assays were performed to

examine effect of PRKCZ on migratory properties of all three cell lines. No migration

differences were observed in any of these cells when PRKCZ-transfected cells were

compared with controls (Figure 4-8).

To examine if the endogenous levels of PRKCZ in parental cells are sufficient for

their migration, I repeated wound healing assays with the same ovarian cell lines that

have been subjected to PRKCZ siRNA knockdown (Figure 4-9). I observed that

knocking down the transcript levels of PRKCZ can in fact decrease migration rate of

SKOV3 cells. This phenotype was not observed in HEY and OVCAR3 cell lines (data

not shown).

Interestingly, despite the lack of difference in migration rate, a distinct

morphological phenotype was observed in HEY cells that over-express PRKCZ in the

scratch wound healing assay. It was noted that while HEY parental or empty-vector

control cells were both able to migrate to re-establish the monolayer and that their

movements were primarily perpendicular to the wound, the movements of PRKCZ-

expressing HEY cells were disorganized and appeared to travel in various directions

rather than continuing in a perpendicular direction, as observed through cell migration

videos (Figure 4-10). This phenotype was specific to the HEY cell line as PRKCZ-

expressing SKOV3 and OVCAR3 cells did not exhibit differences in rate or direction of

migration.

Page 146: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

130

Figure 4-8. Migration of ovarian cancer cells. Migration rate of a) HEY, b) SKOV3, and c) OVCAR3 cell lines as measured by scratch wound assays. No difference in migration was observed between empty vector control and PRKCZ-expressing cells in any of the ovarian cancer cell lines. PC = parental control; N2 = empty vector control N2 a) b)

0

0.1

0.2

0.3

0.4

0.5

0.6

N2 PRKCZ#1 PRKCZ#2 PRKCZ#3

Dist

ance

Tra

velle

d (m

m)

0.00

0.05

0.10

0.15

0.20

0.25

0.30

0.35

N2 PRKCZ#1 PRKCZ#4 PRKCZ#8

Mig

ratio

n di

stanc

e (m

m)

c)

0

0.01

0.02

0.03

0.04

0.05

0.06

0.07

0.08

N2 PRKCZ#1 PRKCZ#2 PRKCZ#3

Dis

tanc

e Tr

avel

led

(um

)

Page 147: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

131

Figure 4-9. Effect of PRKCZ gene knockdown on SKOV3 parental cells migration as observed by wound healing assay. a) Confirmation of PRKCZ gene expression knockdown by quantitative real-time PCR. B) Knockdown of PRKCZ expression by siRNA inhibits migration compared to controls as observed in wound healing assays. (*p<0.01) (n=2) a)

0.0

0.2

0.4

0.6

0.8

1.0

1.2

Rel

ativ

e E

xpre

ssio

n

PRKCZ mRNA Expression

Page 148: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

132

b)

PRKCZ siRNA Mock Control

0 hr 0 hr

20 hr 20 hr

0

0.05

0.1

0.15

0.2

0.25

0.3

0.35

PRKCZ siRNA Scramble siRNA Mock siRNA

Dis

tanc

e tr

avel

led

(mm

) *

0

0.05

0.1

0.15

0.2

0.25

0.3

0.35

PRKCZ siRNA Scramble siRNA Mock siRNA

Dis

tanc

e tr

avel

led

(mm

) *

Page 149: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

133

Figure 4-10. Disorganized cell movement of HEY cells over-expressing PRKCZ. Despite similar migration rates between HEY control and PRKCZ-tranfected cells in scratch wound assays, the movement of PRKCZ-expressing cells appeared to be disorganized compared to parental cells (as observed by time-lapse migration video). N2 = empty vector control N2. HEY-N2 HEY-PRKCZ 0 hr 4 hr 8 hr 12 hr

Page 150: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

134

Furthermore, to investigate if polarity plays a role in directionality of HEY cells, I

performed polarity assays by measuring the orientation of microtubule organization

centre (MTOC) of the leading edge of cells subjected to scratch wound. However, no

differences were observed between PRKCZ-expressing and control HEY cells (Figure 4-

11).

To examine the effect of PRKCZ on single-cell motility of ovarian cancer cells, I

performed phagokinetic track assays using HEY, SKOV3, and OVCAR3 cell lines

(Figure 4-12). As expected, the areas travelled by each cell line correlate with their

invasive properties, with HEY cells having the highest motility track area, followed by

SKOV3, and OVCAR3 cells being the least motile. Despite the random cell movement

observed in scratch wound healing assay, HEY cells that over-express PRKCZ did not

show an increased level of single-cell motility compared to vector control cells. However,

it is possible that PRKCZ-expressing HEY cells have migrated to areas that they have

previously travelled, thus giving lower measurements of track areas. Interestingly,

phagokinetic track assays showed that SKOV3 cells over-expressing PRKCZ decreased

cell motility compared to the parental cells; however, this decrease in cell motility did not

affect the overall migration rate of these cells. PRKCZ-expressing OVCAR3 cells did

not show a difference in motility compared to control cells.

Page 151: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

135

Figure 4-11. Measurement of cell polarity of HEY by pericentrin orientation assay. Monolayer of HEY cells were subjected to scratch wound and fixed at 4 hours post wounding. a) Co-labelling of wounded HEY cells for tubulin (green), pericentrin (red), and nucleus (blue). White arrows depict direction of cell movement. White dotted lines indicate quadrant of cell facing wound. b) The percentage of wound-edge cells with MTOC orientation facing wound. a)

Scratch wound

b)

0

10

20

30

40

50

60

70

80

90

100

N2 PRKCZ#1 PRKCZ#3

% o

f MT

OC

Fac

ing

the

Wou

nd

Page 152: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

136

Figure 4-12. Quantitation of ovarian cancer cell motility. Phagokinetic track assays were performed to quantitate the effect of PRKCZ on ovarian cancer cell motility. a) Comparison of cell motility between the three parental ovarian cancer cell lines. HEY exhibited the highest level of motility, followed by SKOV3, while OVCAR3 displayed minimal movement. b) & c) No effect was observed between controls and PRKCZ-expressing HEY and OVCAR3 cells. d) Decreased motility was observed in SKOV3 cells (p<0.05). (n=2) N2 = empty vector control N2 a) b)

0

2000

4000

6000

8000

10000

12000

14000

16000

HEY SKOV3 OVCAR3

Trac

k ar

ea/c

ell (

um2 )

0

5000

10000

15000

20000

25000

30000

HEY N2 HEY PRKCZ#1 HEY PRKCZ#2 HEY PRKCZ#3

Trac

k ar

ea/c

ell (

um2 )

c) d)

0500

100015002000250030003500400045005000

SKOV3 N2 SKOV3PRKCZ#1

SKOV3PRKCZ#2

SKOV3PRKCZ#4

SKOV3PRKCZ#8

Trac

k ar

ea/c

ell (

um2 )

0

50

100

150

200

250

300

350

400

450

500

OVCAR3 N2 OVCAR3 PRKCZ#1 OVCAR3 PRKCZ#2 OVCAR3 PRKCZ#3

Trac

k ar

ea/c

ell (

um2 )

Page 153: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

137

4.3.4 Identification of Potential Downstream Effectors of PRKCZ

PRKCZ is involved in various cell signalling pathways, thus may alter multiple

downstream targets. To identify downstream effectors of PRKCZ that may be involved in

ovarian tumourigenesis, I first performed molecular network analyses with the

microarray data to identify potential interactors of PRKCZ (Figure 4-13). Upon

examination of the molecular network as identified by IPA, I decided to focus on IGF1R

(insulin-like growth factor 1 receptor) and ITGB3 (integrin beta 3) in my subsequent

analyses.

4.3.4.1 IGF1R and ITGB3 as Potential Downstream Effectors of PRKCZ

The first step I took was to examine if there is a relationship between PRKCZ,

IGF1R and ITGB3 was to determine if over-expression PRKCZ could exert any effect on

the mRNA and protein expression of IGF-IR in the three different ovarian cancer cell

lines by quantitative real-time PCR and western blot analyses (Figure 4-14). No

differences in transcript or protein expression were observed in PRKCZ-expressing HEY

cells when compared to parental and empty-vector control cells. However, while the

transcript level was not altered in SKOV3 cells, an increase in PRKCZ protein expression

correlated with an increased level of IGF1R protein, suggesting that PRKCZ may

participate in IGF1R translation or protein stability. Additionally, the expression level of

phosphorylated IRS-2 (insulin-receptor substrate-2), a known downstream target for IGF-

IR, was also increased in SKOV3 cells, confirming that PRKCZ is involved in the

activation of IGF1R signalling pathway in this particular cell line.

Page 154: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

138

Figure 4-13. Identification of potential interactors of PRKCZ. Ingenuity pathway analysis was performed to identify interactors of PRKCZ using microarray results from Chapter 1. Integrin beta 3 (ITGB3) and Insulin Growth Factor 1 Receptor (IGF1R) were identified as indirect interactors of PRKCZ and were chosen for further analyses.

Page 155: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

139

Figure 4-14. Transcript and protein expression of IGF1R in PRKCZ-expressing SKOV3 cells. a) No significant increase in IGF1R transcript level was observed in SKOV3. b) Protein level of IGF1R is elevated in PRKCZ-expressing cells. c) The level of p-IRS-2, a downstream effector of IGF1R, was elevated in 3 of the 4 PRKCZ clones. PC = parental control; N2 = empty vector control N2. Western blot shown is representative of 3 independent analyses. a)

0.0

0.5

1.0

1.5

2.0

2.5

N2 PRKCZ#1 PRKCZ#2 PRKCZ#4 PRKCZ#8

Rela

tive

Expr

essio

n

b)

PRKCZ clones PC N2 #1 #2 #4 #8

PRKCZ

IGF1R

Beta-actin c)

p-IRS2

Beta-actin

Page 156: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

140

Interestingly, in contrast to SKOV3 cells, expression of IGF1R was decreased in

OVCAR3 cells that over-express PRKCZ, at both the transcript and protein levels (Figure

4-15).

The transcript and protein expression of ITGB3 was also determined in the three

ovarian cancer cell lines by real-time quantitative RT-PCR and western blot analyses.

The expression of ITGB3 was unaltered in HEY cells that over-express PRKCZ, at both

transcriptional and protein levels. Interestingly, mRNA and protein expression of ITGB3

were significantly decreased in both SKOV3 cells and OVCAR3 cells that over-express

PRKCZ, compared to parental and empty-vector controls (Figures 4-16, 4-17).

The concurrent expression alterations observed for IGF1R and ITGB3 in PRKCZ-

expressing cells prompted the question of whether these changes are occurring within the

same biological pathway. I first performed IGF1R siRNA gene knockdown experiments

in SKOV3 cells to determine if reducing IGF1R expression would have an effect on

ITGB3 gene expression. Results from quantitative real-time PCR analysis indicated that

IGF1R knockdown can lead to de-repression of ITGB3 mRNA expression in cells over-

expressing PRKCZ (Figure 4-18). To further address whether activation of the IGF1-

signalling pathway plays a role in ITGB3 gene regulation, I examined the expression

level of ITGB3 after stimulation of SKOV3 cells with IGF1, a known ligand for IGF1R.

Interestingly, similar to the IGF1R siRNA knockdown experiment, ITGB3 expression

was de-repressed in PRKCZ-expressing cells when stimulated with IGF1 (Figure 4-19).

Page 157: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

141

Figure 4-15. Transcript and protein expression of IGF1R in PRKCZ-expressing OVCAR3 cells. Both transcript and protein levels of IGF1R are decreased in PRKCZ-expressing OVCAR3 cells. This observation is in contrast to SKOV3 cells, which showed increased IGF1R expression at protein levels. PC = parental control; N2 = empty vector control N2. Western blot shown is representative of 3 independent analyses.

0

0.2

0.4

0.6

0.8

1

1.2

N2 PRKCZ#1 PRKCZ#2 PRKCZ#3

Rela

tive

Expr

essio

n

PC N2 #1 #2 #3 PRKCZ clones

PRKCZ IGF1R

Beta-actin

Page 158: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

142

Figure 4-16. Transcript and protein expression of ITGB3 in PRKCZ-expressing SKOV3 cells. Both transcript and protein levels of ITGB3 are decreased in PRKCZ-expressing SKOV3 cells. PC = parental control; N2 = empty vector control N2. Western blot shown is representative of 3 independent analyses.

0

0.2

0.4

0.6

0.8

1

1.2

N2 PRKCZ#1

PRKCZ#2

PRKCZ#4

PRKCZ#8

Rela

tive

Expr

essio

n

p < 0.05

* *

* *

PRKCZ clones

PC N2 #1 #2 #4 #8

PRKCZ

ITGB3

Beta-actin

Page 159: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

143

Figure 4-17. Transcript and protein expression of ITGB3 in PRKCZ-expressing OVCAR3 cells. Similar to SKOV3 cell line, both transcript and protein levels of ITGB3 in OVCAR3 cell line are decreased in PRKCZ-expressing cells. PC = parental control; N2 = empty vector control N2. Western blot shown is representative of 3 independent analyses.

0

0.2

0.4

0.6

0.8

1

1.2

N2 PRKCZ#1 PRKCZ#2 PRKCZ#3

Rela

tive

Expr

essio

n

p < 0.05

*

*

*

PRKCZ clones

PC N2 #1 #2 #3

PRKCZ

ITGB3

Beta-actin

Page 160: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

144

Figure 4-18. Knockdown of IGF1R rescues transcript expression of ITGB3 in PRKCZ-expressing cells. The expression of ITGB3 is de-repressed in PRKCZ-expressing SKOV3 cells subjected to IGF1R gene knockdown by siRNA (*p < 0.05), but IGF1R knockdown has no effect on empty vector control. (n=3) N2 = empty vector control N2.

0

0.2

0.4

0.6

0.8

1

1.2

1.4

N2 PRKCZ#1 PRKCZ#4

Rela

tive

Expr

essio

n

Mockscrambled siRNA IGF1R siRNA

*

*

Page 161: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

145

Figure 4-19. IGF1 increases ITGB3 transcript expression in PRKCZ-expressing SKOV3 cells. To examine if transcript expression of ITGB3 is directly regulated by IGF1-signalling, SKOV3 cells were treated with IGF1 (50 ng/ml) and its expression was measured by quantitative real-time RT-PCR. The addition of IGF1 had no effect on empty vector control but significantly increased the expression of ITGB3 in three of the four PRKCZ clones tested when compared to their non-treated counterpart (*p < 0.05). N2 = empty vector control N2.

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

N2 PRKCZ#1 PRKCZ#2 PRKCZ#4 PRKCZ#8

Rel

ativ

e Ex

pres

sion

No treatmentIGF-I (50 ng/ml)

*

**

Page 162: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

146

Moreover, to examine if there is a negative feedback mechanism that regulates

expression of IGF1R, I induced SKOV3 cells with IGF1 and examined their IGF1R

mRNA expression (Figure 4-20). Upon stimulation with IGF1, the transcript expression

of IGF1R decreased in all SKOV3 cells, except for PRKCZ clone #4, as observed by

quantitative RT-PCR. This illustrates that IGF1 stimulation in SKOV3 cells may in fact

be able to repress the transcript expression of IGF1R.

4.3.4.2 TIMP-1 as a Potential Downstream Effector in ITGB3 and IGF1 Signalling

Based on my observations that both ITGB3 mRNA and protein levels are

decreased in PRKCZ-expressing cells in two different ovarian cancer cell lines (SKOV3

and OVCAR3), I sought to identify potential downstream players within this signalling

pathway that may play role in ovarian cancer. Transcription of TIMP-1 (TIMP

metallopeptidase inhibitor 1) has previously been shown to be up-regulated by ITGB3 in

the ovarian cancer cell line MDAH 2774 and thus may be a candidate target (231).

Therefore, I decided to examine the mRNA level of TIMP-1 in SKOV3 and OVCAR3

cells. Interestingly, TIMP-1 expression was decreased in PRKCZ clones of both of these

cell lines, which correlated with the expression of ITGB3 (Figure 4-21). To further

examine if the decrease in TIMP-1 expression was directly related to the decreased level

of ITGB3, SKOV3 parental cells were subjected to ITGB3 knockdown. No difference in

TIMP-1 expression was observed between these cells (Figure 4-22), suggesting that the

decreased level of TIMP-1 in PRKCZ-expressing cells was ITGB3-independent.

Page 163: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

147

Figure 4-20. IGF1 stimulation decreases IGF1R gene expression in SKOV3 cells. When SKOV3 cells are induced with IGF1, the transcript level of IGF1R is significantly decreased (*p<0.05), except for one of the PRKCZ clones (PRKCZ#4).

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

PC EV PRKCZ#1 PRKCZ#4 PRKCZ#8

Rela

tive

Expr

essio

n

no IGF1IGF1 (50 ng/ml)

*

*

* *

Page 164: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

148

Figure 4-21. TIMP-1 gene expression decreases in PRKCZ-expressing ovarian cancer cells. The mRNA expression of TIMP-1, a potential downstream target of ITGB3, is decreased in PRKCZ-expressing SKOV3 cells and OVCAR3 cells, as determined by quantitative real-time RT-PCR (p < 0.05). N2 = empty vector control N2. (n=3)

0.0

0.2

0.4

0.6

0.8

1.0

1.2

N2 PRKCZ#1 PRKCZ#2 PRKCZ#4 PRKCZ#8

Rel

ativ

e Ex

pres

sion p < 0.05

SKOV3

0.0

0.2

0.4

0.6

0.8

1.0

1.2

N2 PRKCZ#1 PRKCZ#2 PRKCZ#3

Rel

ativ

e Ex

pres

sion

p < 0.05

OVCAR3

Page 165: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

149

Figure 4-22. TIMP-1 gene expression regulation is independent of ITGB3 gene expression in SKOV3 cells. The transcript levels of TIMP-1, a potential downstream target of ITGB3 was determined by quantitative real-time RT-PCR following ITGB3 siRNA knockdown in SKOV3 parental cells. a) Confirmation of ITGB3 expression knockdown. b) Transcript levels of TIMP1 in SKOV3 cells with and without ITGB3 siRNA treatment. No significant TIMP1 transcript level difference was observed between ITGB3 siRNA treated and control cells. (n=3) a)

0

0.2

0.4

0.6

0.8

1

1.2

Mock Scrambled siRNA ITGB3 siRNA

Rel

ativ

e m

RN

A E

xpre

ssio

n

ITGB3 mRNA Expression

b)

0

0.2

0.4

0.6

0.8

1

1.2

Mock siRNA Scramble siRNA ITGB3 siRNA

Rel

ativ

e Ex

pres

sion

TIMP-1 mRNA expression

0

0.2

0.4

0.6

0.8

1

1.2

Mock siRNA Scramble siRNA ITGB3 siRNA

Rel

ativ

e Ex

pres

sion

TIMP-1 mRNA expression

Page 166: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

150

In addition to ITGB3, I examined if induction of IGF1 signalling has an effect on TIMP-1

expression. I observed that upon IGF1 stimulation, SKOV3 parental and empty-vector

controls exhibited a 2-fold decrease in TIMP-1 mRNA, but no further decrease in TIMP-1

expression was observed in PRKCZ clones (Figure 4-23). This is an interesting finding,

as IGF1 and PRKCZ signalling pathways may converge through their potential roles in

the regulation of TIMP-1 expression.

4.3.4.3 Effects of IGF and ITGB3 Signalling on Cell Migration/Invasion in SKOV3

Cells

The lack of migratory changes in PRKCZ-expressing SKOV3 cells as observed

from my migration experiments as described above may be due to lack of stimulation.

Since an increase in IGF-IR expression was observed in cells over-expressing PRKCZ, I

repeated the migration assays with the addition of IGF1. As seen in Figure 4-24, an

increase in migration was observed in parental and empty-vector controls upon

stimulation with IGF1 in the wound healing assay, illustrating that the IGF1 signalling

pathway is involved in migration of SKOV3 cells. However, this effect was not observed

in PRKCZ-expressing cells. The lack of response in PRKCZ-expressing cells may

perhaps be due to negative feedback exerted by the over-expression of IGF1 receptor in

these cells. Matrigel migration assays also did not show an increased level of invasion in

any of cells upon stimulation of IGF1 (Figure 4-25). However, upon stimulation with

EGF, both control and PRCKZ clones exhibited increased migration, and the effect of

EGF was significantly higher in two of the PRKCZ clones when compared to vector

control cells.

Page 167: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

151

Lastly, to evaluate if ITGB3 plays a role in ovarian cancer migration, scratch

wound healing assays were repeated with SKOV3 cells treated with ITGB3 siRNA. No

difference in migration rate was observed between siRNA-treated and control cells

(Figure 4-26).

Page 168: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

152

Figure 4-23. Effect of IGF1 on TIMP-1 transcript expression in SKOV3 cells. Treatment with IGF1 (50 ng/ml) decreased TIMP-1 transcrpt levels in SKOV3 parental and empty-vector control cells but not PRKCZ clones (* p < 0.05). (n=2) PC = parental control; N2 = empty vector control N2

0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

PC N2 PRKCZ#1 PRKCZ#2 PRKCZ#4 PRKCZ#8

Rel

ativ

e Ex

pres

sion

No Treatment

IGF-I (50 ng/ml)

* *

Page 169: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

153

Figure 4-24. Effect of IGF1 signalling on SKOV3 migration as observed by wound healing assay. No significant increase in migration was observed between controls and PRKCZ-expressing SKOV3 cells in the absence of stimulation (black bars). However, upon treatment with IGF1, parental and vector-control cells showed an increase in migration (white bars; * p<0.05). This observation was not seen in PRKCZ-expressing clones. (n=2) PC = parental control; N2 = empty vector control N2

0.00

0.05

0.10

0.15

0.20

0.25

0.30

0.35

0.40

PC N2 PRKCZ#1 PRKCZ#4 PRKCZ#8

Mig

ratio

n D

istan

ce (m

m)

No treatment

IGF1 (50 ng/ml) * *

Page 170: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

154

Figure 4-25. Effects of IGF1 and EGF on migration of SKOV3 as determined by transwell migration assay. No difference in migration was observed between SKOV3 vector control and PRKCZ-expressing cells in non-treated and IGF1-treated cells. When treated with EGF, both the control and PRCKZ clones exhibited an increased in migration (*p<0.05); the effect of EGF was significantly higher in two of the PRKCZ clones when compared to vector control cells (**p<0.05). (n=2) N2 = empty vector control N2

0100020003000

4000500060007000

N2 PRKCZ#1 PRKCZ#4 PRKCZ#8

# of

cel

ls m

igra

ted

EGF (10ng/ml)IGF-1 (50 ng/ml) + 10% FBS10% FBS

*

* *

** **

*

Page 171: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

155

Figure 4-26. Effect of ITGB3 on SKOV3 parental cells migration as observed by wound healing assay. Knockdown of ITGB3 has no effect on cell migration in SKOV3 cells. a) Confirmation of ITGB3 knockdown in SKOV3 cells. b) Distance travelled by SKOV3 cells with and without treatment of ITGB3 siRNA. (n=3) a)

ITGB3 mRNA Expression

0

0.2

0.4

0.6

0.8

1

1.2

Mock Scramble siRNA ITGB3 siRNA

Rel

ativ

e Ex

pres

sion

b)

Migration of SKOV3 Cells

0

0.05

0.1

0.15

0.2

0.25

0.3

0.35

Mock Scramble siRNA ITGB3 siRNA

Dis

tanc

e T

rave

lled

(mm

)

Page 172: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

156

4.4 Discussion

As discussed in Chapter 2, my microarray study of familial ovarian cancer

comparing the gene expression profiles of tumours from patients with strong and weak

family history of breast and/or ovarian cancer has identified genes that are differentially

expressed. One of the most significant genes identified was PRKCZ, which was more

highly expressed in tumours from patients with strong family history, and this differential

expression was validated with quantitative real-time PCR. This observation, as well as

the previously suggested role of PRKCZ in cancer development, provided me with the

rationale for further investigations of PRKCZ.

As defined by Hanahan and Weinberg, human tumourigenesis is a multistep

process in which cells can acquire properties, through genetic alterations, that allow them

to transform into a higher malignant derivative (3). In this Chapter, I aimed to determine

if alteration in PRKCZ expression can drive such processes, by manipulating the

expression levels of PRKCZ in ovarian cancer cell lines, and examining their effect on

cell survival and migration of these cells.

My cell viability assays data revealed that over-expression of PRKCZ increases

survival of SKOV3 ovarian cancer cells while no changes were observed for HEY or

OVCAR3 cells. The variable results are likely due to the different genetic attributes

associated with each of the cell lines, which might have led to differential response to

PRKCZ-signalling. Subsequent apoptotic and proliferation experiments on SKOV3 cells

suggested that the increase in cell viability observed for PRKCZ-expressing cells was due

to an increase in proliferation and not due to suppression of apoptosis.

Page 173: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

157

The role of PRKCZ in apoptosis remains controversial as it appears to act as both

a positive and a negative regulator (232-235). Interestingly, while I did not observe an

apoptotic effect of PRKCZ in SKOV3 cells, a recently published study by Nazarenko et

al. reported PRKCZ as a negative regulator of cell survival in ovarian cancer, as its over-

expression was able to mediate HRSL3 tumour suppressor-dependent apoptosis in

ovarian cancer cell line OVCAR3 (234). While I did not further examine the apoptotic

role of PRKCZ in OVCAR3 cells specifically (since no change in cell viability was

initially observed for this cell line), the observation made by Nazarenko et al.

nevertheless suggests that PRKCZ may be a critical player in ovarian cancer cell

apoptosis, and that its role may be dependent on additional molecular players. On the

other hand, through immunohistochemical analyses, the same group found that a

significant proportion of ovarian carcinomas express high levels of PRKCZ, as compared

to normal and benign carcinomas, and its expression was positively correlated with poor

prognosis (234). This observation appears to be contradictory to their in vitro results

described above, as one may expect a decreased expression of pro-apoptotic proteins in

advanced cancers; however, the authors also suggested that while PRKCZ expression

remains high in these tumours, the pro-apoptotic activity of PRKCZ may be reduced by

other genetic or epigenetic changes that occur in advanced cases of ovarian cancer (234).

Thus, the exact role of PRKCZ in apoptosis remains elusive and requires further

investigations.

The ability of cancer cells to migrate during cancer progression is associated with

the acquisition of abnormal motile behaviour resulting from various molecular

alterations. Aberrant expression of PRKCZ is an example of such alteration, as its role in

Page 174: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

158

migration has previously been demonstrated in head and neck tumour cells, pancreatic

cancer, as well as breast cancer (177, 236, 237). The comparisons I made between

parental cell lines with their PRKCZ-expressing counterparts through various migration

experiments showed that over-expressing PRKCZ alone is not sufficient to exert

increased migratory properties in the three ovarian cancer cell lines tested. Nevertheless,

I showed that siRNA knockdown of PRKCZ expression in SKOV3 parental cells can

decrease its rate of migration. This observation suggests that the endogenous level of

PRKCZ is sufficient for cell motility in this cell line. Additionally, in concordance with

previous studies examining the migratory role of PRKCZ in different types of cancer cell

lines, I showed that PRKCZ can increase cell motility in SKOV3 cells to a substantial

level compared to control cells when the EGF signalling pathway is activated, thus

demonstrating that PRKCZ signalling can augment EGF-induced chemotaxis in multiple

cancer types.

In addition to PRKCZ, it is also important to note the potential roles of PRKCI

(protein kinase C iota) in ovarian cancer. Similar to PRKCZ, PRKCI belongs to the

atypical protein kinase C family group, and it has been implicated in the establishment of

cell polarity, motility, proliferation, and survival of cancer cells (238-241). Interestingly,

the PRKCI gene has been shown to be amplified and over-expressed in serous epithelial

ovarian cancers, and an increase in its DNA copy number is associated with a decrease in

progression-free survival for the disease (239, 242). Since PRKCZ and PRKCI are

highly homologous to one another, sharing ~70% overall amino acid sequence identity

(243), it is possible that these two proteins function redundantly. Indeed, it has

previously been demonstrated that disruption of either PRKCZ or PRKCI expression can

Page 175: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

159

inhibit tight junction formation in cultured epithelial cells, suggesting that these two

proteins have an overlapping role in establishment of cell polarity (244). For that reason,

it may be important to further investigate if PRKCI can also contribute to the phenotype

that I have observed in PRKCZ-expressing cells. Additionally, since siRNA knockdown

of PRKCZ did not have an effect in HEY and OVCAR3 cell lines, it may be of interest to

determine if these cell lines are more dependent on the activity of PRKCI.

The original aim of my study was to identify the genetic alterations that occur in

the subset of patients with strong family history; therefore, I was interested in finding the

connection between PRKCZ and familial ovarian cancer. While BRCA1 and BRCA2 are

two well-studied high-penetrance genes associated with ovarian cancer, I was especially

interested in identifying expression alterations of other genes potentially related to

familial ovarian cancer which may be under the regulatory control of PRKCZ. By

examining protein and genetic interaction networks generated using my ovarian cancer

microarray expression data described in Chapter 1, I identified IGF1R (Insulin-like

Growth Factor 1 Receptor) and ITGB3 (Integrin beta 3) as potential interactors or targets

of PRKCZ.

The IGF1R promoter has previously been identified as a molecular target for

BRCA1 in breast cancer cell lines (245, 246). This observation suggests that BRCA1

mutations may result in transcriptional de-repression of the IGF1R promoter, thus

increasing the level of IGF1R, and may be one of the mechanisms responsible for breast

and ovarian tumourigenesis in BRCA1 mutation carriers. Indeed, a more recent study

conducted by the same group revealed that primary breast tumour samples from BRCA1

mutation carriers have elevated levels of IGF1R protein compared to non-carriers (247),

Page 176: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

160

further confirming the association between BRCA1 and IGF1R. Moreover, a large body

of evidence has supported the importance of IGF1R expression in ovarian cancer, and

that increased expression of IGF1R is associated with aggressiveness, as well as drug

resistance of the disease (248-253).

My studies gave evidence that over-expressing PRKCZ in certain ovarian cancer

cell lines can in fact alter the expression of IGF1R, and the type of expression alteration

is cell-line dependent. Specifically, while no IGF1R transcript levels alteration was

observed in SKOV3 cells, its protein expression level is increased in cells that over-

express PRKCZ, which may be explained by post-transcriptional processes such as

protein translation, post-translation modification and decrease in protein degradation;

however, the exact mechanism involved remains to be investigated. On the other hand,

both gene and protein expression levels of IGF1R are decreased in the OVCAR3 cell line

when PRKCZ is over-expressed, suggesting that regulation of IGF1R by PRKCZ in this

particular cell line may be occurring at the transcript level. These results imply that

regulation of IGF1R by PRKCZ may also be occurring within different biological

pathways and may be dependent on other molecular characteristics specific to each of the

cell lines, once again illustrating the heterogeneity of this disease, and further

investigations are required to determine the exact mechanisms responsible for the dual

effect PRKCZ has on IGF1R expression.

In addition to IGF1R, ITGB3 has also previously been suggested to be associated

with familial ovarian cancer. In a population study conducted by Jakubowska et al., it

was found that the Leu33Pro polymorphic allele of ITGB3 is associated with an increased

risk of ovarian cancer in individuals with BRCA1 mutations (123). While subsequent

Page 177: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

161

analysis by the same group revealed that this association may only be specific to the

Polish population from their original study (254), it nevertheless suggests the importance

of ITGB3 in ovarian cancer in a specific cohort of BRCA1 carriers. The exact molecular

mechanisms involved in the increased ovarian cancer risk in ITGB3 Leu33Pro carriers in

this Polish population is still unknown; however, it was suggested this particular allele

may be able to enhance the adhesive properties of tumour cells as well as activation of

MAPK pathway (255, 256), which may contribute to the malignant potential of cancer

cells.

The role of ITGB3 in ovarian cancer has also been implicated in a study in which

over-expression of ITGB3 in SKOV3ip1 cells (cell line generated from ascites developed

in nu/nu mouse by administering an intraperitoneal injection of SKOV3 cells) was found

to be associated with decreased invasion, protease expression, as well as colony

formation; these observations were consistent with their subsequent in vivo experiments,

which showed that tumours expressing ITGB3 were less aggressive compared to those

that do not express this protein (257). Moreover, upon examination of ITGB3 expression

in ovarian tissue of patients with invasive ovarian cancer, the same group found that

patients with high ITGB3 expression had a significantly better prognosis, which is

consistent with two other recent studies showing that an increased level of ITGB3 is

associated with better survival in patients with stage III serous ovarian cancer, and was

suggested to be a potential prognostic marker (183, 257, 258). Interestingly, my

assessment of ITGB3 expression in PRKCZ-expressing SKOV3 and OVCAR3 ovarian

cancer cells showed that ITGB3 is down-regulated in the presence of PRKCZ, as its gene

and protein expressions were both decreased compared to controls. Additional IHC

Page 178: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

162

studies should be performed to examine whether this correlation occurs in ovarian tumour

specimens, and whether there is a correlation between family history and ITGB3

expression.

The concomitant expression alterations of IGF1R and ITGB3 in PRKCZ-

expressing cells led me to question whether these genes are activated in the same

signalling pathway. Indeed, my IGF1R siRNA knockdown experiments using SKOV3

cells revealed that ITGB3 transcription may be dependent on expression of IGF1R;

however, the effects of IGF1R on OVCAR3 cells may differ since its expression is

decreased in PRKCZ-expressing cells. Nevertheless, results from my studies suggest one

possible mechanism by which ITGB3 expression may be altered, and as a consequence, a

more aggressive phenotype of ovarian cancer cells is developed.

Given that PRKCZ expression correlates with the expression of both IGF1R and

ITGB3, I further examined if alteration of these genes can affect the migration phenotype

of ovarian cancer cells that over-express PRKCZ. As mentioned earlier, over-expression

of PRKCZ alone did not alter the migration rate of any of the ovarian cancer cell lines but

did show significant increased migration in SKOV3 cells upon activation of EGF

signalling. Contrary to what I expected, scratch wound healing migration assays showed

that upon IGF1 stimulation, SKOV3 control cells but not PRKCZ-expressing cells,

displayed an increase in cellular motility. The lack of response in PRKCZ-expressing

cells may perhaps be due to a negative feedback mechanism exerted by the over-

expression of IGF1 receptor in these cells, thus hindering the cells’ ability to respond to

IGF1 signalling. Additionally, unlike results obtained from scratch wound assays, IGF1

stimulation had no effects on any of these cells in matrigel migration assays, suggesting

Page 179: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

163

that while IGF1 signalling may be involved in cell migration, it may be insufficient for

increasing the invasive properties of these cells, because the cells are incapable of

breaking down the matrigel matrix to cross the barrier.

Based on the findings from this Chapter, I propose the following model by which

PRKCZ may participate during tumour progression in a subset of ovarian cancer (Figure

4-27). In cells with normal expression of PRKCZ, the expressions of IGF1R and ITGB3

are in equilibrium. When PRKCZ is deregulated and over-expressed, it increases

translation or stability of IGF1R, thus enhancing IGF1 signalling, leading to a repression

of ITGB3 expression, which ultimately can lead to changes in cellular processes that can

enhance the aggressiveness of a tumour cell (eg. impaired apoptosis, increased

proliferation). When IGF1R gene expression is decreased (eg. via siRNA knockdown) in

these PRKCZ-expressing cells, de-repression of ITGB3 occurs. Additionally, when cells

are stimulated with IGF1, the overall expression of IGF1R decreases due to a negative

feedback mechanism that leads to suppression of IGF1R transcription and de-repression

of ITGB3; however, there may be another yet to be identified pathway downstream of

IGF1 signalling that can lead to de-repression of ITGB3. One possible pathway may be

PI3K/AKT, as IGF1 is a potent activator of this signalling cascade (259). Further

investigations are worthwhile to evaluate and confirm the roles of these important players

in ovarian cancer development; a better understanding of the interaction of the molecules

involved may be useful in development of therapeutics for the subset of ovarian cancer

patients who display expression alterations of these genes.

Page 180: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

164

Figure 4-27. Proposed model of ITGB3 transcriptional regulation through IGF-1 signalling in PRKCZ-expressing SKOV3 cells. a) Expression of IGF1R and ITGB3 are in equilibrium in the presence of normal level of PRKCZ. b) Over-expression of PRKCZ increases either translation or stability of IGF1R, possibly leading to constitutive activation of IGF1 signalling cascade that results in transcriptional repression of ITGB3 and increase cell survival through proliferation. c) Repression of ITGB3 in PRKCZ-expressing cells is dependent on IGF1R expression as IGF1R knockdown by siRNA de-represses its expression. a) b)

c)

Page 181: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

165

Figure 4-27 (Cont’d). d) IGF1 stimulation activates negative feedback mechanism, leading to a decrease in IGF1R transcription; ITGB3 expression is de-repressed. e) In addition to IGF1R transcription suppressor, other signalling pathway downstream of IGF1 cascade, such as PI3K/AKT, may be activated to de-repress expression of ITGB3. d) e)

Page 182: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

CHAPTER 5

Conclusions and Future Directions

166

Page 183: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

167

5.1 Summary and Implications of Thesis Findings

Epithelial ovarian cancer is one of the most lethal gynaecological malignancies.

One of the most important risk factors for this disease is family history; therefore,

identification of the molecular changes involved in familial ovarian cancer development

is fundamental in developing preventive and diagnostic measures for the group of

patients with family history of breast and/or ovarian cancers.

My thesis aimed to examine the molecular changes involved in familial ovarian

cancer. To achieve this, I utilized cDNA microarrays to identify the gene expression

profiles of ovarian tumours from patients with different family history. Results from this

analysis prompted me to further examine hCDC4 and PRKCZ in relation to ovarian

cancer. I carried out various genetic analyses to explore the mechanisms that may alter

hCDC4 expression in ovarian tumours. Additionally, to characterize the potential roles

of PRKCZ in ovarian cancer, functional and biochemical analyses were performed.

5.1.1 Expression Profiling of Familial Ovarian Cancer

Ovarian cancer is a heterogeneous disease that is attributable to a diverse group of

molecular alterations. Since the development of high-throughput microarrays,

identification of these alterations has escalated greatly; however, information regarding

the genetic changes that occur in familial cases of ovarian cancer remains minimal. To

address this, I performed gene expression profiling of ovarian tumours from patients with

family history of breast and/or ovarian cancer, as discussed in Chapter 2 of this thesis.

Statistical analysis of microarray results revealed the expression alterations that

occur in familial ovarian cancer, which included genes known to be linked to apoptosis,

Page 184: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

168

cell migration, cell adhesion, and cell cycle regulation. Differentially expressed genes

such as VTN and PDCD4 have previously been associated with ovarian cancer; however,

a majority of the genes identified have yet to be examined in relation to this disease, such

as MCM5, CYP11A1, as well as PRKCZ, which I have chosen for additional functional

analyses as described further below.

The development and progression of ovarian cancer is likely to be due to not one,

but multiple aberrant genetic events that ultimately lead to changes in important cellular

pathways. For that reason, I integrated my gene expression data onto various protein

interaction and biological pathway databases through Ingenuity Pathway analysis, in an

attempt to reveal and explore the altered biological networks/pathways that may play

roles in familial ovarian cancer. Significantly altered interaction networks were

identified, including those that revolve around MAPK, HNF4A, histone 3, HGF, as well

as beta-estradiol. While some of these molecules and pathways have previously been

implicated in ovarian cancer, their associations with the development of familial cases of

ovarian cancer require further investigations.

As mentioned, multiple genetic events are likely to be responsible for

manifestation of ovarian cancer; as such, high-throughput identification and subsequent

detailed examination of concomitant genetic alterations occurring in familial ovarian

cancer may reveal relevant biological information that can be used in the development of

early detection methods and clinical treatments of the disease in the subset of patients

with ovarian cancer predispositions.

Page 185: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

169

5.1.2 hCDC4 and Ovarian Cancer

My microarray study of ovarian cancer showed that the tumour suppressor gene

hCDC4 was expressed at lower levels in ovarian tumours from patients with strong

family history of breast and/or ovarian cancer compared to tumours from patients with

weak family history. Consequently I attempted to elucidate the mechanisms by which

ovarian cancer cells can deactivate the expression of hCDC4, as described in Chapter 3.

My mutation screening experiments showed that although hCDC4 mutations may

be important in malignancies such as colorectal and endometrial cancer (202, 203), it is

an infrequent event in ovarian cancer, as only one nucleotide change was observed from

my analysis of 28 ovarian tumours. This observation is in concordance with other

hCDC4 mutational studies conducted recently (207, 209). Our findings, however, could

not rule out other mechanisms by which hCDC4 expression can be repressed in ovarian

cancer. Epigenetics such as promoter methylation can play significant roles in gene

expression regulation. Indeed, numerous studies have reported the presence of

hypermethylation within promoters of tumour suppressor genes, including BRCA1 in

sporadic ovarian cancer (260). However, my analysis ruled out promoter methylation as

a method by which ovarian cancer cells can inactivate hCDC4, as all of the ovarian

tumours tested showed absence of methylation within the hCDC4 promoter. Loss of

heterozygosity may be another mechanism by which hCDC4 gene expression can be

reduced. I examined the LOH status of hCDC4 using DNA from four ovarian tumours

with their normal-matched tissues but no LOH was observed. However, due the small

number of normal samples available for this part of study, the results from this

Page 186: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

170

experiment remain inconclusive. Thus a larger collection of normal-tumour matched

samples will be required for robust analysis.

The protein expression of hCDC4 varies among ovarian tumours, as observed by

immunohistochemical staining of ovarian TMA. However, the significance of decreased

hCDC4 protein expression in familial cases of ovarian cancer was indeterminate, due to

the limited number of familial tumours present on the array. Therefore, further analyses

using a larger sample size may provide a better insight on the significance of hCDC4

expression in familial ovarian cancer.

5.1.3 PRKCZ and Ovarian Cancer

The expression of PRKCZ is more highly expressed in ovarian tumours from

patients with strong history of breast and/or ovarian cancer, as observed in familial

ovarian cancer gene expression profiling described in Chapter 2. While the roles of

PRKCZ have previously been discussed in various malignancies (174, 176-178, 223,

261), its relation to ovarian cancer is unclear.

In Chapter 4 of this thesis, I described the in vitro approaches I have taken to

examine the potential roles of PRKCZ in ovarian cancer. I showed that the SKOV3

ovarian cancer cell line stably expressing PRKCZ exhibited a higher rate of cell growth

compared to controls. This effect, however, was cell line specific, since no changes were

observed in HEY and OVCAR3 ovarian cancer cell lines. In addition to cell

proliferation, I showed that endogenous PRKCZ plays a role in ovarian cancer cell

migration, as endogenous PRKCZ expression knockdown by siRNA decreases the

migration rate of SKOV3 cells compared to controls. Moreover, while the HEY cell line

Page 187: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

171

stably expressing PRKCZ did not show an increased rate of cell migration in scratch

wound healing assays, it did exhibit random patterns of cell movement compared to

controls. Subsequent measurement of single cell movement by phagokinetic track

assays, however, showed that individual HEY cells over-expressing PRKCZ did not have

increased cell motility relative to non-transfected cells. These observations suggest that

PRKCZ-induced random cell motility in HEY cells likely requires the presence of

signalling cues from other cells over-expressing PRKCZ.

The ultimate aim for my thesis project is to identify molecular alterations

involved in the development of ovarian cancer in patients with predisposition. Hence, I

attempted to examine PRKCZ in relation to other proteins or pathways that have

previously been implicated in familial ovarian cancer. Upon examination of the

interaction network involving PRKCZ as generated by IPA using expression profiling

data, I decided to focus on the expression of IGF1R and ITGB3 in relation to PRKCZ,

since both of these genes have previously been suggested to be associated with BRCA1-

related breast and ovarian cancers (123, 245-247, 262).

Altered expression of IGF1R in various types of tumours have previously been

shown to occur after other molecular events, such as repression of tumour suppressor

genes, or gain of function mutations in p53 (263). My biochemical analyses revealed that

an increase in PRKCZ expression may be one of the molecular events leading to

alteration in IGF1R protein expression in ovarian cancer. Intriguingly, while over-

expression of PRKCZ increases protein expression of IGF1R in the SKOV3 ovarian

cancer cell line, it had the opposite effect in the OVCAR3 ovarian cancer cell line, as its

over-expression was correlated with decreased mRNA and protein expression of IGF1R.

Page 188: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

172

Strikingly, the expression of ITGB3 was decreased at both mRNA and protein levels in

PRKCZ-expressing SKOV3 and OVCAR3 cells, compared to controls. Additionally, the

mRNA level of TIMP-1, a target of ITGB3, was also decreased in cells that over-express

PRKCZ.

From these studies it is apparent that over-expression of PRKCZ can affect

downstream pathways in some ovarian cancer cells, which may lead to phenotypic

changes associated with increased aggressiveness of the disease. Further elucidation of

these pathways may provide insight on the biological events that occur in the

development of ovarian cancer in predisposed patients.

Page 189: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

173

5.2 Future Directions

The work presented in this thesis provided insight into some of the genetic and

molecular events that occur in familial ovarian cancer. Based on data presented in the

preceding chapters, additional studies can be proposed to further understand the biological

alterations responsible for ovarian cancer development in predisposed patients.

5.2.1 High-Throughput Analyses of Familial Ovarian Cancer

As mentioned earlier, the development of high-throughput techniques have guided

researchers in generating a plethora of scientific data. My initial expression microarray

analyses of an enriched set of familial ovarian cancer have also produced a collection of data

that can be further analyzed, thus providing additional insights on the biology of this disease.

5.2.1.1 Gene Set Analysis of Expression Microarrays Data

To complement our single-gene microarray analysis that was described in Chapter 2,

Gene Set Analysis (GSA) was performed in collaboration with biostatistician Dr.

Pinnaduwage. GSA differs from the aforementioned identification of differentially expressed

genes as it evaluates the differences in biologically relevant functional gene units rather than

as single genes between the two subject groups (264). Since this method uses the entire

collection of gene expression microarray data without pre-filtering for a short list of strongly

differentiated genes, it allows us to detect subtle coordinated gene expression changes in

specific biological pathways that may be important in familial ovarian cancer development.

In our preliminary GSA analysis, we examined a total of 4984 gene sets from various

curated databases, and identified six gene sets that are significantly differentially expressed

between the strong and weak familial ovarian cancer groups (Table 5-1). Furthermore, IPA

Page 190: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

174

was applied using these results to identify potential biological pathways and processes in

which these genes may be involved (Table 5-2). Intriguingly, five of the six significant gene

sets contain interaction networks that center on HNF4A, a nuclear transcription factor, but

each with different biological interactors (Figure 5-1). This is a remarkable finding since

HNF4A was also the main node in one of the protein interaction networks that I have

previously identified through IPA using our significant gene list from the single-gene

approach (Figure 2-4-B). The expression of HNF4A was not measured in my microarray

analysis because this gene was not present on the microarray platform that was used;

nevertheless, the observation made from the IPA analysis strongly suggests the diverse

pathways that HNF4A may be involved in during the development and/or progression of

familial ovarian cancer, through the deregulation of its target genes. To date, evidence for a

role for HNF4A in ovarian cancer is lacking; therefore, it may be worthwhile to further

examine the expression levels of HNF4A, as well as its gene targets in the subset of ovarian

cancer patients with strong family history in order to examine its relevance in the disease.

Additional functional studies involving manipulation of HNF4A expression may be useful in

identifying molecular alterations important in ovarian cancer predisposition.

Page 191: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

175

Table 5-1. Gene Set Analysis (GSA) of familial ovarian cancer expression microarray data. Gene sets identified as significantly altered based on gene expression data of strong and weak familial ovarian tumours. * = lower expression of most genes in the gene set correlates with strong familial cases; ** = higher expression of most genes in the gene set correlates with strong familial cases; ES = enrichment score. FDR = false discovery rate

Gene Sets Source ES p-value FDR Top Cellular Processes as determined by IPA

MORF_PRKDC*

(neighbourhood of PRKDC, protein kinase, DNA-activated, catalytic polypetide)

MSigDb v2.0; C4

-0.2755

<0.001

<0.001

cellular assembly & organization; cell cycle; cell morphology; DNA replication, replication & repair; cell-to-cell signaling and interaction, cellular growth and proliferation; cancer

GCM_PTPRU* (neighbourhood of PTPRU, protein tyrosine phosphatase, receptor type U)

MSigDb v2.0; C4

-0.8247

<0.001

<0.001

metabolic disease; cellular assembly & organization; lipid metabolism; cellular growth and proliferation; gene expression; DNA replication, recombination & repair

MORF_CCNF* (neighbourhood of cyclin F)

MSigDb v2.0; C4

-0.4705

0.002 0.2135

drug metabolism; small molecule biochemistry; cell death, cellular function & maintenance; cancer

GCM_MAX* (neighbourhood of MYC associated factor X)

MSigDb v2.0; C4

-0.8078

0.002 0.2135

gene expression; infection mechanism; cancer, cellular assembly and organization

Module412*

Segal Laboratory Cancer Modules

-1.2709

<0.001

<0.001

lipid metabolism; small molecule biochemistry; cellular compromise

FATTY_ACID_ DEGRADATION**

MSigDb v2.0; C2

0.8565

<0.001

<0.001

lipid metabolism; genetic disorder; molecule biochemistry, molecular transport

Page 192: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

176

Table 5-2. Top functions of networks belonging to significant gene sets as identified by Gene Set Analysis (GSA) and Ingenuity Pathway Analysis (IPA). Unique genes present from both microarray dataset and gene sets are shown in bold (focus molecules). A score of >3 was considered statistically significant (p<0.001).

Gene Set Molecules in Network Score # of Focus Molecules Top Functions

26s Proteasome, BRD8, BUB3, BUB1B, Caspase, CDC7, CDKN2C, Cyclin A, DIAPH1, E2f, HCFC1, HDAC2, Histone h3, HNRNPD, Hsp70, LBR, MCM2, MCM5, MCM6, NFkB (complex), NUP62, POLR2A, PP1-C, PRKDC, PSMA1, RPA2, SAFB, SSRP1, TFDP1, TRAP/Media, TRRAP, TYMS, XPO1, XPO6, XRCC5

54 26

Cellular Assembly and Organization, Cell Cycle, Cell Morphology

ALDH6A1, APRT, ARPC1A, ATP5H (includes EG:641434), ATP5I, ATP5O, ATP6AP1, ATP6V1D, ATP6V1F, CS, DLD, ESD, FH, GRB2, GSK3B, IDH3A, KCNMA1, MLEC, NAGA, PDHB, PGRMC2, PRPS2, PRPSAP2, PRUNE, RNPEP, RPS21, RPS12 (includes EG:6206), SEPT7, SHMT2, SLC2A4, SSBP1, SUCLG2, UPF3A, VARS, VDAC2

28 16

Energy Production, Nucleic Acid Metabolism, Small Molecule Biochemistry

Actin, Alpha-tubulin, ARHGEF18, CAP2, CCT4, CCT5, Ck2, DKC1, DNAJC11, DOCK5, FBXO45, GNB1, GPAA1, GTF2A2, IMMT, Insulin, KHDRBS1, KIF20B, OSBP, PCSK9, PDCL, PFDN4, PFDN6, PIN1, PMVK, PPP1R1A, RNA polymerase II, RUVBL1, SREBF2, SRRM1, SSB, STON1-GTF2A1L, TFIIF, VBP1, VTI1A

24 14

Carbohydrate Metabolism, Small Molecule Biochemistry, Post-Translational Modification

14-3-3(β, γ, θ, η, ζ), APOL3 (includes EG:80833), ARIH2, C22ORF9, Calmodulin-Camkk-Ca2+, Camkk, CAMKK1, CAMKK2, CDKN1A, COPS5, DDB1, DEAF1, DNMT1, EI24, EML3 (includes EG:256364), KAT5, KLC4, KRAS, LARP1, MAPK9, NR3C1, PCNA, PDIA3, SAMD4B, SHPRH, SKIV2L2, SLC1A6, USP1, USP14, USP37 (includes EG:57695), VANGL2, XPO7, YWHAD, YWHA, YWHAQ (includes EG:10971)

19 12

DNA Replication, Recombination, and Repair, Cell Cycle, Connective Tissue Development and Function

MORF_PRKDC

ANKRD17, APON, BUD31 (includes EG:8896), Ca2+, CALML4, CEACAM21, CXCL10, DISP2, DULLARD, ELF3, EPRS, GARS, GGT6, GRHL1, HNF4A, IL4, MCFD2, MRPS27, MS4A8B, NFKBIE, ODZ4 (includes EG:26011), RFC4, SLC22A18, SLC26A11, SLC35A1, SLC35A5, SLC39A1, TARS, TGFB1, THOP1, TMEM17, TMIGD1, TTC22, TXLNA (includes EG:200081), YARS

17 11

Cell-To-Cell Signaling and Interaction, Cellular Growth and Proliferation, Hematological System Development and Function

Page 193: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

177

1, 3, 4, 5-IP4, AKT1, ALG3, BPGM, C19ORF50, CCDC33, COL8A1, CYB5R2, DCTD, DDX46, DEGS1, dihydrosphingosine 1-phosphate, Foxo, FXR2, GFRA1, LNX1, MTCP1, NTRK2, Ntrk dimer, NUDT1, PABPC4, PCBD1, PLAC8, PTN, RBMX,RPIA, SHC1, SHC2, SHC3, SLC25A1, SNRPA, TRIP13, VCL, WDYHV1, ZBTB8A

15 10

Cancer, Reproductive System Disease, Cell Death

ANP32A, APP, ATP5G3, CALU, CD82,CDK5R2, ELP3 (includes EG:55140), FKBP5, FZD4, Histone h4, HOXB7, HOXC8, IKBKAP, MIR363 (includes EG:574031), MLL, MT1F, MTA2, NAE1, NFKBIA, nitrite, Notch, NUMBLPELP1, POLD2, POM121, PPIB, PPIE, Rab5, SET, SMARCA5, THAP7, TNFSF15, TP53BP2, ZBTB7A, ZNF131

6 5

Cellular Function and Maintenance, Gene Expression, Cell Morphology

ADCYAP1, Akt, APOC3, AVPR1B, C13ORF15, CDH5, Creb, CREB-NFkB, CSPG4, ELK4, ERK, ERK1/2, FABP3, FCGR2A, GCK, GHRHR, hCG, HMGA2, IFN Beta, IFNA21, IL1, Insulin, ITSN1, MVK, NDUFA1, NFkB (complex), OVGP1, PI3K, PRKCA, Ras homolog, SEC14L2, SSTR3, STAT4, TAF1, TEC

45 20

Metabolic Disease, Cellular Assembly an Organization, Lipid Metabolism

AHR, AUH, beta-estradiol, CCR9, CD40LG, CD8B, CHRM5, CUZD1, CYP1, DARS2, FEV, FMO2, FMO3, GH2, GZMC, HMGA1, HTR6, HTR5A, HTR5B, ICAM1, ICAM5, IL4, IL15, IRS1, KRT12, KRT83, NUDT1, P2RY6, PTPRU, SLC17A2, SLC30A3, SPRR2B, SPRR2G (includes EG:6706), SYT11, TNFSF8 (includes EG:944)

18 10

Cellular Growth and Proliferation, Hematological System Development and Function, Cellular Development

15-(S)-hydroperoxyeicosatetraenoic acid, AANAT, BNIP1, C11ORF82, CHRNE, COL14A1, CTSF, CYP26B1, KRT35, LRRC8C, MGST2, MMP13, norepinephrine, NQO2, PC, PDRG1, PEPD, PEX11A, PMM1, PPARG, PRKRIR, PRODH, PTGS2, RNASE4, SCO2 (includes EG:9997), SLC14A2, SMAD3, SMARCD1, SUPT4H1, testosterone, TNF, TNN, TP53, VPS72, ZNF8

18 10

Gene Expression, Cardiovascular System Development and Function, Organismal Development

GCM_PTPRU

acetic acid, AGTRAP, BLVRA, CSTA, EPGN, ERCC1, ERCC4, FHL3, FIGF, FOS, GCNT1, GHRHR, GPER, GRIN2B, HMMR, KLB, LGI1, LIN7A, MAPK1, MOS, MPP2, MSK1/2, MYH7, NMB, PTPN5, PTPN7, RLN2, RPS6KA4, spermidine, SRF-ELK1, ST8SIA1, TMSB4, TPSD1, TREM1, UCN2

8 5

DNA Replication, Recombination, and Repair, Cell Signaling, Nucleic Acid Metabolism

Page 194: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

178

5430435G22RIK, CA3, CDC7, CDK1/2, CDK2-CyclinE, CDKN1A, Cyclin A, Cyclin E, dihydrotestosterone, EGFBP2, ERK1/2, FRYL, HELB, Jun-GABP, KIAA0101, leucovorin, MAD2L1, MAK, MAPK8, ME1, MFN2, MGC29506, NFYB, PAXIP1, PEBP4, PLK4, POLA1, progesterone, SIGMAR1, SYNJ2, TFF2, TRIM27, TRIP13, WHSC2, ZNF346

27 13 Drug Metabolism, Small Molecule Biochemistry, Cell Death

ABCC6, ADH6, ADH1B, APLP1, ARL4C, ATG4C, C11ORF1, CEBPA, CHMP1B, CLPX, DHX8, ECE2, EXTL2, GIPC2, HNF4A, LARS2, LPGAT1, MMRN1, MPHOSPH6, MRPL44, PEPD, PEX3, PEX16, PHF10, RBL2, RFC5, RQCD1, SETDB1, SHFM1, SPAST, SPCS3, TMEM11, TPP2, WRNIP1, ZFP64

25 12 Small Molecule Biochemistry, Cellular Assembly and Organization, Cellular Development

MORF_CCNF

AKR1A1, ATP6V1B2, CACNG5, CAMK1, CAMK2G, CAMK2N1, CES1 (includes EG:1066), CETN3, CYCS (includes EG:13063), CYP2D12, DLG4, ERK, HOXD12, INPP5A, IQSEC2, JRK, LAP3, LOC388344, LPHN1, MIR214 (includes EG:406996), MUC5B, NAGPA, PCP4, PLEKHB1, retinoic acid, RPS12 (includes EG:20042), SMG1, SP1, SPRED2, STK10, SUCLA2, SYN1, TAF2, UBE2V2, UGT1A9 (includes EG:54600)

17 9 Cellular Function and Maintenance, Lipid Metabolism, Molecular Transport

ABT1, BANF1, C11ORF82, C14ORF106, CCAR1, CCDC21, CDK9, Cyclin T, CYP1A1, DCUN1D1, HNF4A, LARP7 (includes EG:51574), LSM14A, MIR24-1 (includes EG:407012), PRPF40A, RNF138, RTP3, SH3BGRL2, SMAD2, SMAD4, SMG1, STAG2, TCERG1, TM9SF2, TMEM30A, TNF, TP53, TPRKB, TRIM15, USPL1, ZBTB11, ZCCHC8, ZNF175, ZNF318 (includes EG:24149), ZNHIT6

32 13 Gene Expression, Infection Mechanism, Embryonic Development

GCM_MAX

AMD1, BCL2A1, CAPZB, CBX3, CCNA2, CDH2, CLIC4, DHX15, DSTYK, EEF1G, KPNA4, KPNB1, MAT2A, MIR199A1, MIR199A2, MT1F, MTPN, MYC, MYCBP2, MYH7, MYO1C, NCAM1, NFYC, NPPB, RAB10, RBM25 (includes EG:58517), RBMS1, ROCK1, S100A6, SDC1, SERBP1, SPARC, TSC2, XRN1, ZNF217

5 3 Cancer, Renal and Urological Disease, Cellular Assembly and Organization

Page 195: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

179

Module412 ABCA6, ABHD6, AEBP1, AGT, AQP8, ASAH1, ATP6V1C1, ATP6V1D, ATP6V1H, cholesterol, COX7A2, COX7C (includes EG:1350), CPA1, CPB2, CTSA, DPAGT1, ETNK2, FUCA1, GLA, GLB1, HEXA, HNF4A, hydrogen peroxide, IER5, MIPEP, NAGA, NFkB (complex), NLN, RNF19B, SC5DL, SGK2, SPCS3, TMEM123, WNT10A, ZNF71

25 9

Lipid Metabolism, Small Molecule Biochemistry, Cellular Compromise

ACAA2, ACAA1B, ACSL3, ACSL5, ACSL6, CES2 (includes EG:8824), Cpt, CPT1, CPT1A, CPT1B, CPT1C, CRAT, DDX10, GPD2, HADHB, HNF4A, IGF1, LIF, LIPT1, LPL, LRRC8C, oleic acid, PNLIPRP1, PPAR ligand-PPAR-Retinoic acid-RXRα, PPAR ligand-PPARγ-Retinoic acid-RXR, PPARG, PPARGC1A, PPARγ ligand-PPARγ-Retinoic acid-RARα, retinoic acid, SLC25A20, SLC5A3, TMEM49, TMEM176A, TPI1, ZNF133

29 11

Lipid Metabolism, Small Molecule Biochemistry, Molecular Transport FATTY_ACID_

DEGRADATION

9330129D05RIK, acad, ACAD8, ACAD9, ACAD10, ACAD11, ACADL, ACADM, ACADS, ACADSB, ACADVL, acyl-CoA dehydrogenase, GCDH, IVD, IWS1, PPARA, TNF

7 3

Genetic Disorder, Metabolic Disease, Lipid Metabolism

Page 196: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

180

Figure 5-1. Potential significance of HNF4A in familial ovarian cancer as suggested by Gene Sets Analysis (GSA) of gene expression microarray data. Of the six gene sets identified as significantly altered from GSA analysis of familial ovarian cancer gene expression microarrays, five contain an interaction network that centralizes on HNF4A (hepatocyte nuclear factor 4, alpha), as identified by IPA. HNF4A interaction networks from A) MORF_PRKDC gene set, B) MORF_CCNF gene set, C) GCM_MAX gene set, D) Cancer Module 412 gene set, and E) FATTY_ACID_DEGRADATION gene set. A) B)

Page 197: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

181

C) D) E)

Page 198: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

182

5.2.1.2 Genomic Signature of Familial Ovarian Cancer As detailed in Chapter 1, DNA copy number alterations can have effects on gene

expressions in a cell. As such, in parallel to the cDNA expression microarray experiments, I

have initially performed array-comparative genomic hybridization (aCGH) experiments using

genomic DNA from the same cohort of tumour samples in order to identify DNA copy

number changes that occur in familial ovarian cancer. In collaborations with biostatisticians

Dr. Shelley Bull and Sarah Colby, chromosomal regions of alterations were identified in our

preliminary analysis, which included gains in 3q26.3 (where well-known ovarian cancer

oncogene PIK3CA resides), 7p14, 10q21, 15q23, 16q24, 17q, and 20q, and one deletion at

22q. These gains and loss were identified in both strong and weak familial ovarian tumours.

While I subsequently decided to focus on examining the gene expression data, the results

generated from my aCGH study can nonetheless be used to further identify genetic

alterations that occur in ovarian cancer. Regions of gains and losses should be validated by

quantitative real-time RT-PCR and fluorescence in situ hybridization (FISH), respectively,

and their correlations with mRNA and protein levels of candidate genes should be analyzed.

Appropriate functional experiments can then be designed and performed to interrogate their

roles in ovarian cancer.

5.2.2 hCDC4 and Ovarian Cancer

The mutational analysis that I performed in Chapter 3 examined the coding regions of

hCDC4 in ovarian cancer. The presence of hCDC4 variations/polymorphisms in non-coding

regions, however, has yet to be examined in ovarian cancer. Indisputably, single-nucleotide

polymorphism within intronic regions of a gene can play significant roles in cancer

susceptibility, as suggested in previous epidemiological studies (265-271). In fact, the

Page 199: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

183

presence of specific SNP alleles within intronic regions of hCDC4 was recently reported in

acute myeloid leukemia (AML) and breast cancer (272, 273). The SNP allele described in

the AML study did not show significance in genotype frequency between patients and

healthy controls (272). On the other hand, Yu et al. observed an association between specific

hCDC4 intronic SNP alleles and breast cancer susceptibility; additionally, these alleles

showed a joint effect with SNP alleles found for the cyclin E gene, thus further elucidating

the importance of cell cycle and ubiquitin ligase genes interactions in cancer development

(273). On the basis of these observations, it would be of interest to identify the presence of

intronic SNP allele variations of hCDC4 in ovarian cancer, and to explore the correlation

between different alleles with disease susceptibility.

The notion of hCDC4 as a haploinsufficient tumour suppressor gene, as previously

suggested in a study using a hCDC4+/- ; p53+/- mouse model that can develop a wide range

of malignancies, including ovarian epithelial tumours (213), also prompts the question of

whether individuals with p53 mutations may be more susceptible to ovarian cancer if they

also have one dysfunctional allele of hCDC4, either through gene mutation or LOH.

Therefore, future genetic and clinical studies examining concurrent mutational events of p53

and hCDC4 in ovarian tumours may provide some insight on some of the molecular events

responsible for ovarian tumourigenesis, and information gained from such studies may be

used for development of novel prognostic tools in clinical settings.

5.2.3 PRKCZ and Ovarian Cancer

It is also important to further explore the functions of PRKCZ in familial ovarian

cancer in relation to BRCA proteins. The absence of a well-characterized human BRCA-null

ovarian cancer cell line prevented me from examining this aspect in detail in my in vitro

Page 200: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

184

functional studies. However, a novel BRCA1-null ovarian cancer cell line, UWB1.289, and

its BRCA1-expressing counterpart have been propagated and generated by DelloRusso et al.,

which were made publicly available (274). By over-expressing PRKCZ in these cell lines, it

would now be possible to address the question of whether or not PRKCZ can exert its

tumourigenic effect more prominently in cells lacking BRCA1 functions; if so, further

characterization of the signalling pathways involved will be useful in identifying additional

potential therapeutic targets for the subset of patients with BRCA1 mutations.

In this thesis, I have demonstrated in vitro that over-expression of PRKCZ alters the

expression of IGF1R and ITGB3 in some ovarian cancer cells. Since IGF1R and ITGB3

expressions have previously been found to be associated with aggressiveness and outcome of

ovarian cancer (214, 248-250, 258), it would be relevant to determine if there is a correlation

of protein expression among these three proteins in ovarian tumour specimens by

immunohistochemical analyses. Clinical data such as family history, BRCA mutation status,

or other relevant information such as p53 and ER status of a larger cohort of familial ovarian

cancer should also be collected and used to determine the significance of concurrent protein

expression alterations in these tumours, and to relate this correlation to patient outcome.

My identification of IGF1R and ITGB3 as potential downstream effectors of

PRKCZ has shed new light on some of the mechanisms by which PRKCZ can affect

ovarian tumour progression. The regulatory and cooperative roles of these proteins in

relation to ovarian cancer should therefore be examined in additional functional studies.

The involvement of the IGF1/IGF1R axis in ovarian cancer has been

demonstrated in multiple studies (180, 248, 249, 253, 275, 276). My observation of

PRKCZ-induced IGF1R expression alterations in ovarian cancers is a novel finding and

requires additional investigations to address the exact biological mechanisms that are

Page 201: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

185

involved. PRKCZ appears to have multiple roles in the regulation of IGF1R, as shown

by its ability to both increase and decrease IGF1R expression levels in SKOV3 and

OVCAR3 cells, respectively. The lack of increased IGF1R mRNA expression in SKOV3

cells over-expressing PRKCZ suggests that increased IGF1R protein level may be due to

decreased protein degradation. To examine if direct physical interaction between

PRKCZ and IGF1R plays role in protein stability, localization and co-

immunoprecipitation studies should be conducted in these cells. Alterations in PRKCZ-

related signalling cascades (eg. Raf/Mek/MAPK pathway) may also be involved in

IGF1R stability, by ultimately inhibiting the activity of molecules involved in IGF1R

degradation. This hypothesis can be tested by using antagonists that target the different

signalling molecules, followed by IGF1R protein expression detection. Furthermore,

since over-expression of PRKCZ in OVCAR3 was shown to repress the gene expression

of IGF1R, PRKCZ-siRNA knockdown experiments should be performed to examine if

decreasing PRKCZ levels would have an opposite effect, resulting in increased IGF1R

expression, relative to non-treated parental cells, and further examine if these expression

alterations are associated with changes in proliferation and migration properties of

OVCAR3.

The two recent studies conducted by Partheen et al. have suggested ITGB3 to be a

potential prognostic marker for ovarian cancer, as low protein level of this protein in

advanced stage serous carcinomas is associated with poor patient outcome; however, the

regulatory mechanisms for ITGB3 expression have yet to be determined (183, 258).

Given that I have shown in my functional studies that over-expression of PRKCZ in

ovarian cancer cells decreases the expression of ITGB3, it may be relevant to target

Page 202: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

186

molecules in PRKCZ-associated pathways in vitro, by the use of antagonists, to elucidate

the pathway(s) of which PRKCZ can regulate ITGB3 expression. Moreover, since

normal ovarian epithelium and highly-differentiated ovarian carcinomas have been

shown to have higher expression levels of ITGB3 compared to poorly differentiated

carcinomas (277), it may be useful to examine if over-expressing PRKCZ in cell lines

derived from normal ovarian epithelium and highly-differentiated carcinomas can indeed

decrease expression of ITGB3, to result in a less-differentiated and a more aggressive

phenotype in these cells. Additionally, as mentioned in discussion section of Chapter 4,

the apoptotic role of ITGB3 has yet to be examined in ovarian cancer. Therefore, it may

also be worthwhile to examine if decreased expression of ITGB3 in ovarian cancer cells

are associated with cell survival, by correlating its expression with apoptosis markers

such as Bax and Bcl-2, either through in vitro or immunohistochemical studies.

These proposed experiments will expand our knowledge in the involvement of

PRKCZ as well as its downstream targets in ovarian tumourigenesis. Elucidation of their

expressions in a larger cohort of ovarian tumour samples, as well as their correlation with

disease outcome and family history, may therefore be helpful in the development of novel

therapeutics for ovarian cancer patients with disease predisposition.

Page 203: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Appendix

187

Page 204: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

188

Appendix A1. Family history data of ovarian cancer patients from present study. Classification of serous ovarian tumour samples according to the patients’ family history data. Nine tumours were classified as “strong familial”, and 27 tumours were classified as “weak familial”. The mean age of diagnosis for the “strong familial” and “weak familial” groups were 51 and 62 years, respectively.

Page 205: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

Bibliography

189

Page 206: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

190

1. Herceg Z, Hainaut P. Genetic and epigenetic alterations as biomarkers for cancer detection, diagnosis and prognosis. Mol Oncol 2007;1(1):26-41. 2. Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007;128(4):683-92. 3. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100(1):57-70. 4. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5):646-74. 5. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990;61(5):759-67. 6. Carethers JM. The cellular and molecular pathogenesis of colorectal cancer. Gastroenterol Clin North Am 1996;25(4):737-54. 7. Macleod K. Tumor suppressor genes. Curr Opin Genet Dev 2000;10(1):81-93. 8. Kinzler KW, Vogelstein B. Cancer-susceptibility genes. Gatekeepers and caretakers. Nature 1997;386(6627):761, 3. 9. Michor F, Iwasa Y, Nowak MA. Dynamics of cancer progression. Nat Rev Cancer 2004;4(3):197-205. 10. Knudson AG, Jr. Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A 1971;68(4):820-3. 11. Santarosa M, Ashworth A. Haploinsufficiency for tumour suppressor genes: when you don't need to go all the way. Biochim Biophys Acta 2004;1654(2):105-22. 12. Katso RM, Manek S, O'Byrne K, Playford MP, Le Meuth V, Ganesan TS. Molecular approaches to diagnosis and management of ovarian cancer. Cancer Metastasis Rev 1997;16(1-2):81-107. 13. Palii SS, Robertson KD. Epigenetic control of tumor suppression. Crit Rev Eukaryot Gene Expr 2007;17(4):295-316. 14. Richards FM. Molecular pathology of von HippelLindau disease and the VHL tumour suppressor gene. Expert Rev Mol Med 2001;2001:1-27. 15. Boente MP, Hurteau J, Rodriguez GC, Bast RC, Jr., Berchuck A. The biology of ovarian cancer. Curr Opin Oncol 1993;5(5):900-7. 16. Munger K. Disruption of oncogene/tumor suppressor networks during human carcinogenesis. Cancer Invest 2002;20(1):71-81. 17. Smith MR, Matthews NT, Jones KA, Kung HF. Biological actions of oncogenes. Pharmacol Ther 1993;58(2):211-36. 18. Rak J, Filmus J, Finkenzeller G, Grugel S, Marme D, Kerbel RS. Oncogenes as inducers of tumor angiogenesis. Cancer Metastasis Rev 1995;14(4):263-77. 19. Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer 2008;8(12):976-90. 20. Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med 2004;10(8):789-99. 21. Levitt NC, Hickson ID. Caretaker tumour suppressor genes that defend genome integrity. Trends Mol Med 2002;8(4):179-86. 22. Iwasa Y, Michor F, Komarova NL, Nowak MA. Population genetics of tumor suppressor genes. J Theor Biol 2005;233(1):15-23. 23. Weaver VM, Gilbert P. Watch thy neighbor: cancer is a communal affair. J Cell Sci 2004;117(Pt 8):1287-90. 24. Polyak K, Haviv I, Campbell IG. Co-evolution of tumor cells and their microenvironment. Trends Genet 2009;25(1):30-8.

Page 207: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

191

25. Weber F, Shen L, Fukino K, et al. Total-genome analysis of BRCA1/2-related invasive carcinomas of the breast identifies tumor stroma as potential landscaper for neoplastic initiation. Am J Hum Genet 2006;78(6):961-72. 26. Kurose K, Gilley K, Matsumoto S, Watson PH, Zhou XP, Eng C. Frequent somatic mutations in PTEN and TP53 are mutually exclusive in the stroma of breast carcinomas. Nat Genet 2002;32(3):355-7. 27. Weber F, Fukino K, Sawada T, et al. Variability in organ-specific EGFR mutational spectra in tumour epithelium and stroma may be the biological basis for differential responses to tyrosine kinase inhibitors. Br J Cancer 2005;92(10):1922-6. 28. Moloughney B, Snider J, Villeneuve L. Ovarian cancer in Canada. Cmaj 2000;162(5):690. 29. Canadian Cancer Society's Steering Committee: Canadian Cancer Statistics 2009. Toronto: Canadian Cancer Society, 2009.; 2009. 30. Ramus SJ, Gayther SA. The contribution of BRCA1 and BRCA2 to ovarian cancer. Mol Oncol 2009;3(2):138-50. 31. Werness BA, Eltabbakh GH. Familial ovarian cancer and early ovarian cancer: biologic, pathologic, and clinical features. Int J Gynecol Pathol 2001;20(1):48-63. 32. Cannistra SA. Cancer of the ovary. N Engl J Med 2004;351(24):2519-29. 33. Hennessy BT, Coleman RL, Markman M. Ovarian cancer. Lancet 2009;374(9698):1371-82. 34. Landen CN, Jr., Birrer MJ, Sood AK. Early events in the pathogenesis of epithelial ovarian cancer. J Clin Oncol 2008;26(6):995-1005. 35. Cho KR, Shih Ie M. Ovarian cancer. Annu Rev Pathol 2009;4:287-313. 36. Koulouris CR, Penson RT. Ovarian stromal and germ cell tumors. Semin Oncol 2009;36(2):126-36. 37. Lawrenson K, Gayther SA. Ovarian cancer: a clinical challenge that needs some basic answers. PLoS Med 2009;6(2):e25. 38. Shih Ie M, Kurman RJ. Ovarian tumorigenesis: a proposed model based on morphological and molecular genetic analysis. Am J Pathol 2004;164(5):1511-8. 39. Cho KR. Ovarian cancer update: lessons from morphology, molecules, and mice. Arch Pathol Lab Med 2009;133(11):1775-81. 40. Dehari R, Kurman RJ, Logani S, Shih Ie M. The development of high-grade serous carcinoma from atypical proliferative (borderline) serous tumors and low-grade micropapillary serous carcinoma: a morphologic and molecular genetic analysis. Am J Surg Pathol 2007;31(7):1007-12. 41. Barnes MN, Grizzle WE, Grubbs CJ, Partridge EE. Paradigms for primary prevention of ovarian carcinoma. CA Cancer J Clin 2002;52(4):216-25. 42. Sueblinvong T, Carney ME. Current understanding of risk factors for ovarian cancer. Curr Treat Options Oncol 2009;10(1-2):67-81. 43. Parazzini F, Franceschi S, La Vecchia C, Fasoli M. The epidemiology of ovarian cancer. Gynecol Oncol 1991;43(1):9-23. 44. Wong AS, Auersperg N. Ovarian surface epithelium: family history and early events in ovarian cancer. Reprod Biol Endocrinol 2003;1:70. 45. Russo A, Calo V, Bruno L, Rizzo S, Bazan V, Di Fede G. Hereditary ovarian cancer. Crit Rev Oncol Hematol 2009;69(1):28-44.

Page 208: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

192

46. Piver MS. Hereditary ovarian cancer. Lessons from the first twenty years of the Gilda Radner Familial Ovarian Cancer Registry. Gynecol Oncol 2002;85(1):9-17. 47. Prat J, Ribe A, Gallardo A. Hereditary ovarian cancer. Hum Pathol 2005;36(8):861-70. 48. Lynch HT, Casey MJ, Snyder CL, et al. Hereditary ovarian carcinoma: heterogeneity, molecular genetics, pathology, and management. Mol Oncol 2009;3(2):97-137. 49. Boardman LA, Thibodeau SN, Schaid DJ, et al. Increased risk for cancer in patients with the Peutz-Jeghers syndrome. Ann Intern Med 1998;128(11):896-9. 50. Gorlin RJ. Nevoid basal cell carcinoma syndrome. Dermatol Clin 1995;13(1):113-25. 51. Nelen MR, Padberg GW, Peeters EA, et al. Localization of the gene for Cowden disease to chromosome 10q22-23. Nat Genet 1996;13(1):114-6. 52. Pharoah PD, Ponder BA. The genetics of ovarian cancer. Best Pract Res Clin Obstet Gynaecol 2002;16(4):449-68. 53. Bewtra C, Watson P, Conway T, Read-Hippee C, Lynch HT. Hereditary ovarian cancer: a clinicopathological study. Int J Gynecol Pathol 1992;11(3):180-7. 54. Drake AC, Campbell H, Porteous ME, Dunlop MG. The contribution of DNA mismatch repair gene defects to the burden of gynecological cancer. Int J Gynecol Cancer 2003;13(3):262-77. 55. Peltomaki P. Role of DNA mismatch repair defects in the pathogenesis of human cancer. J Clin Oncol 2003;21(6):1174-9. 56. Wheeler JM, Bodmer WF, Mortensen NJ. DNA mismatch repair genes and colorectal cancer. Gut 2000;47(1):148-53. 57. Arzimanoglou, II, Lallas T, Osborne M, Barber H, Gilbert F. Microsatellite instability differences between familial and sporadic ovarian cancers. Carcinogenesis 1996;17(9):1799-804. 58. Christie M, Oehler MK. Molecular pathology of epithelial ovarian cancer. J Br Menopause Soc 2006;12(2):57-63. 59. Tomasini R, Mak TW, Melino G. The impact of p53 and p73 on aneuploidy and cancer. Trends Cell Biol 2008;18(5):244-52. 60. Okuda T, Otsuka J, Sekizawa A, et al. p53 mutations and overexpression affect prognosis of ovarian endometrioid cancer but not clear cell cancer. Gynecol Oncol 2003;88(3):318-25. 61. Gatcliffe TA, Monk BJ, Planutis K, Holcombe RF. Wnt signaling in ovarian tumorigenesis. Int J Gynecol Cancer 2008;18(5):954-62. 62. Liu F, Millar SE. Wnt/beta-catenin signaling in oral tissue development and disease. J Dent Res 2010;89(4):318-30. 63. Wu R, Zhai Y, Fearon ER, Cho KR. Diverse mechanisms of beta-catenin deregulation in ovarian endometrioid adenocarcinomas. Cancer Res 2001;61(22):8247-55. 64. Di Cristofano A, Ellenson LH. Endometrial carcinoma. Annu Rev Pathol 2007;2:57-85. 65. Campbell IG, Russell SE, Choong DY, et al. Mutation of the PIK3CA gene in ovarian and breast cancer. Cancer Res 2004;64(21):7678-81.

Page 209: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

193

66. Kolasa IK, Rembiszewska A, Felisiak A, et al. PIK3CA amplification associates with resistance to chemotherapy in ovarian cancer patients. Cancer Biol Ther 2009;8(1):21-6. 67. Shiue H, Musch MW, Wang Y, Chang EB, Turner JR. Akt2 phosphorylates ezrin to trigger NHE3 translocation and activation. J Biol Chem 2005;280(2):1688-95. 68. Nakayama K, Nakayama N, Kurman RJ, et al. Sequence mutations and amplification of PIK3CA and AKT2 genes in purified ovarian serous neoplasms. Cancer Biol Ther 2006;5(7):779-85. 69. Bellacosa A, de Feo D, Godwin AK, et al. Molecular alterations of the AKT2 oncogene in ovarian and breast carcinomas. Int J Cancer 1995;64(4):280-5. 70. Rychahou PG, Kang J, Gulhati P, et al. Akt2 overexpression plays a critical role in the establishment of colorectal cancer metastasis. Proc Natl Acad Sci U S A 2008;105(51):20315-20. 71. Li AJ, Karlan BY. Genetic factors in ovarian carcinoma. Curr Oncol Rep 2001;3(1):27-32. 72. Berchuck A, Carney M. Human ovarian cancer of the surface epithelium. Biochem Pharmacol 1997;54(5):541-4. 73. Gemignani ML, Schlaerth AC, Bogomolniy F, et al. Role of KRAS and BRAF gene mutations in mucinous ovarian carcinoma. Gynecol Oncol 2003;90(2):378-81. 74. Masamha CP, Benbrook DM. Cyclin D1 degradation is sufficient to induce G1 cell cycle arrest despite constitutive expression of cyclin E2 in ovarian cancer cells. Cancer Res 2009;69(16):6565-72. 75. Nam EJ, Kim YT. Alteration of cell-cycle regulation in epithelial ovarian cancer. Int J Gynecol Cancer 2008;18(6):1169-82. 76. Dhar KK, Branigan K, Parkes J, et al. Expression and subcellular localization of cyclin D1 protein in epithelial ovarian tumour cells. Br J Cancer 1999;81(7):1174-81. 77. Barbieri F, Lorenzi P, Ragni N, et al. Overexpression of cyclin D1 is associated with poor survival in epithelial ovarian cancer. Oncology 2004;66(4):310-5. 78. D'Andrilli G, Kumar C, Scambia G, Giordano A. Cell cycle genes in ovarian cancer: steps toward earlier diagnosis and novel therapies. Clin Cancer Res 2004;10(24):8132-41. 79. Fujita M, Enomoto T, Murata Y. Genetic alterations in ovarian carcinoma: with specific reference to histological subtypes. Mol Cell Endocrinol 2003;202(1-2):97-9. 80. Marone M, Scambia G, Giannitelli C, et al. Analysis of cyclin E and CDK2 in ovarian cancer: gene amplification and RNA overexpression. Int J Cancer 1998;75(1):34-9. 81. Sui L, Dong Y, Ohno M, et al. Implication of malignancy and prognosis of p27(kip1), Cyclin E, and Cdk2 expression in epithelial ovarian tumors. Gynecol Oncol 2001;83(1):56-63. 82. Meinhold-Heerlein I, Bauerschlag D, Hilpert F, et al. Molecular and prognostic distinction between serous ovarian carcinomas of varying grade and malignant potential. Oncogene 2005;24(6):1053-65. 83. Maihle NJ, Baron AT, Barrette BA, et al. EGF/ErbB receptor family in ovarian cancer. Cancer Treat Res 2002;107:247-58.

Page 210: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

194

84. Lassus H, Leminen A, Vayrynen A, et al. ERBB2 amplification is superior to protein expression status in predicting patient outcome in serous ovarian carcinoma. Gynecol Oncol 2004;92(1):31-9. 85. Mano MS, Awada A, Di Leo A, et al. Rates of topoisomerase II-alpha and HER-2 gene amplification and expression in epithelial ovarian carcinoma. Gynecol Oncol 2004;92(3):887-95. 86. McAlpine JN, Wiegand KC, Vang R, et al. HER2 overexpression and amplification is present in a subset of ovarian mucinous carcinomas and can be targeted with trastuzumab therapy. BMC Cancer 2009;9:433. 87. Tuefferd M, Couturier J, Penault-Llorca F, et al. HER2 status in ovarian carcinomas: a multicenter GINECO study of 320 patients. PLoS One 2007;2(11):e1138. 88. Vermeij J, Teugels E, Bourgain C, et al. Genomic activation of the EGFR and HER2-neu genes in a significant proportion of invasive epithelial ovarian cancers. BMC Cancer 2008;8:3. 89. Reibenwein J, Krainer M. Targeting signaling pathways in ovarian cancer. Expert Opin Ther Targets 2008;12(3):353-65. 90. Badache A, Hynes NE. A new therapeutic antibody masks ErbB2 to its partners. Cancer Cell 2004;5(4):299-301. 91. Chen GG, Zeng Q, Tse GM. Estrogen and its receptors in cancer. Med Res Rev 2008;28(6):954-74. 92. Pujol P, Rey JM, Nirde P, et al. Differential expression of estrogen receptor-alpha and -beta messenger RNAs as a potential marker of ovarian carcinogenesis. Cancer Res 1998;58(23):5367-73. 93. Issa RM, Lebeau A, Grob T, et al. Estrogen receptor gene amplification occurs rarely in ovarian cancer. Mod Pathol 2009;22(2):191-6. 94. Chan KK, Wei N, Liu SS, Xiao-Yun L, Cheung AN, Ngan HY. Estrogen receptor subtypes in ovarian cancer: a clinical correlation. Obstet Gynecol 2008;111(1):144-51. 95. Bardin A, Hoffmann P, Boulle N, et al. Involvement of estrogen receptor beta in ovarian carcinogenesis. Cancer Res 2004;64(16):5861-9. 96. Hall JM, Lee MK, Newman B, et al. Linkage of early-onset familial breast cancer to chromosome 17q21. Science 1990;250(4988):1684-9. 97. Narod SA, Feunteun J, Lynch HT, et al. Familial breast-ovarian cancer locus on chromosome 17q12-q23. Lancet 1991;338(8759):82-3. 98. Miki Y, Swensen J, Shattuck-Eidens D, et al. A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 1994;266(5182):66-71. 99. Wooster R, Neuhausen SL, Mangion J, et al. Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science 1994;265(5181):2088-90. 100. Carvalho MA, Couch FJ, Monteiro AN. Functional assays for BRCA1 and BRCA2. Int J Biochem Cell Biol 2007;39(2):298-310. 101. Narod SA, Foulkes WD. BRCA1 and BRCA2: 1994 and beyond. Nat Rev Cancer 2004;4(9):665-76. 102. Huen MS, Sy SM, Chen J. BRCA1 and its toolbox for the maintenance of genome integrity. Nat Rev Mol Cell Biol 2010;11(2):138-48. 103. Williams RS, Green R, Glover JN. Crystal structure of the BRCT repeat region from the breast cancer-associated protein BRCA1. Nat Struct Biol 2001;8(10):838-42.

Page 211: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

195

104. Gudmundsdottir K, Ashworth A. The roles of BRCA1 and BRCA2 and associated proteins in the maintenance of genomic stability. Oncogene 2006;25(43):5864-74. 105. Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet 2003;72(5):1117-30. 106. Venkitaraman AR. Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell 2002;108(2):171-82. 107. Chenevix-Trench G, Healey S, Lakhani S, et al. Genetic and histopathologic evaluation of BRCA1 and BRCA2 DNA sequence variants of unknown clinical significance. Cancer Res 2006;66(4):2019-27. 108. Meric-Bernstam F. Heterogenic loss of BRCA in breast cancer: the "two-hit" hypothesis takes a hit. Ann Surg Oncol 2007;14(9):2428-9. 109. Chan KY, Ozcelik H, Cheung AN, Ngan HY, Khoo US. Epigenetic factors controlling the BRCA1 and BRCA2 genes in sporadic ovarian cancer. Cancer Res 2002;62(14):4151-6. 110. Dworkin AM, Spearman AD, Tseng SY, Sweet K, Toland AE. Methylation not a frequent "second hit" in tumors with germline BRCA mutations. Fam Cancer 2009. 111. Mazoyer S. Genomic rearrangements in the BRCA1 and BRCA2 genes. Hum Mutat 2005;25(5):415-22. 112. Walsh T, Lee MK, Casadei S, et al. Detection of inherited mutations for breast and ovarian cancer using genomic capture and massively parallel sequencing. Proc Natl Acad Sci U S A;107(28):12629-33. 113. Sluiter MD, van Rensburg EJ. Large genomic rearrangements of the BRCA1 and BRCA2 genes: review of the literature and report of a novel BRCA1 mutation. Breast Cancer Res Treat;125(2):325-49. 114. del Valle J, Feliubadalo L, Nadal M, et al. Identification and comprehensive characterization of large genomic rearrangements in the BRCA1 and BRCA2 genes. Breast Cancer Res Treat;122(3):733-43. 115. Kasprzak L, Foulkes WD, Shelling AN. Forth nightly review: hereditary ovarian carcinoma. Bmj 1999;318(7186):786-9. 116. Gayther SA, Mangion J, Russell P, et al. Variation of risks of breast and ovarian cancer associated with different germline mutations of the BRCA2 gene. Nat Genet 1997;15(1):103-5. 117. Sowter HM, Ashworth A. BRCA1 and BRCA2 as ovarian cancer susceptibility genes. Carcinogenesis 2005;26(10):1651-6. 118. Thompson D, Easton D. Variation in cancer risks, by mutation position, in BRCA2 mutation carriers. Am J Hum Genet 2001;68(2):410-9. 119. Szabo CI, King MC. Population genetics of BRCA1 and BRCA2. Am J Hum Genet 1997;60(5):1013-20. 120. Tonin PN, Mes-Masson AM, Futreal PA, et al. Founder BRCA1 and BRCA2 mutations in French Canadian breast and ovarian cancer families. Am J Hum Genet 1998;63(5):1341-51. 121. Tonin PN, Perret C, Lambert JA, et al. Founder BRCA1 and BRCA2 mutations in early-onset French Canadian breast cancer cases unselected for family history. Int J Cancer 2001;95(3):189-93.

Page 212: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

196

122. Yarden RI, Friedman E, Metsuyanim S, Olender T, Ben-Asher E, Papa MZ. MDM2 SNP309 accelerates breast and ovarian carcinogenesis in BRCA1 and BRCA2 carriers of Jewish-Ashkenazi descent. Breast Cancer Res Treat 2008;111(3):497-504. 123. Jakubowska A, Gronwald J, Menkiszak J, et al. Integrin beta3 Leu33Pro polymorphism increases BRCA1-associated ovarian cancer risk. J Med Genet 2007;44(6):408-11. 124. Jakubowska A, Rozkrut D, Antoniou A, Hamann U, Lubinski J. The Leu33Pro polymorphism in the ITGB3 gene does not modify BRCA1/2-associated breast or ovarian cancer risks: results from a multicenter study among 15,542 BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 2010;121(3):639-49. 125. Ramus SJ, Kartsonaki C, Gayther SA, et al. Genetic Variation at 9p22.2 and Ovarian Cancer Risk for BRCA1 and BRCA2 Mutation Carriers. J Natl Cancer Inst 2010. 126. Narod SA, Risch H, Moslehi R, et al. Oral contraceptives and the risk of hereditary ovarian cancer. Hereditary Ovarian Cancer Clinical Study Group. N Engl J Med 1998;339(7):424-8. 127. Narod SA, Sun P, Ghadirian P, et al. Tubal ligation and risk of ovarian cancer in carriers of BRCA1 or BRCA2 mutations: a case-control study. Lancet 2001;357(9267):1467-70. 128. Lambros MB, Fiegler H, Jones A, et al. Analysis of ovarian cancer cell lines using array-based comparative genomic hybridization. J Pathol 2005;205(1):29-40. 129. Mayr D, Kanitz V, Anderegg B, et al. Analysis of gene amplification and prognostic markers in ovarian cancer using comparative genomic hybridization for microarrays and immunohistochemical analysis for tissue microarrays. Am J Clin Pathol 2006;126(1):101-9. 130. Leunen K, Gevaert O, Daemen A, et al. Recurrent copy number alterations in BRCA1-mutated ovarian tumors alter biological pathways. Hum Mutat 2009;30(12):1693-702. 131. Mhawech-Fauceglia P, Rai H, Nowak N, et al. The use of array-based comparative genomic hybridization (a-CGH) to distinguish metastatic from primary synchronous carcinomas of the ovary and the uterus. Histopathology 2008;53(4):490-5. 132. Osterberg L, Levan K, Partheen K, Staaf J, Sundfeldt K, Horvath G. High-resolution genomic profiling of carboplatin resistance in early-stage epithelial ovarian carcinoma. Cytogenet Genome Res 2009;125(1):8-18. 133. Osterberg L, Levan K, Partheen K, et al. Potential predictive markers of chemotherapy resistance in stage III ovarian serous carcinomas. BMC Cancer 2009;9:368. 134. Walsh CS, Ogawa S, Scoles DR, et al. Genome-wide loss of heterozygosity and uniparental disomy in BRCA1/2-associated ovarian carcinomas. Clin Cancer Res 2008;14(23):7645-51. 135. Yoshihara K, Tajima A, Adachi S, et al. Germline copy number variations in BRCA1-associated ovarian cancer patients. Genes Chromosomes Cancer. 136. Gorringe KL, Ramakrishna M, Williams LH, et al. Are there any more ovarian tumor suppressor genes? A new perspective using ultra high-resolution copy number and loss of heterozygosity analysis. Genes Chromosomes Cancer 2009;48(10):931-42.

Page 213: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

197

137. Goode EL, White KL, Vierkant RA, et al. Xenobiotic-Metabolizing gene polymorphisms and ovarian cancer risk. Mol Carcinog. 138. Engel C, Versmold B, Wappenschmidt B, et al. Association of the variants CASP8 D302H and CASP10 V410I with breast and ovarian cancer risk in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol Biomarkers Prev 2010;19(11):2859-68. 139. Monaco A, Menolascina F, Zhao Y, et al. "Sequencing-grade" screening for BRCA1 variants by oligo-arrays. J Transl Med 2008;6:64. 140. Fehrmann RS, Li XY, van der Zee AG, et al. Profiling studies in ovarian cancer: a review. Oncologist 2007;12(8):960-6. 141. Le Page C, Ouellet V, Madore J, et al. From gene profiling to diagnostic markers: IL-18 and FGF-2 complement CA125 as serum-based markers in epithelial ovarian cancer. Int J Cancer 2006;118(7):1750-8. 142. Berchuck A, Iversen ES, Luo J, et al. Microarray analysis of early stage serous ovarian cancers shows profiles predictive of favorable outcome. Clin Cancer Res 2009;15(7):2448-55. 143. McGuire WP, Ozols RF. Chemotherapy of advanced ovarian cancer. Semin Oncol 1998;25(3):340-8. 144. L'Esperance S, Popa I, Bachvarova M, et al. Gene expression profiling of paired ovarian tumors obtained prior to and following adjuvant chemotherapy: molecular signatures of chemoresistant tumors. Int J Oncol 2006;29(1):5-24. 145. Bild AH, Yao G, Chang JT, et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 2006;439(7074):353-7. 146. Sunde JS, Donninger H, Wu K, et al. Expression profiling identifies altered expression of genes that contribute to the inhibition of transforming growth factor-beta signaling in ovarian cancer. Cancer Res 2006;66(17):8404-12. 147. Crum CP, Drapkin R, Kindelberger D, Medeiros F, Miron A, Lee Y. Lessons from BRCA: the tubal fimbria emerges as an origin for pelvic serous cancer. Clin Med Res 2007;5(1):35-44. 148. Tone AA, Begley H, Sharma M, et al. Gene expression profiles of luteal phase fallopian tube epithelium from BRCA mutation carriers resemble high-grade serous carcinoma. Clin Cancer Res 2008;14(13):4067-78. 149. Motamed-Khorasani A, Jurisica I, Letarte M, et al. Differentially androgen-modulated genes in ovarian epithelial cells from BRCA mutation carriers and control patients predict ovarian cancer survival and disease progression. Oncogene 2007;26(2):198-214. 150. Evangelou A, Letarte M, Jurisica I, et al. Loss of coordinated androgen regulation in nonmalignant ovarian epithelial cells with BRCA1/2 mutations and ovarian cancer cells. Cancer Res 2003;63(10):2416-24. 151. Jiang X, Zhu T, Yang J, et al. Identification of novel epithelial ovarian cancer biomarkers by cross-laboratory microarray analysis. J Huazhong Univ Sci Technolog Med Sci;30(3):354-9. 152. Yoshihara K, Tajima A, Yahata T, et al. Gene expression profile for predicting survival in advanced-stage serous ovarian cancer across two independent datasets. PLoS One;5(3):e9615.

Page 214: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

198

153. Yamaguchi K, Mandai M, Oura T, et al. Identification of an ovarian clear cell carcinoma gene signature that reflects inherent disease biology and the carcinogenic processes. Oncogene 2010;29(12):1741-52. 154. Crijns AP, Fehrmann RS, de Jong S, et al. Survival-related profile, pathways, and transcription factors in ovarian cancer. PLoS Med 2009;6(2):e24. 155. Jazaeri AA, Yee CJ, Sotiriou C, Brantley KR, Boyd J, Liu ET. Gene expression profiles of BRCA1-linked, BRCA2-linked, and sporadic ovarian cancers. J Natl Cancer Inst 2002;94(13):990-1000. 156. Walker LC, Waddell N, Ten Haaf A, Grimmond S, Spurdle AB. Use of expression data and the CGEMS genome-wide breast cancer association study to identify genes that may modify risk in BRCA1/2 mutation carriers. Breast Cancer Res Treat 2008;112(2):229-36. 157. Yang YH, Dudoit S, Luu P, et al. Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res 2002;30(4):e15. 158. Smyth GK. Limma: linear models for microarray data. . New York: Springer; 2005. 159. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A 2001;98(9):5116-21. 160. Allison SJ, Milner J. SIRT3 is pro-apoptotic and participates in distinct basal apoptotic pathways. Cell Cycle 2007;6(21):2669-77. 161. Heyman L, Leroy-Dudal J, Fernandes J, Seyer D, Dutoit S, Carreiras F. Mesothelial vitronectin stimulates migration of ovarian cancer cells. Cell Biol Int 2010;34(5):493-502. 162. Vielmetter J, Chen XN, Miskevich F, et al. Molecular characterization of human neogenin, a DCC-related protein, and the mapping of its gene (NEO1) to chromosomal position 15q22.3-q23. Genomics 1997;41(3):414-21. 163. Latil A, Chene L, Cochant-Priollet B, et al. Quantification of expression of netrins, slits and their receptors in human prostate tumors. Int J Cancer 2003;103(3):306-15. 164. Goke R, Gregel C, Goke A, Arnold R, Schmidt H, Lankat-Buttgereit B. Programmed cell death protein 4 (PDCD4) acts as a tumor suppressor in neuroendocrine tumor cells. Ann N Y Acad Sci 2004;1014:220-1. 165. Gao F, Zhang P, Zhou C, et al. Frequent loss of PDCD4 expression in human glioma: possible role in the tumorigenesis of glioma. Oncol Rep 2007;17(1):123-8. 166. Goke R, Barth P, Schmidt A, Samans B, Lankat-Buttgereit B. Programmed cell death protein 4 suppresses CDK1/cdc2 via induction of p21(Waf1/Cip1). Am J Physiol Cell Physiol 2004;287(6):C1541-6. 167. Wei ZT, Zhang X, Wang XY, et al. PDCD4 inhibits the malignant phenotype of ovarian cancer cells. Cancer Sci 2009. 168. Murphy N, Ring M, Heffron CC, et al. Quantitation of CDC6 and MCM5 mRNA in cervical intraepithelial neoplasia and invasive squamous cell carcinoma of the cervix. Mod Pathol 2005;18(6):844-9. 169. Williams GH, Swinn R, Prevost AT, et al. Diagnosis of oesophageal cancer by detection of minichromosome maintenance 5 protein in gastric aspirates. Br J Cancer 2004;91(4):714-9.

Page 215: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

199

170. Terry K, McGrath M, Lee IM, Buring J, De Vivo I. Genetic variation in CYP11A1 and StAR in relation to endometrial cancer risk. Gynecol Oncol 2010;117(2):255-9. 171. Celhar T, Gersak K, Ovcak Z, Sedmak B, Mlinaric-Rascan I. The presence of the CYP11A1 (TTTTA)6 allele increases the risk of biochemical relapse in organ confined and low-grade prostate cancer. Cancer Genet Cytogenet 2008;187(1):28-33. 172. Rodriguez EM, Dunham EE, Martin GS. Atypical protein kinase C activity is required for extracellular matrix degradation and invasion by Src-transformed cells. J Cell Physiol 2009;221(1):171-82. 173. Grifoni D, Garoia F, Bellosta P, et al. aPKCzeta cortical loading is associated with Lgl cytoplasmic release and tumor growth in Drosophila and human epithelia. Oncogene 2007;26(40):5960-5. 174. Guo H, Gu F, Li W, et al. Reduction of protein kinase C zeta inhibits migration and invasion of human glioblastoma cells. J Neurochem 2009;109(1):203-13. 175. Hirai T, Chida K. Protein kinase Czeta (PKCzeta): activation mechanisms and cellular functions. J Biochem 2003;133(1):1-7. 176. Liu Y, Wang B, Wang J, et al. Down-regulation of PKCzeta expression inhibits chemotaxis signal transduction in human lung cancer cells. Lung Cancer 2009;63(2):210-8. 177. Sun R, Gao P, Chen L, et al. Protein kinase C zeta is required for epidermal growth factor-induced chemotaxis of human breast cancer cells. Cancer Res 2005;65(4):1433-41. 178. Xiao H, Goldthwait DA, Mapstone T. The identification of four protein kinase C isoforms in human glioblastoma cell lines: PKC alpha, gamma, epsilon, and zeta. J Neurosurg 1994;81(5):734-40. 179. Denison SR, Callahan G, Becker NA, Phillips LA, Smith DI. Characterization of FRA6E and its potential role in autosomal recessive juvenile parkinsonism and ovarian cancer. Genes Chromosomes Cancer 2003;38(1):40-52. 180. Gotlieb WH, Bruchim I, Gu J, et al. Insulin-like growth factor receptor I targeting in epithelial ovarian cancer. Gynecol Oncol 2006;100(2):389-96. 181. Helleman J, Jansen MP, Span PN, et al. Molecular profiling of platinum resistant ovarian cancer. Int J Cancer 2006;118(8):1963-71. 182. Heyman L, Kellouche S, Fernandes J, Dutoit S, Poulain L, Carreiras F. Vitronectin and its receptors partly mediate adhesion of ovarian cancer cells to peritoneal mesothelium in vitro. Tumour Biol 2008;29(4):231-44. 183. Partheen K, Levan K, Osterberg L, et al. Four potential biomarkers as prognostic factors in stage III serous ovarian adenocarcinomas. Int J Cancer 2008;123(9):2130-7. 184. Runnebaum IB, Bruning A. Glucocorticoids inhibit cell death in ovarian cancer and up-regulate caspase inhibitor cIAP2. Clin Cancer Res 2005;11(17):6325-32. 185. Sugai M, Umezu H, Yamamoto T, et al. Expression of hepatocyte nuclear factor 4 alpha in primary ovarian mucinous tumors. Pathol Int 2008;58(11):681-6. 186. Wei Y, Xia W, Zhang Z, et al. Loss of trimethylation at lysine 27 of histone H3 is a predictor of poor outcome in breast, ovarian, and pancreatic cancers. Mol Carcinog 2008;47(9):701-6.

Page 216: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

200

187. Caslini C, Capo-chichi CD, Roland IH, Nicolas E, Yeung AT, Xu XX. Histone modifications silence the GATA transcription factor genes in ovarian cancer. Oncogene 2006;25(39):5446-61. 188. Ye H, Pungpravat N, Huang BL, et al. Genomic assessments of the frequent loss of heterozygosity region on 8p21.3-p22 in head and neck squamous cell carcinoma. Cancer Genet Cytogenet 2007;176(2):100-6. 189. Lux MP, Fasching PA, Beckmann MW. Hereditary breast and ovarian cancer: review and future perspectives. J Mol Med 2006;84(1):16-28. 190. Choi KC, Kang SK, Tai CJ, Auersperg N, Leung PC. Estradiol up-regulates antiapoptotic Bcl-2 messenger ribonucleic acid and protein in tumorigenic ovarian surface epithelium cells. Endocrinology 2001;142(6):2351-60. 191. Ding JX, Feng YJ, Yao LQ, Yu M, Jin HY, Yin LH. The reinforcement of invasion in epithelial ovarian cancer cells by 17 beta-Estradiol is associated with up-regulation of Snail. Gynecol Oncol 2006;103(2):623-30. 192. Crusio KM, King B, Reavie LB, Aifantis I. The ubiquitous nature of cancer: the role of the SCF(Fbw7) complex in development and transformation. Oncogene 2010. 193. Tan Y, Sangfelt O, Spruck C. The Fbxw7/hCdc4 tumor suppressor in human cancer. Cancer Lett 2008;271(1):1-12. 194. Minella AC, Clurman BE. Mechanisms of tumor suppression by the SCF(Fbw7). Cell Cycle 2005;4(10):1356-9. 195. Skaar JR, Pagano M. Control of cell growth by the SCF and APC/C ubiquitin ligases. Curr Opin Cell Biol 2009;21(6):816-24. 196. Koepp DM, Schaefer LK, Ye X, et al. Phosphorylation-dependent ubiquitination of cyclin E by the SCFFbw7 ubiquitin ligase. Science 2001;294(5540):173-7. 197. Yada M, Hatakeyama S, Kamura T, et al. Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7. Embo J 2004;23(10):2116-25. 198. Nateri AS, Riera-Sans L, Da Costa C, Behrens A. The ubiquitin ligase SCFFbw7 antagonizes apoptotic JNK signaling. Science 2004;303(5662):1374-8. 199. Wu G, Lyapina S, Das I, et al. SEL-10 is an inhibitor of notch signaling that targets notch for ubiquitin-mediated protein degradation. Mol Cell Biol 2001;21(21):7403-15. 200. Mao JH, Kim IJ, Wu D, et al. FBXW7 targets mTOR for degradation and cooperates with PTEN in tumor suppression. Science 2008;321(5895):1499-502. 201. Schwab M, Tyers M. Cell cycle. Archipelago of destruction. Nature 2001;413(6853):268-9. 202. Rajagopalan H, Jallepalli PV, Rago C, et al. Inactivation of hCDC4 can cause chromosomal instability. Nature 2004;428(6978):77-81. 203. Spruck CH, Strohmaier H, Sangfelt O, et al. hCDC4 gene mutations in endometrial cancer. Cancer Res 2002;62(16):4535-9. 204. Strohmaier H, Spruck CH, Kaiser P, Won KA, Sangfelt O, Reed SI. Human F-box protein hCdc4 targets cyclin E for proteolysis and is mutated in a breast cancer cell line. Nature 2001;413(6853):316-22. 205. Moberg KH, Bell DW, Wahrer DC, Haber DA, Hariharan IK. Archipelago regulates Cyclin E levels in Drosophila and is mutated in human cancer cell lines. Nature 2001;413(6853):311-6.

Page 217: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

201

206. Li LC, Dahiya R. MethPrimer: designing primers for methylation PCRs. Bioinformatics 2002;18(11):1427-31. 207. Akhoondi S, Sun D, von der Lehr N, et al. FBXW7/hCDC4 is a general tumor suppressor in human cancer. Cancer Res 2007;67(19):9006-12. 208. Kimchi-Sarfaty C, Oh JM, Kim IW, et al. A "silent" polymorphism in the MDR1 gene changes substrate specificity. Science 2007;315(5811):525-8. 209. Kwak EL, Moberg KH, Wahrer DC, et al. Infrequent mutations of Archipelago (hAGO, hCDC4, Fbw7) in primary ovarian cancer. Gynecol Oncol 2005;98(1):124-8. 210. Wajed SA, Laird PW, DeMeester TR. DNA methylation: an alternative pathway to cancer. Ann Surg 2001;234(1):10-20. 211. Asadollahi R, Hyde CA, Zhong XY. Epigenetics of ovarian cancer: from the lab to the clinic. Gynecol Oncol 2010;118(1):81-7. 212. Gu Z, Inomata K, Mitsui H, Horii A. Promoter hypermethylation is not the major mechanism for inactivation of the FBXW7 beta-form in human gliomas. Genes Genet Syst 2008;83(4):347-52. 213. Mao JH, Perez-Losada J, Wu D, et al. Fbxw7/Cdc4 is a p53-dependent, haploinsufficient tumour suppressor gene. Nature 2004;432(7018):775-9. 214. Partheen K, Levan K, Osterberg L, Helou K, Horvath G. Analysis of cytogenetic alterations in stage III serous ovarian adenocarcinoma reveals a heterogeneous group regarding survival, surgical outcome, and substage. Genes Chromosomes Cancer 2004;40(4):342-8. 215. Sterian A, Kan T, Berki AT, et al. Mutational and LOH analyses of the chromosome 4q region in esophageal adenocarcinoma. Oncology 2006;70(3):168-72. 216. Milne AN, Leguit R, Corver WE, et al. Loss of CDC4/FBXW7 in gastric carinoma. Cell Oncol 2010;32(5-6):347-59. 217. Rosenberg E, Demopoulos RI, Zeleniuch-Jacquotte A, et al. Expression of cell cycle regulators p57(KIP2), cyclin D1, and cyclin E in epithelial ovarian tumors and survival. Hum Pathol 2001;32(8):808-13. 218. Schraml P, Bucher C, Bissig H, et al. Cyclin E overexpression and amplification in human tumours. J Pathol 2003;200(3):375-82. 219. Courjal F, Louason G, Speiser P, Katsaros D, Zeillinger R, Theillet C. Cyclin gene amplification and overexpression in breast and ovarian cancers: evidence for the selection of cyclin D1 in breast and cyclin E in ovarian tumors. Int J Cancer 1996;69(4):247-53. 220. Marchini S, Mariani P, Chiorino G, et al. Analysis of gene expression in early-stage ovarian cancer. Clin Cancer Res 2008;14(23):7850-60. 221. Rosen DG, Yang G, Deavers MT, et al. Cyclin E expression is correlated with tumor progression and predicts a poor prognosis in patients with ovarian carcinoma. Cancer 2006;106(9):1925-32. 222. Farley J, Smith LM, Darcy KM, et al. Cyclin E expression is a significant predictor of survival in advanced, suboptimally debulked ovarian epithelial cancers: a Gynecologic Oncology Group study. Cancer Res 2003;63(6):1235-41. 223. Donson AM, Banerjee A, Gamboni-Robertson F, Fleitz JM, Foreman NK. Protein kinase C zeta isoform is critical for proliferation in human glioblastoma cell lines. J Neurooncol 2000;47(2):109-15.

Page 218: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

202

224. Huang L, Muthuswamy SK. Polarity protein alterations in carcinoma: a focus on emerging roles for polarity regulators. Curr Opin Genet Dev 2010;20(1):41-50. 225. Magdalena J, Millard TH, Machesky LM. Microtubule involvement in NIH 3T3 Golgi and MTOC polarity establishment. J Cell Sci 2003;116(Pt 4):743-56. 226. Moscat J, Sanz L, Sanchez P, Diaz-Meco MT. Regulation and role of the atypical PKC isoforms in cell survival during tumor transformation. Adv Enzyme Regul 2001;41:99-120. 227. Wang YM, Seibenhener ML, Vandenplas ML, Wooten MW. Atypical PKC zeta is activated by ceramide, resulting in coactivation of NF-kappaB/JNK kinase and cell survival. J Neurosci Res 1999;55(3):293-302. 228. Joung I, Kim HJ, Kwon YK. p62 modulates Akt activity via association with PKCzeta in neuronal survival and differentiation. Biochem Biophys Res Commun 2005;334(2):654-60. 229. Win HY, Acevedo-Duncan M. Atypical protein kinase C phosphorylates IKKalphabeta in transformed non-malignant and malignant prostate cell survival. Cancer Lett 2008;270(2):302-11. 230. Martin P, Duran A, Minguet S, et al. Role of zeta PKC in B-cell signaling and function. Embo J 2002;21(15):4049-57. 231. Kim DS, Jeon OH, Lee HD, Yoo KH, Kim DS. Integrin alphavbeta3-mediated transcriptional regulation of TIMP-1 in a human ovarian cancer cell line. Biochem Biophys Res Commun 2008;377(2):479-83. 232. Peng Y, Sigua CA, Murr MM. Protein kinase C-zeta mediates apoptosis of mouse Kupffer cells via ERK-1/2: A novel mechanism. Surgery. 233. Xin M, Gao F, May WS, Flagg T, Deng X. Protein kinase Czeta abrogates the proapoptotic function of Bax through phosphorylation. J Biol Chem 2007;282(29):21268-77. 234. Nazarenko I, Jenny M, Keil J, et al. Atypical protein kinase C zeta exhibits a proapoptotic function in ovarian cancer. Mol Cancer Res 2010;8(6):919-34. 235. Mustafi R, Cerda S, Chumsangsri A, Fichera A, Bissonnette M. Protein Kinase-zeta inhibits collagen I-dependent and anchorage-independent growth and enhances apoptosis of human Caco-2 cells. Mol Cancer Res 2006;4(9):683-94. 236. Cohen EE, Lingen MW, Zhu B, et al. Protein kinase C zeta mediates epidermal growth factor-induced growth of head and neck tumor cells by regulating mitogen-activated protein kinase. Cancer Res 2006;66(12):6296-303. 237. Della Peruta M, Giagulli C, Laudanna C, Scarpa A, Sorio C. RHOA and PRKCZ control different aspects of cell motility in pancreatic cancer metastatic clones. Mol Cancer 2010;9:61. 238. Moscat J, Diaz-Meco MT. The atypical protein kinase Cs. Functional specificity mediated by specific protein adapters. EMBO Rep 2000;1(5):399-403. 239. Eder AM, Sui X, Rosen DG, et al. Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proc Natl Acad Sci U S A 2005;102(35):12519-24. 240. Baldwin RM, Barrett GM, Parolin DA, et al. Coordination of glioblastoma cell motility by PKCiota. Mol Cancer 2010;9:233.

Page 219: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

203

241. Win HY, Acevedo-Duncan M. Role of protein kinase C-iota in transformed non-malignant RWPE-1 cells and androgen-independent prostate carcinoma DU-145 cells. Cell Prolif 2009;42(2):182-94. 242. Zhang L, Huang J, Yang N, et al. Integrative genomic analysis of protein kinase C (PKC) family identifies PKCiota as a biomarker and potential oncogene in ovarian carcinoma. Cancer Res 2006;66(9):4627-35. 243. Suzuki A, Akimoto K, Ohno S. Protein kinase C lambda/iota (PKClambda/iota): a PKC isotype essential for the development of multicellular organisms. J Biochem 2003;133(1):9-16. 244. Suzuki A, Yamanaka T, Hirose T, et al. Atypical protein kinase C is involved in the evolutionarily conserved par protein complex and plays a critical role in establishing epithelia-specific junctional structures. J Cell Biol 2001;152(6):1183-96. 245. Abramovitch S, Glaser T, Ouchi T, Werner H. BRCA1-Sp1 interactions in transcriptional regulation of the IGF-IR gene. FEBS Lett 2003;541(1-3):149-54. 246. Abramovitch S, Werner H. Functional and physical interactions between BRCA1 and p53 in transcriptional regulation of the IGF-IR gene. Horm Metab Res 2003;35(11-12):758-62. 247. Maor S, Yosepovich A, Papa MZ, et al. Elevated insulin-like growth factor-I receptor (IGF-IR) levels in primary breast tumors associated with BRCA1 mutations. Cancer Lett 2007;257(2):236-43. 248. An Y, Cai L, Wang Y, Zhu D, Guan Y, Zheng J. Local expression of insulin-like growth factor-I, insulin-like growth factor-I receptor, and estrogen receptor alpha in ovarian cancer. Onkologie 2009;32(11):638-44. 249. Ouban A, Muraca P, Yeatman T, Coppola D. Expression and distribution of insulin-like growth factor-1 receptor in human carcinomas. Hum Pathol 2003;34(8):803-8. 250. Eckstein N, Servan K, Hildebrandt B, et al. Hyperactivation of the insulin-like growth factor receptor I signaling pathway is an essential event for cisplatin resistance of ovarian cancer cells. Cancer Res 2009;69(7):2996-3003. 251. Yee D, Morales FR, Hamilton TC, Von Hoff DD. Expression of insulin-like growth factor I, its binding proteins, and its receptor in ovarian cancer. Cancer Res 1991;51(19):5107-12. 252. Spentzos D, Cannistra SA, Grall F, et al. IGF axis gene expression patterns are prognostic of survival in epithelial ovarian cancer. Endocr Relat Cancer 2007;14(3):781-90. 253. Beck EP, Russo P, Gliozzo B, et al. Identification of insulin and insulin-like growth factor I (IGF I) receptors in ovarian cancer tissue. Gynecol Oncol 1994;53(2):196-201. 254. Jakubowska A, Rozkrut D, Antoniou A, Hamann U, Lubinski J. The Leu33Pro polymorphism in the ITGB3 gene does not modify BRCA1/2-associated breast or ovarian cancer risks: results from a multicenter study among 15,542 BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat;121(3):639-49. 255. Feng D, Lindpaintner K, Larson MG, et al. Increased platelet aggregability associated with platelet GPIIIa PlA2 polymorphism: the Framingham Offspring Study. Arterioscler Thromb Vasc Biol 1999;19(4):1142-7.

Page 220: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

204

256. Vijayan KV, Liu Y, Dong JF, Bray PF. Enhanced activation of mitogen-activated protein kinase and myosin light chain kinase by the Pro33 polymorphism of integrin beta 3. J Biol Chem 2003;278(6):3860-7. 257. Kaur S, Kenny HA, Jagadeeswaran S, et al. {beta}3-integrin expression on tumor cells inhibits tumor progression, reduces metastasis, and is associated with a favorable prognosis in patients with ovarian cancer. Am J Pathol 2009;175(5):2184-96. 258. Partheen K, Levan K, Osterberg L, Claesson I, Sundfeldt K, Horvath G. External validation suggests Integrin beta 3 as prognostic biomarker in serous ovarian adenocarcinomas. BMC Cancer 2009;9:336. 259. Toker A, Yoeli-Lerner M. Akt signaling and cancer: surviving but not moving on. Cancer Res 2006;66(8):3963-6. 260. McCoy ML, Mueller CR, Roskelley CD. The role of the breast cancer susceptibility gene 1 (BRCA1) in sporadic epithelial ovarian cancer. Reprod Biol Endocrinol 2003;1:72. 261. Della Peruta M, Giagulli C, Laudanna C, Scarpa A, Sorio C. RHOA and PRKCZ control different aspects of cell motility in pancreatic cancer metastatic clones. Mol Cancer;9:61. 262. Neuhausen SL, Brummel S, Ding YC, et al. Genetic variation in insulin-like growth factor signaling genes and breast cancer risk among BRCA1 and BRCA2 carriers. Breast Cancer Res 2009;11(5):R76. 263. Chitnis MM, Yuen JS, Protheroe AS, Pollak M, Macaulay VM. The type 1 insulin-like growth factor receptor pathway. Clin Cancer Res 2008;14(20):6364-70. 264. Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005;102(43):15545-50. 265. Amundadottir L, Kraft P, Stolzenberg-Solomon RZ, et al. Genome-wide association study identifies variants in the ABO locus associated with susceptibility to pancreatic cancer. Nat Genet 2009;41(9):986-90. 266. Do TN, Ucisik-Akkaya E, Davis CF, Morrison BA, Dorak MT. An intronic polymorphism of IRF4 gene influences gene transcription in vitro and shows a risk association with childhood acute lymphoblastic leukemia in males. Biochim Biophys Acta 2010;1802(2):292-300. 267. Gu F, Qureshi AA, Niu T, et al. Interleukin and interleukin receptor gene polymorphisms and susceptibility to melanoma. Melanoma Res 2008;18(5):330-5. 268. Jou YS, Lo YL, Hsiao CF, et al. Association of an EGFR intron 1 SNP with never-smoking female lung adenocarcinoma patients. Lung Cancer 2009;64(3):251-6. 269. Levin AM, Machiela MJ, Zuhlke KA, Ray AM, Cooney KA, Douglas JA. Chromosome 17q12 variants contribute to risk of early-onset prostate cancer. Cancer Res 2008;68(16):6492-5. 270. Wang-Gohrke S, Weikel W, Risch H, et al. Intron variants of the p53 gene are associated with increased risk for ovarian cancer but not in carriers of BRCA1 or BRCA2 germline mutations. Br J Cancer 1999;81(1):179-83. 271. Zhou J, Nagarkatti PS, Zhong Y, Creek K, Zhang J, Nagarkatti M. Unique SNP in CD44 intron 1 and its role in breast cancer development. Anticancer Res 2010;30(4):1263-72.

Page 221: Identification of Genes and Potential Pathways Involved in ... · Identification of Genes and Potential Pathways Involved in . Familial Ovarian Cancer . Degree of Doctor of Philosophy,

205

272. Nowak D, Mossner M, Baldus CD, Hopfer O, Thiel E, Hofmann WK. Mutation analysis of hCDC4 in AML cells identifies a new intronic polymorphism. Int J Med Sci 2006;3(4):148-51. 273. Yu JC, Ding SL, Chang CH, et al. Genetic susceptibility to the development and progression of breast cancer associated with polymorphism of cell cycle and ubiquitin ligase genes. Carcinogenesis 2009;30(9):1562-70. 274. DelloRusso C, Welcsh PL, Wang W, Garcia RL, King MC, Swisher EM. Functional characterization of a novel BRCA1-null ovarian cancer cell line in response to ionizing radiation. Mol Cancer Res 2007;5(1):35-45. 275. Hirano S, Ito N, Takahashi S, Tamaya T. Clinical implications of insulin-like growth factors through the presence of their binding proteins and receptors expressed in gynecological cancers. Eur J Gynaecol Oncol 2004;25(2):187-91. 276. Terry KL, Tworoger SS, Gates MA, Cramer DW, Hankinson SE. Common genetic variation in IGF1, IGFBP1 and IGFBP3 and ovarian cancer risk. Carcinogenesis 2009;30(12):2042-6. 277. Carreiras F, Denoux Y, Staedel C, Lehmann M, Sichel F, Gauduchon P. Expression and localization of alpha v integrins and their ligand vitronectin in normal ovarian epithelium and in ovarian carcinoma. Gynecol Oncol 1996;62(2):260-7.