81
Investigating the Role of PHIP1 in Breast Cancer by Chan Mi Lee A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Laboratory Medicine & Pathobiology University of Toronto, Canada © Copyright by Chan Mi Lee, 2011.

Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

Investigating the Role of PHIP1 in Breast Cancer

by

Chan Mi Lee

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Department of Laboratory Medicine & Pathobiology

University of Toronto, Canada

© Copyright by Chan Mi Lee, 2011.

Page 2: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

ii

Investigating the Role of PHIP1 in Breast Cancer

Chan Mi Lee

Master of Science

Laboratory Medicine & Pathobiology

University of Toronto

2011

Abstract

PHIP1 is a novel downstream transcriptional co-regulator of insulin-like growth factor-I receptor (IGF-

IR), a tyrosine kinase receptor that is often elevated and autophosphorylated in breast cancer. In this study,

I show that PHIP1 is upregulated in MCF10A cells stably overexpressing IGF-IR signaling components

and that knock-down of PHIP1 significantly inhibits breast cancer cell proliferation by inducing

transcriptional upregulation of p21 and downregulation of cyclin D2. I also show that stable

overexpression of PHIP1 in MCF10A cells can lead to its proteasomal degradation. Together, our data

indicate that PHIP1 is implicated in breast cancer cell growth and suggest a number of avenues that await

exciting discovery.

Page 3: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

iii

Acknowledgments

My time here at the University of Toronto has been an enormous learning experience and one of the

most memorable moments of my life. However, despite the ups and downs I had, it was a journey

worth undergoing, and it made me stronger and more knowledgeable. As I present this fruit of the

labor, I would like to express appreciation to all those who stood by me on this journey.

My first thanks goes to my supervisor, Dr. Maria Rozakis-Adcock, for her patience throughout the

years, and the opportunity to be part of the exciting research team. I am also grateful all the members

of the Rozakis lab, for sharing their scientific knowledge and for making my time in the laboratory

stimulating and enjoyable.

I would like to sincerely thank my committee members, Dr. Eldad Zacksenhaus and Dr. Stephane

Angers, for their generous time and expertise to advise and guide me through my research education.

I also express my gratitude to all my friends on the sixth floor MSB and the LMP staffs, for their

friendships and all the fun times we had together.

Finally, I specially thank my family for the limitless support, understanding and love. And also to

God, who is my friend for life, for His eternal presence within me.

Page 4: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

iv

Table of Contents

ACKNOWLEDGMENTS ........................................................................................................................ III

TABLE OF CONTENTS ......................................................................................................................... IV

LIST OF TABLES .................................................................................................................................... VI

LIST OF FIGURES ................................................................................................................................. VII

LIST OF ABBREVIATIONS ............................................................................................................... VIII

CHAPTER 1. INTRODUCTION ...............................................................................................................1

1.1 Breast Cancer ..................................................................................................................................1

1.1.1 Mammary gland structure ......................................................................................................1

1.1.2 Classification of breast cancer ...............................................................................................3

1.1.3 Classical oncogenes in mammary gland tumorigenesis ........................................................4

1.1.3.1 HER2 ................................................................................................................................................................ 4

1.1.3.2 c-Myc ................................................................................................................................................................ 5

1.1.3.3 Cyclin D1 .......................................................................................................................................................... 5

1.2 Insulin-like Growth Factor type I Receptor (IGF-IR) pathway .................................................6

1.2.1 Insulin-like growth factor type I receptor (IGF-IR) structure ..............................................6

1.2.2 Downstream signaling ............................................................................................................7

1.2.2.1 PI3K/p-Akt pathway ....................................................................................................................................... 8

1.2.2.2 Ras/MAPK pathway ....................................................................................................................................... 9

1.2.3 Involvement in breast cancer .................................................................................................9

1.3 Pleckstrin-homology domain Interacting Protein (PHIP) ........................................................ 10

1.3.1 Discovery and characterization ............................................................................................ 10

1.3.2 PHIP1 structure and functional domains ............................................................................ 12

1.3.2.1 WD40-repeat domain .................................................................................................................................... 12

1.3.2.2 Bromodomains ............................................................................................................................................... 13

1.3.3 PHIP1 as DCAF14 ............................................................................................................... 14

1.3.3.1 Ubiquitin-Proteasome Pathway (UPP) and CUL4-DDB1 Ubiquitin Ligase ............................................. 14

1.3.3.2 Involvement of Cul4 in breast cancer .......................................................................................................... 16

1.4 Hypothesis and Rationale............................................................................................................. 17

Page 5: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

v

CHAPTER 2. MATERIALS AND METHODS ..................................................................................... 18

2.1 Reagents and antibodies ............................................................................................................... 18

2.2 Cell culture .................................................................................................................................... 18

2.3 Virus production and transduction ............................................................................................. 19

2.4 siRNA transfection for transient knockdown experiments ....................................................... 20

2.5 Cell proliferation assays ............................................................................................................... 21

2.6 Soft agar anchorage-independent colony formation assay ....................................................... 21

2.7 SDS-PAGE Electrophoresis and Western Blotting ................................................................... 22

2.8 Immunoprecipitations .................................................................................................................. 23

2.9 Propidium iodide (PI) staining and FACS analysis ................................................................... 25

2.10 RNA Isolation and real-time quantitative PCR (RT-qPCR) .................................................... 25

CHAPTER 3. RESULTS .......................................................................................................................... 28

3.1 PHIP1 is overexpressed in primary breast tumor samples of MMTV transgenic mouse

models and in MCF10A stable cell lines overexpressing members of the IGF-IR pathway. . 28

3.2 MCF10A-PHIP1 stable cell line spontaneously silences ectopic PHIP1 expression at the

protein level. .................................................................................................................................. 28

3.3 AdPHIP1 is not the ideal system for the soft agar colony-forming assay ................................ 34

3.4 TRIPZ shRNA-PHIP constructs fail to maintain PHIP knockdown. ...................................... 36

3.5 PHIP1 is implicated in cancer cell proliferation ........................................................................ 40

3.6 PHIP1 may complex with p53 ..................................................................................................... 45

CHAPTER 4. DISCUSSION AND FUTURE DIRECTIONS ............................................................... 48

4.1 PHIP1 is differentially expressed in cancer cells. ...................................................................... 48

4.2 PHIP1 as a component of the nuclear proteasome and transcriptional machinery ............... 51

4.3 Role of PHIP1 in cell-cycle progression ...................................................................................... 56

4.4 Putting PHIP1 in the context of breast cancer and IGF-IR signaling ..................................... 59

REFERENCES .......................................................................................................................................... 62

Page 6: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

vi

List of Tables

Table 1. Table of antibodies used in the study with their dilutions ............................................................. 23

Table 2. Sequence of qPCR primers used in the study ................................................................................ 26

Page 7: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

vii

List of Figures

Figure 1. The human mammary gland. .........................................................................................................2

Figure 2. Structure of IGF-IR. .......................................................................................................................6

Figure 3. IGF-IR signaling pathway. .............................................................................................................8

Figure 4. Different variants of PHIP. .......................................................................................................... 12

Figure 5. E3 ubiquitin ligases of different Cullin proteins. ......................................................................... 16

Figure 6. PHIP is manifested in the primary breast tumor samples of MMTV transgenic mouse models of

classical oncogenes, and in MCF10A stable cell lines with constitutively active IGF-IR pathway. .......... 29

Figure 7. Retroviral infection of MCF10A leads to robust expression of HA. ........................................... 31

Figure 8. Loss of HA-PHIP expression in the MCF10A stables. ................................................................ 32

Figure 9. PHIP is targeted for protein degradation in MCF10A-PHIP stable cell line. .............................. 33

Figure 10. Soft agar results with MCF10A cells infected with PHIP1 adenovirus. .................................... 35

Figure 11 Analyses of MCF10A-IGF-IR TRIPZ stable cell lines. .............................................................. 38

Figure 12. Validation of TRIPZ constructs in 293T and MDA-MB-231 cells. .......................................... 39

Figure 13. PHIP knockdown inhibits the growth of MCF10A-IGFR cells. ................................................ 41

Figure 14. PHIP1 knockdown inhibits the growth of both ER positive and ER negative cell lines............ 42

Figure 15. PHIP knockdown in MDA-MB-231 arrests G1 to S phase transition of cell cycle. .................. 43

Figure 16. PHIP complexes with p53. ......................................................................................................... 46

Figure 17. A model for role of PHIP1 as part of the ubiquitin-proteasome complex and in the regulation

of transcription initiation. ............................................................................................................................ 53

Page 8: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

viii

List of Abbreviations

7-AAD aa 3’UTR

7-aminoactinomycin D amino acid 3’untranslated region

BD bHLH

bromodomain (domain) basic helix loop helix

BSA bovine serum albumin CAF CBP CDK

chromatin assembly factor-1 CREB binding protein cyclin dependent kinase

CKI cyclin dependent kinase inhibitor CIS carcinoma in situ CRL CUL

cullin-RING finger ligase cullin (protein)

DCAF DDB1 DMEM

DDB1 and CUL4-associated factor damaged DNA binding protein Dulbecco's modified eagle medium

DMSO DNA DTT ECL

dimethyl sulfoxide deoxyribonucleic acid dithiothreitol enhanced chemiluminescent

ECM extracellular matrix EDTA EGF EGFP EMT

ethylenediaminetetraacetic acid epidermal growth factor enhanced green fluorescent protein epithelial-to-mesenchymal transition

ErbB ER ERK1/2 ETD

avian erythroblastosis oncogene B estrogen receptor extracellular signal-regulated kinase 1/2 extralobular terminal duct

FAK FACS FBS

focal adhesion kinase fluorescence activated cell sorting fetal bovine serum

FISH G1 phase G2 phase GAP GAPDH

fluorescent in situ hybridization cell cycle gap 1 cell cycle gap 2 GTPase activating protein glyceraldehyde 3-phosphate dehydrogenase

GFP green fluorescent protein GSK-3β glycogen synthase kinase-3β GST glutathione S-transferase (tag) HA hemagglutinin (tag) HAT HDAC HEK293

histone acetyltranferase histone deacetylase human embryonic kidney cells

HEPES HER2

N-2-hydroxyethyl piperazine-N-ethanesulphonic acid human epidermal growth factor receptor 2

Page 9: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

ix

HRP horseradish peroxidase IGF-I IGF-IR IHC IR IRES IRS-1 ITD L MAPK MIEV MMTV MOI MSCV MTT NF-κB NGF ORF P300 PAGE PBS PCAF PCNA PCR PEI PHIP PI PI3K PIC PMA PMSF Pol II PPi PR PTB RB RING RIPA RTK SCID S phase SDS SFM SHC shRNA siRNA SNP TAD TBP TBST

insulin-like growth factor-1 insulin-like growth factor-1 receptor immunohistochemistry insulin receptor internal ribosome entry site insulin receptor substrate-1 intralobular terminal duct litre mitogen-activated protein kinase MSCV-IRES-EGFP vector mouse mammary tumor virus multiplicity of infection murine stem cell virus 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide nuclear factor-κ B nerve growth factor open reading frame E1A binding protein p300 polyacrylamide gel electrophoresis phosphate buffered saline P300/CBP-associated factor proliferating cell nuclear antigen polymerase chain reaction polyethyleneimine pleckstrin-homology domain interacting protein propidium iodide phosphatidyl inositol-3 kinase protease inhibitor cocktail phorbol ester phenylmethanesulphonylfluoride RNA polymerase II inorganic pyrophosphate progesterone receptor phosphotyrosine binding retinoblastoma susceptibility gene really interesting new gene (domain) radioimmuno precipitation assay buffer receptor tyrosine kinase severe combined immunodeficiency DNA synthesis phase sodium dodecyl sulphate serum free media src/collagen-homology short hairpin RNA small interfering RNA single nucleotide polymorphism transactivation domain TATA-binding protein tris-buffered saline tween-20

Page 10: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

x

TDLU TF TGF-β TMA UPP WAP WDR WT

terminal ductal-lobular unit transcription factor transforming growth factor β tissue microarray ubiquitin-proteasome pathway whey acidic protein tryptophan-aspartic acid repeat (domain) wild-type

Page 11: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

1

Chapter 1. Introduction

1.1 Breast Cancer

Breast cancer is the most common cause of cancer among women globally (Bray et al, 2004) as well

as in Canada (Canadian Cancer Society, 2010). It imposes a significant burden to public health with

high morbidity and mortality rates, and in 2010 alone, 23,200 women in Canada are estimated to be

diagnosed with breast cancer while resulting in 5,300 deaths. In addition, approximately 1 in 9

women will develop breast cancer during their lifetime while 1 out of 28 will die of the disease.

The probability of being diagnosed with breast cancer in women increases dramatically with

age. As an example, when compared to women in their 20s of age, the risk is more than 10-fold for

women in their 30s, more than 100-fold for women in 50s and larger than 1,000-fold for women in

their 80s (Schedin, 2006). Given the fact that more women are giving birth to first child in their 30s

in modern society and the average age of child-bearing is increasing steadily (Kroman et al, 2003),

the morbidity and mortality due to or associated with breast cancer are also likely to be augmented in

the future. However, despite the huge research efforts to elucidate the mechanism of breast

tumorigenesis over the past decades, our understanding of the communication networks is yet

incomplete. There is thus a notable need for more comprehensive knowledge of the complex cellular

events that are involved in the transformation, development and metastasis of breast cancer cells.

1.1.1 Mammary gland structure

Each breast harbors the mammary gland, which is comprised of 15 to 20 lobes, or compartments.

Each lobe, in turn, consists of smaller lobules that contain milk-secreting glands called alveoli in

grapelike clusters surrounded by the connective tissue (Tortora and Derrickson, 2006). The

functional unit of the mammary gland is called the terminal ductal lobular unit (TDLU), which

consists of extralobular terminal duct (ETD) and intralobular terminal duct (ITD) (Allred et al, 2001;

Ohuchi et al, 1984; Figure 1A). The ITD refers to the central space of the lobule and have

outpouchings called acini, or ductules. (Ohuchi et al, 1984). The acini, in turn, contain a hollow

lumen surrounded by a single polarized layer of luminal epithelial cells (Allred et al, 2001; Bissell et

al, 2002).

There are two major different tissue structures that define each mammary gland: the

epithelium and the stroma. The adenocarcinoma of the breast is thought to originate from the

epithelial cells of the TDLU (Bissell et al, 2002). The epithelium forms the ducts and alveoli, which

Page 12: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

2

are collectively connected as the lactiferous duct and open to the nipple for the release of milk. Most

of the epithelial cells are luminal and undergo functional differentiation during pregnancy to become

secretory for milk production, which is largely stimulated by the hormone prolactin from the anterior

pituitary (Tortora and Derrickson, 2006; Hennighausen and Robinson, 2006). The luminal cells are

enclosed by basal, myoepithelial cells that are contractile (Figure 1B) and aid in the delivery of milk

under the influence of the hormone oxytocin, which is released from the posterior pituitary upon

suckling. The stroma, also called the mammary fat pad, refers to the connective tissue which is

composed mostly of the adipocytes, fibroblasts, blood vessels, and neurons and embeds the extensive

branches of epithelial cells. The mammary epithelial extracellular matrix containing the

myoepithelial basement membrane and the stroma supports the mammary gland structure and acts as

communicative bridge between the inner epithelium and the surrounding environment during the

organ maturation and hormonal regulation (Wiseman and Werb, 2002). During pregnancy, the

alveolar epithelium expands and produces milk during lactation, but after weaning where the level of

prolactin wanes, it undergoes massive apoptosis and extensive remodeling process called involution

(Hennighausen and Robinson, 2006).

A. B.

Figure 1. The human mammary gland.

(A). Nomenclature of the ductal system. The terminal ductal-lobular unit (TDLU) is the functional unit of the

mammary gland and consists of an ETD, extralobular terminal duct, and ductules. ITD stands for the intralobular

terminal duct. Adopted from (Ohuchi et al, 1984). (B). Schematic diagram of human mammary gland lobule.

Adapted (from Debnath et al, 2003).

Page 13: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

3

1.1.2 Classification of breast cancer

Breast cancer is a clinically heterogeneous disease, and there are a number of variables that are taken

into consideration to determine the prognosis and the likely response to therapies. Breast cancer can

be classified by different criteria to serve different purposes, and examples of the breast cancer

categorizations include histological type, grade, tumor size, involvement of lymph node, estrogen

receptor (ER), progesterone receptor (PR) and HER2/Neu-receptor status, as well as protein and

genetic markers. Tumor pathology refers to the histological appearance of the tumor tissue, and as

the majority of breast cancers are derived from ductal and lobular epithelium, the most common

forms of breast cancer under this category are carcinoma in situ (CIS) and invasive carcinoma.

According to the MERCK Professional Manual, CIS refers to the proliferation and invasion of the

cancer cells within the epithelial cells, while the invasive carcinoma denotes invasion of cancer cells

to the surrounding tissues. The grading system is another form of the histological analysis that

distinguishes between well differentiated (low grade), moderately differentiated (intermediate grade),

and poorly differentiated (high grade) as cells progressively depart from the normal phenotype.

Unsurprisingly, breast cancer of high grade has the worst prognosis. The use of stages for breast

cancer follows the tumor, node, metastasis, or TNM classification, based on the size of tumor (T),

presence of metastasis to the lymph nodes in the armpits, neck and chest (N) or the rest of the body

(M). Staging is usually more accurately determined during surgery and is numerated from 0 to 4,

with 0 being the pre-malignant stage and 4 being the ‘advanced’, or highly metastatic state.

The current most widely used molecular classification in clinical treatments is the receptor

status, according to the presence (positive) or absence (negative) for hormonal receptors ER, PR, and

HER2/Neu. ER status and probably PR contents, for example, are a good prognostic factor and a

positive predictor of response to endocrine therapy (Andre and Pusztai, 2006). Overexpression of

HER2, on the other hand, is generally associated with poor disease outcomes, and basal-like cells

that express none of these receptors are considered more aggressive. The recent recommended

guideline for the use of tumor markers in breast cancer by the American Society of Clinical

Oncology is reviewed by (Harris et al, 2007). The molecular classification is constantly being

updated with more large-scale genomic analyses of breast cancers and the discovery of subsets of

molecular markers within the existing categories, which is hoped to improve selection criteria for

patient treatment for maximum efficacy (reviewed in Andre and Pusztai, 2006).

Page 14: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

4

1.1.3 Classical oncogenes in mammary gland tumorigenesis

In breast cancer, direct activation of proto-oncogenes to transform into oncogenes generally occurs

by one of the three mechanisms: 1. point mutations or sequence changes in the coding or the

promoter region of the gene, as in the case for the RAS family, 2. gene amplification, such as ERBB2

amplification in breast cancer and 3. gene arrangements, most frequently chromosome translocations

which is a prominent form of oncogenic activation in chronic myelogenous leukaemia and Burkitts

lymphoma (Oliveira and Fletcher, 2005). Indirect activation can also occur through suppression of

normal regulatory mechanisms, by the inactivation of tumor suppressors such as p53, RB or BRCA1.

Few examples of the classical oncogenes that are relevant in breast cancer have been outlined below.

1.1.3.1 HER2

HER2, also known as ErbB2, is a member of the epidermal growth factor receptor (EGFR) family of

receptor tyrosine kinases (RTK) normally involved in signal transduction of cell growth and

differentiation (Reese and Slamon, 1997). The HER family consists of HER1, 2, 3 and 4 and upon

ligand binding, it can either homodimerize or heterodimerize with a different member of the family.

However, HER2 is the preferential dimerisation partner of other members of the ErbB family, and its

heterodimers are more stable and lead to more potent signaling (Cardoso et al, 2005). The proto-

oncogene is located on chromosome 17q21-q22 and is amplified in 20 to 30% of human mammary

carcinomas, which are associated with poor prognosis and increased disease recurrence (Cardoso et

al, 2005). Because of its ability to predict response to treatment such as trastuzumab (Herceptin) or

Lapatinib that target the HER2 signaling pathway, HER2 is a well used molecular marker routinely

screened in breast cancer for prognosis. There is much in vivo evidence for HER2 oncogenic activity

to date. MMTV-Neu transgenic mice overexpressing the activated rat homolog of ErbB2 develops

multifocal adenocarcinoma and lung metastases in approximately 15 weeks after pregnancy (Muller

et al, 1988). Similarly, transgenic mammary gland-specific expression of wild-type ErbB2 also leads

to mammary tumor formation at an average age of 7 months and many of the mice developed

secondary metastatic tumor in the lungs (Guy et al, 1992). More recently, Moody and colleagues

demonstrated, using a doxycyline-inducible conditional mouse model, that tumor formation could be

partially regressed upon withdrawing doxycycline. However, most mice exhibited recurrence, which

was accompanied by upregulation of Snail and epithelial-to-mesenchymal transition (EMT) (Moody

et al, 2005).

Page 15: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

5

1.1.3.2 c-Myc

The family of myc proto-oncogenes code for proteins belonging to classical transcription factors with

a characteristic basic helix-loop-helix leucine zipper (bHLHZ) domain and participate in various

aspects of cell function, such as replication, cell growth, metabolism, differentiation and apoptosis

(Liao and Dickson, 2000). The c-Myc protein heterodimerizes with Max, another transcription factor,

and binds to an E-box element containing CAC(G/A)TG motif in the middle of the DNA recognition

site (Haynes and Lane, 2001). The c-myc proto-oncogene, which is located on chromosome 8q24, is

amplified in approximately 16% of breast cancer cases and is associated with decreased disease-free

survival of the patient (Oliveira and Fletcher, 2005). c-myc expression is frequently upregulated in

early breast lesions and the protein c-Myc is overexpressed in approximately 70% of breast tumors,

which is correlated with poor clinical outcome (Liao and Dickson, 2000). In vivo models containing

c-Myc transgene such as MMTV-Myc (Stewart et al, 1984), whey acidic protein (WAP) promoter-

Myc (Schoenenberger et al, 1988), or tetracycline inducible system (D’Cruz et al, 2001) develop

mammary tumors in 7 to 14 months in 50 to 80% of mice. c-Myc is now known to be a downstream

effector of HER2 signaling (Haynes and Lane, 2001).

1.1.3.3 Cyclin D1

Cyclin D1 complexes with cyclin-dependent kinase (CDK) 4 and 6 during the G1 phase of the cell

cycle to phosphorylate the retinoblastoma (Rb) family of tumor suppressors, to release the

checkpoint on the entry into the DNA synthesis (S phase). Cyclin D1 protein is overexpressed in

most breast cancer cases (Steeg and Zhou, 1998) and is associated with poor prognosis (Sutherland

and Musgrove, 2004). The cyclin D1 gene, or CCND1 is localized on chromosome 11q13, and the

locus amplification has been found in up to 15% of breast cancer (Rennstam et al, 2001). As an

activator of cell cycle progression, cyclin D1 overexpression leads to enhanced cell proliferation and

has been shown to participate in the transition from the premalignant breast lesion to carcinoma

(Weinstat-Saslow et al, 1995). Similarly, MMTV-cyclin D1 mouse models show mammary

hyperplasia due to proliferative disturbances and eventually develop mammary adenocarcinoma

(Wang et al, 1994).

Page 16: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

6

1.2 Insulin-like Growth Factor type I Receptor (IGF-IR) pathway

1.2.1 Insulin-like growth factor type I receptor (IGF-IR) structure

Insulin-like growth factor type I receptor (IGF-IR) is a transmembrane tyrosine kinase cell surface

receptor that undergoes dimerization and autophosphorylation of their C-terminal tyrosine residues

upon binding of their ligands, IGF-I and IGF-II (Surmacz, 2000). The functional receptor is a

heterotetramer, with two α and two β chains that are linked by disulfide bonds (Figure 2). The α

chains participate in the ligand binding and provide the binding specificity, while the β subunits

transmit the ligand-induced signal into the cell. There are three major domains that are recognized in

the β subunit: a short juxtramembrane or transmembrane portion that provide docking sites to major

signaling substrates such as insulin receptor substrates (IRS) 1-4 and src/collagen-homology (SHC)

proteins, a tyrosine kinase domain containing the autophosphorylation tyrosine residue clusters, and

the carboxy-terminal domain (C-terminus) that contains important residues necessary in the

association of adaptor proteins required for IGF-I signaling (Dupont et al, 2003). Examples of

proteins that associate with the C-terminal region of the IGF-IR include GRB10, 14-3-3 epsilon, and

p85 subunit of phosphatidyl inositol-3 kinase (PI3K) or SHPTP2 phophatase.

α α

β β

Figure 2. Structure of IGF-IR.

The key residues and binding elements involved in signaling are depicted. Adapted from (Surmacz, 2000).

Page 17: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

7

As its name suggests, IGF-IR is structurally and functionally homologous with the insulin

receptor (IR) and they can heterodimerize to create an insulin/IGF-IR hybrid receptor. Structurally,

the kinase domains of the two receptors are 80-90% identical. However, the C-terminal regions,

which are important in the specificity of signaling pathway triggered through the recruitment of

adaptor proteins, are only 40% equivalent. Tartare and colleagues demonstrated the C-terminal

signaling specificity in signaling using chimeric receptors, which either contained the ligand-binding

domain of IR and cytoplasmic segment of the IGF-IR or vice versa (Tartare et al, 1994). The

functional analyses showed high correlation with the C-terminal region, more than the ligand-binding

domain. Thus, the two receptors have distinct as well as similar physiological roles (Surmacz, 2000).

1.2.2 Downstream signaling

Upon the binding of its ligand, IGF-I, the IGF-IRs cluster and activate their tyrosine kinase to

autophosphorylate and cross-phosphorylate the tyrosine residues in the beta chain, whereby the

phosphorylated specific Tyrosine (Tyr) and Serine (Ser) residues create docking sites for the IGF-IR

signaling molecules. Some of the most well known substrates are IRS-1 and SHC, both of which

bind to Tyr 950 via phosphotyrosine binding (PTB) domain (Surmacz, 2000). The phosphorylated

tyrosine residues of IGF-IR upon activation become the docking sites for adaptor proteins such as

insulin receptor substrate (IRS)-1 and Grb-2, which lead to subsequent activation of downstream

signaling (Figure 3). Although there are four different forms of IRS proteins in the mammalian

system, only IRS-1 and 2 are the most prominent players in the IGF-IR or IR signaling (Surmacz,

2000). There are two major anti-apoptotic pathways induced by IGF-IR activation, which are covered

in more depth in the following sections; 1. Lipid kinase PI3K/Akt and 2. GTPase Ras-Raf-

ERK/MAPK pathway. PI3K/Akt is involved in mitogenesis, metabolism, cell adhesion and motility,

while Ras/MAPK through the activation of Grb2 and Sos is implicated in cell growth and

differentiation (Chitnis et al, 2008; Valentinis et al, 2001).

Page 18: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

8

Figure 3. IGF-IR signaling pathway.

The IGF-IR has an extracellular α domain and a cytosolic β domain. Binding of IGF-I to the receptor leads to

homodimerization of IGF-IR and activation of multiple downstream signaling pathways. Adapted from (Dupont et

al 2003).

1.2.2.1 PI3K/p-Akt pathway

IRS-1 acts as a potent effector of IGF-IR by recruiting and phosphorylating various signaling

molecules, especially PI3K, through the interaction of phosphotyrosine binding domains such as SH2

(Dupont et al, 2003). 3-phosphoinositide-dependent protein kinase 1 (PDK1), some isoforms of

protein kinase C (PKC) and AKT kinases are activated upon increased level of lipid secondary

messengers by PI3K and subsequent localization to the plasma membrane. The PI3K/p-Akt pathway

is a major downstream effector of IRS-1 and is important in cell growth, survival and metabolism.

Akt function covers several biological processes such as cell-cycle progression, growth and apoptosis

through the regulation of key molecules, including mTOR, IKK, and BAD (Tao et al, 2007). The

enhanced cell proliferation induced by Akt occurs in both anti-apoptotic and pro-cell division manner,

as it also impedes the expression and function of cell cycle inhibitors such as, p21, p27, Chk1, and

GSK3 (Chitnis et al, 2008). p70S6 kinase is another effector of Akt, which elevates the expression of

cyclin D1 to initiate the cell cycle progression (Hashemolhosseinin et al, 1998).

Page 19: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

9

1.2.2.2 Ras/MAPK pathway

The Ras/MAP cascade of kinases of the Ras/MAPK pathway are activated downstream of the IGF-

IR signaling when IRS-1 associate with the GRB2/SOS complex. The Ras/MAPK pathway is also

involved in a wide variety of biological processes, such as cell growth and differentiation. The

downstream components of the Ras GTPase include RAF, MEK and extracellular signal-regulated

kinase (ERK) kinase/MAPK proteins. The main role of Ras signaling in G1 to S phase of cell cycle

progression often mediated by the IGF-I or other growth factor signaling is to inactivate RB, as

indicated by studies where cells with RB knockout no longer require RAS activity (Mittnacht et al,

1997; Peeper et al, 1997). Ras increases cyclin D1 activity, through transcriptional elevation and

protein stabilization and also participates in the assembly of cyclin D1 with CDK4 or 6 (Aktas et al,

1997; Lavoie et al, 1996; Winston et al, 1996). The indispensable role of cyclin D1 in the Ras-

mediated cell proliferation is demonstrated by the resistance to H-Ras induced mammary tumor

development in mice that lack cyclin D1 (Yu et al, 2001).

1.2.3 Involvement in breast cancer

The involvement of insulin-like growth factor-I receptor (IGF-IR) in breast cancer is very well

established. Elevated expression of IGF-IR in breast carcinoma cells lines and the potent mitogenic

effect of IGF-I (more than IGF-II) was initially noted by Cullen et al in 1990 (Cullen et al, 1990) and

the area has received considerable attention with numerous subsequent studies in last two decades.

The critical role of IGF-IR in breast cancer was consistently confirmed by knockdown studies such

as with antibodies or siRNA, where inhibition of IGF-IR effectively abrogated cell proliferation in

different breast cancer cell lines. IGF-IR was also required for anchorage-independent growth, and

ectopic expression of dominant negative IGF-IR lacking the C-terminus showed that the C-terminal

region was vital for transformation and tumorigenesis (Surmacz et al, 1998). IGF-IR has been also

shown to be elevated in estrogen receptor (ER)-positive breast cancer cells and that its level is

correlative with the ER and PR status in primary tumors (Peyrat et al, 1988).

The importance of IGF-IR in the breast cancer mortality was further heightened when Kim et

al demonstrated that overexpression of IGF-IR in immortalized normal human mammary epithelial

cells MCF10A was sufficient to cause epithelial-to-mesenchymal transition (EMT) (Kim et al, 2007).

EMT of primary cancer cells is implicated in the tissue invasion and metastasis, which is the lethal

factor in most cancers. Stable overexpression of constitutively active CD8-IGF-IR fusion protein in

MCF10A not only led to full transformation, but also conferred migratory ability to the cells,

Page 20: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

10

drastically downregulating epithelial adherens junctions and increasing the transcription of Snail

(Kim et al, 2007). The study done by Kim et al, therefore, made a hallmark that IGF-IR is involved

in virtually all stages of breast cancer progression.

In other recent studies, transgenic overexpression of IGF-IR resulted in perturbed mammary

ductal morphogenesis such as marked hyperplasia and impaired ductal elongation, and accelerated

tumor formation, with mice developing papable tumors in approximately 8-11 weeks of age (Carboni

et al, 2005; Jones et al, 2006). Interestingly, histological and cytokeratin immunohistochemical

analyses indicated that some of IGF-IR-induced mouse mammary tumors exhibited features of

ErbB2/Ras tumors, while more advanced tumors shared characteristics of Wnt-oncogene tumors

(Jones et al, 2006). In general, ErbB2/Ras pathway leads to solid, nodular tumors with little stroma

and metaplastic myoepithelial cells, while Wnt oncogene forms ductular tumor with abundant stroma

and lymphocytic infiltration (Rosner et al, 2002). However, IGF-IR did not specifically fit under

those categories, and this may support that IGF-IR can cross-talk with multiple cellular signaling

cascades to cause rapid transformation and hyper-proliferation. As well as in breast cancer, IGF-IR is

involved in multiple types of cancers (Tao et al, 2007) due to its broad spectrum of signaling

pathway activation and gene regulation (Dupont et al, 2003). For example, Creighton and colleagues

identified more than 800 genes whose expression was influenced by IGF-I (Creighton et al, 2008).

Importantly, IGF-IR has also been implicated in clinical resistance to trastuzumab

(Herceptin) in breast cancer. Overexpression of IGF-IR has been shown to antagonize downstream

effects of trastuzumab on G1 cyclins and Cdk2 inhibitor p27Kip1 (Lu et al, 2001) and unique

heterodimerization and crosstalk of IGF-IR with HER-2 has been suggested to contribute to

trastuzumab-resistance (Nahta et al, 2005; Knowlden et al, 2005). This called for a closer

examination of IGF-IR signaling as a potential therapeutic target (reviewed in Tao et al, 2007 and

Chitnis et al, 2008).

1.3 Pleckstrin-homology domain Interacting Protein (PHIP)

1.3.1 Discovery and characterization

PHIP, or pleckstrin-homology domain interacting protein, was first cloned and characterized as a

902-amino acid (aa)-long variant 9 (Farhang-Fallah et al, 2000), which was involved in mitogenesis

and insulin receptor-mediated GLUT4 translocation in myoblasts (Farhang-Fallah et al, 2002). It was

Page 21: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

11

discovered as a novel strong binding partner of IRS-1 through yeast two hybrid screening of murine

10.5-day embryonic cDNA library using the PH domain from rat IRS-1 as bait, followed by

screening of mouse thymus and human fetal brain cDNA libraries. The 902-aa protein, though not

sharing significant sequence homology with any previously known proteins, was found to harbor two

characteristic bromodomain (BD) structural motifs that were most similar (44% identity, 61%

homology) to that of mouse CREB-binding protein (CBP) (Farhang-Fallah et al, 2000). This

suggested that the BD motif in the PHIP family might confer them the function of transcriptional co-

activators.

NCBI mouse database (NCBI Build 36; mm8) identifies at least four variants of PHIP, from

alternative splicing and multiple translational initiation start sites (Figure 4) (Podcheko et al, 2007).

My study focuses on the larger 1,821-aa, 206kDa isoform of PHIP (denoted as PHIP1), which

contains two additional WD repeat domains to the smaller variant 9. The human version of PHIP1

shares 96% primary amino acid sequence identity with the mouse counterpart. Based on the fact that

the smaller variant of PHIP was involved in the insulin signaling pathway, PHIP1 was identified and

characterized in the context of pancreatic β-cell function, as it was the most prominently expressed

isoform in the whole-cell lysates of the mouse islets and insuloma cell lines MIN6 and INS-1 cells

through immunoblot analysis (Podcheko et al, 2007).

Immunohistochemical and immunoblot analyses in pancreatic β-cells showed that PHIP1 was

confined to the nucleus. (Podcheko et al, 2007), though interestingly the 902-aa isoform of PHIP was

shown to selectively interact with IRS1 and IRS2 PH domains as a downstream target (Farhang-

Fallah et al, 2000). During the functional analyses, adenoviral PHIP1 overexpression stimulated

insulin-like growth factor 1(IGF-I) -dependent and -independent proliferation of INS-1 cells through

PKB phosphorylation and transcriptional regulation of cyclin D2 expression, while PHIP1 knock-

down inhibited IRS-2-dependent mitogenesis (Podcheko et al, 2007). The fact that PHIP1 alone

could drive the cyclin D2 transcription was demonstrated by promoter-reporter gene analysis in INS-

1 cells transiently transfected with firefly luciferase gene downstream of the promoter fragment of

mouse cyclin D2. These results consolidated PHIP1 as a novel downstream effector of IGF-IR

signaling pathway with crucial functions in the regulation of cellular growth and apoptosis.

Page 22: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

12

Figure 4. Different variants of PHIP.

The originally identified isoform was the 902-aa variant 9, and the one concerned in this project is the human

PHIP1, which is the WD40-containing 206 kDa protein on the very top. Adapted from (Podcheko et al, 2007).

1.3.2 PHIP1 structure and functional domains

As revealed by sequence analysis for conserved protein motifs by PFAM database, the 1821-aa

PHIP1 was found to contain eight WD40 repeats (residues 171 to 211, 214 to 253, 256 to 299, 310 to

349, 354 to 393, 408 to 452, 455 to 495, and 498 to 542) on its N-terminus and two bromodomains

(BD) (residues 1158 to 1261 and 1318 to 1423) on the C-terminus, representing a new member of the

WD40-repeat (WDR) protein family. PHIP1 additionally contains two putative nuclear localization

signals (NLSs), as identified by the PredictNLS program, and the human PHIP is 96% identical in

sequence with the predicted mouse PHIP isoform (Podcheko et al, 2007). The full-length hPHIP1

was first cloned in our laboratory by reverse-transcriptase PCR from MCF-7 cells to produce 5.53 kb

cDNA that correspond to the complete open reading frame (ORF) of the predicted genome sequence.

1.3.2.1 WD40-repeat domain

WD40-repeat domains consist of 40 to 60 amino acids, characteristically starting with glycine-

histidine (GH) dipeptide 11 to 24 residues from the N-terminus and ending with a tryptophan-

aspartic acid (WD) at the C-terminus. The WD40 domains are usually referred to as ‘WD-repeats’, as

most proteins characterized by this motif contain a cluster of 4 or more tandem repeats. The motif

was first identified in 1986 in the transducin β subunit by Fong et al (Fong et al, 1986) and the

protein family has been expanding with research and discovery. The WD-repeat proteins are

diversified by sequence differences in the two variable regions within the WD-repeat as well as by

Page 23: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

13

the number of structural repeats that are present. The common functionality of the family is to serve

as a scaffold to coordinate multi-protein complex formations and facilitate protein-protein interaction

(Li & Roberts, 2001; Neer et al, 1994). Examples of the WD-40 repeat proteins include β subunit of

heterotrimeric G proteins, Apaf-1, most of α and β-COP proteins, chromatin assembly factor-1

(CAF-1), and the substrate specific adaptor in the CUL4-DDB1 E3 ubiquitin ligase complex (which

is further discussed in the next section). The β subunit of the G protein has been the classically

studied member of the protein group, and the only protein for which the crystal structure has been

elucidated (Sondek et al, 1996). Given the conservation of the domain, WD-repeat proteins are

thought to probably take a shape of a circularized β-propeller, with each repeat comprising a four-

stranded antiparallel β sheet deduced from the β subunit of the G protein. As represented by the

diverse members of the group, WD40-repeat containing proteins serve a wide range of biological

regulatory functions, such as signal transduction, cell cycle regulation, RNA processing, chromatin

and cytoskeleton assembly (Li & Roberts, 2001). However, there are many areas that need

investigation to better understand this protein family. For example, there are a number of WD-repeat

proteins, with PHIP1 also named as WDR11, that currently have no known function registered in the

Human Gene Nomenclature Database.

1.3.2.2 Bromodomains

The bromodomain was initially discovered by Tamkun et al in Drosophila melanogaster Brahma

protein, which is closely related to the SWI2/SNF2 family of transcriptional co-activators (Tamkun

et al, 1992). Bromodomain was also identified in a number of eukaryotic transcriptional factors and

co-activators, including GCN5 and p300/CBP histone acetyltransferases (HAT) (Haynes et al, 1992).

The fold of the bromodomain contains four α helices to form a four-helix bundle arranged in

anti-parallel twist. The double bromodomain, such as in TAFII250 and in PHIP1, the two domains

are held by the electrostatic interface by the interaction between the glutamate and lysine residues in

the interface of the two domains, and the two loops on the opposite side to the N-terminal and C-

terminal ends form a hydrophobic pocket. Interestingly, the presence of both conserved amino acids

as well as the variability from residue insertions and deletions occur in the loop region, hinting that

the region is likely involved in substrate specificity (Marmorstein and Berger, 2001).

The structural analysis of the human P300/CBP-associated factor (PCAF) by Dhalluin and

co-workers (Dhalluin et al, 1999) was the first study that gave insight into the function of the

bromodomain. Because GCN5/PCAF was a member of the histone acetylase (HAT) protein family,

the authors tested whether the bromodomain targeted the acetyl-lysine residues on histone H4 in

Page 24: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

14

NMR titration experiments and showed that the interaction was specific (Dhalluin et al, 1999).

Further studies by Jacobson et al, which analyzed functional and structural characteristics of

TAFII250, indicated that fully acetylated histone H4 peptide interacted with the double

bromodomain with a stoichiometric ratio of 1:1, as determined by isothermal titration calorimetry,

while no binding was detected with the non-acetylated peptide (Jacobson et al, 2000). As

bromodomains can bind to acetylated histone lysine residues, they may promote interaction of the

HATs with chromatin to begin transcriptional activation (reviewed in Marmorstein and Berger, 2001;

Loyola and Almouzni, 2004). In a model of bromodomain function, Marmorstein and Berger

proposed that factor acetyltransferase (FAT)-mediated acetylation of a transcriptional activator and

facilitation of its DNA binding recruits the SWI/SNF remodeling and subsequently the HAT domain

complex through sequential interactions between the bromodomains. This, in turn, leads to the

acetylation of histones within the promoter region and ultimately the recruitment of transcription-

initiation complex, such as TATA box binding protein (TBP), TAF250 and RNA polymerase II, thus

increasing the gene transcription and activation (Marmorstein and Berger, 2001). Therefore, the

presence of these highly functional domains supports that PHIP is a novel transcriptional co-activator

with important roles in many aspects of cell life-cycle.

1.3.3 PHIP1 as DCAF14

WDR proteins are becoming an emerging topic of interest within the scientific community. There

are still a number of WDR proteins less well understood and some have been newly classified as

DCAFs, or DDB1 and CUL4-associated factors, suggested to be putative substrate receptors for

CUL4-DDB1ubiquitin ligase complex (Lee and Zhou, 2007). PHIP1 has been recently discovered as

a subunit of the CUL4-DDB1 E3 ubiquitin ligase, known in alias as DCAF14, which suggests of

more diverse function for PHIP1 in the control of regulatory and signaling proteins.

1.3.3.1 Ubiquitin-Proteasome Pathway (UPP) and CUL4-DDB1 Ubiquitin Ligase

The ubiquitin-proteasome pathway (UPP) is a protein-degradative mechanism whereby 80-90% of

the intracellular proteins are proteolyzed after being targeted for ubiquitination. Ubiquitin (Ub) is a

76-amino acid polypeptide, which post-translationally modifies the substrate by covalent transfer,

which involves conjugation of the Ub to the ε-amino group of the lysine residue via the C-terminus

to form the isopeptide bond. The conjugation takes place in three consecutive steps, involving three

different enzymes, E1, E2 and E3. E1 is the ubiquitin-activating enzyme, which adds the Ub onto

itself by a ATP-requiring process. E2, the ubiquitin-conjugating enzyme, transfers this ubiquitin to a

Page 25: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

15

cysteine residue of the enzyme from E1. Ubiquitin ligase, E3, finally transfers the bound Ub

molecule on E2 to the amino –NH2 side chain of the lysine residue in the target protein, forming a

peptide bond (Lafarga et al, 2002). These three steps are repeated numerous times and the

polyubiquitin chain is recognized and degraded by the proteasome. The specificity of protein

turnover is provided in part by the E3 enzymes. This is exemplified by the fact that there is only one

E1, several E2s, but hundreds of E3 enzymes (O’Connell and Harper, 2005). Non-degradative

function of the UPP include monoubiquitination, which plays a role in protein-protein interaction,

protein trafficking and localization, facilitation of endocytic internalization, regulation of plasma

membrane protein activity, and in histone coding, such as in the methylation of H3 and gene

silencing in yeast (Wang et al, 2006).

The cullin-RING finger ligases (CRL) are members of the RING-finger domain-containing

class of ubiquitin ligases. The E3 ubiquitin ligase that utilizes the cullin subunit as a scaffold in a

multiprotein complex was first reported in 1997 (Skowyra et al, 1997; Feldman et al, 1997). There

are seven cullins in vertebrates (CUL1, 2, 3, 4A, 4B, 5, and 7), and while the different cullins form

variations of E3 ubiquitin ligase (Petroski and Deshaies, 2005; O’Connell and Harper, 2007;

Figure 5), the insight into the spatial configuration of the largest family of E3 ubiquitin ligases was

derived from crystal structure of the archetypical Cul1-Rbx1-Skp1-F boxSkp2 SCF ubiquitin ligase

complex by Zheng and co-workers (2002). In this CRL, Skp1 acts as the adaptor and F-box proteins

as the substrate receptors. Cullins have an extended and rigid structure that contains a long stalk and

a globular domain. The N-terminal region of cullin binds with the substrate receptors through

specific adaptor molecules and the C-terminal, globular domain interacts with the RING finger

protein Rbx1/ROC1/Hrt1, which forms a two-subunit catalytic core and provides the site of E2

ubiquitin-conjugating enzyme recruitment (Zheng et al, 2002).

There is evidence that PHIP1 is part of the CUL4 E3 ligase complex, which is composed of

DDB1 (damaged DNA binding protein), as the adaptor and DCAF as the substrate receptor (Figure

5). DDB1 contains 21 WD40-like repeats which folds into three β-propeller domains, where the

double-propeller pocket participates in the substrate presentation and the third domain binds to the

cullin (Li et al, 2006). In 2006, Angers and co-workers reported on structural and proteomic analyses

of DDB1-CUL4A-ROC1 complex bound to the simian virus 5 V protein (SV5V), which

ubiquitinates and degrades STAT1 to inhibit the host antiviral response, in order to elucidate the

molecular architecture of the enzyme complex as well as to identify its protein interaction networks.

Interestingly, PHIP1 was identified as one of the substrate receptors of CUL4-DDB1 complex during

the tandem-affinity analyses. As CUL4-DDB1 E3 ligases have been reported to be involved in

Page 26: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

16

histone ubiquitination, methylation and responses to DNA damage (Wang et al, 2006; Higa et al,

2006), this enthralling new discovery potentiates the role of PHIP in cell survival and function.

Figure 5. E3 ubiquitin ligases of different Cullin proteins.

Substrates become poly-ubiquitinated by the E3 ubiquitin ligases and targeted for protein degradation by 26S

proteasome. The E3 ubiquitin ligases have conserved molecular assembly composed of the core Cullin-Rbx1 and a

determinant of specificity (Skp1-F-box, BTB, EloBC/BC/SOCS-box, DCAF or FBW8 depending on the Cullin

type). Adapted from (O’Connell and Harper, 2007).

1.3.3.2 Involvement of Cul4 in breast cancer

In support of role of cullin-RING ubiquitin ligase in breast cancer, cul-4, the human homologue for

the Caenorhabditis elegans cul-4 gene, was found to be amplified and overexpressed in primary

breast cancer by Chen and colleagues in 1998. In the study, cul-4 was amplified in 16% of untreated

primary breast cancers and overexpressed in 47% of the cases, as determined by RNA in situ

hybridization. Likewise, in 14 breast cancer cells analyzed, the authors showed that cul-4 gene was

both amplified and overexpressed in only 3 cell lines, but overexpressed without amplification in the

majority, which indicated that cul-4 could accumulate via other mechanisms and the upregulation

Page 27: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

17

may play a role in tumorigenesis (Chen et al, 1998). In a recent study by Schindl et al,

overexpression of cul-4 gene has been related to poor outcome in lymph node-negative invasive

breast cancer through immunohistochemistry analyses of 167 human breast cancer specimens, where

moderate to high levels of cul-4 were observed in over 50% of the cases. The data provided further

evidence that CUL-4 may participate in tumor development and progression (Schindl et al, 2007).

1.4 Hypothesis and Rationale

The study of PHIP1 in islet β-cell function and survival demonstrated that PHIP1 is a positive

regulator of cellular growth and that it potentiates the mitogenic effect of IGF-I. Additionally,

adenoviral overexpression of PHIP1 in NIH3T3 cells promoted proliferation, indicating a growth-

promoting ability of PHIP1. Consistent with this finding, preliminary analysis suggested that PHIP1

protein levels were markedly elevated in a number of human breast cancer cell lines compared to

normal human mammary epithelial cell line (HMEC). Additionally, one of the MMTV-PHIP1

transgenic mouse lines established in the laboratory developed mammary hyperplasia within 8

months of age upon multiple pregnancy. Given this supportive preliminary information and the well-

established implication of IGF-IR in breast cancer, we hypothesized that PHIP1 will play an

important role in mammary gland turmorigenesis and oncogenic transformation. We further aimed to

study PHIP1 action in molecular signaling that drives breast cancer tumorigenesis.

Page 28: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

18

Chapter 2. Materials and Methods

2.1 Reagents and antibodies

Polyclonal rabbit antibody against the bromodomain of human PHIP was generated in our laboratory

using glutathione S-transferase (GST)-PHIP fusion protein corresponding to residues 1548-1821 of

mouse PHIP1 injected into rabbits and purifying the rabbit sera through a GST affinity column, as

previously described (Podcheko et al, 2007). Mouse monoclonal antibody against the hemmaglutinin

(HA) epitope (Cat. # sc-7392) was purchased from Santa Cruz Biotechnology (Santa Cruz, CA). The

rabbit polyclonal antibody against tubulin (Cat. # 2148) was purchased from Cell Signaling

Technology (Danvers, MA). The mouse monoclonal antibody specific for Cyclin D2 (Cat. # MS-

213-PO) was purchased from Thermo Fisher Scientific (Rockford, IL) and mouse antibody against β-

actin (Cat.# A3853) from Sigma Aldrich (St. Louis, MO). The secondary antibodies, both the goat

anti-mouse HRP-conjugate (Cat. # 170-5047) and goat anti-rabbit HRP-conjugate (Cat. # 170-5046)

were purchased from Biorad Life Science (Hercules, CA). The rabbit ExactaCruz was purchased

from Santa Cruz (Cat. # sc-45043). The GIPZ and TRIPZ shRNA lentiviral constructs expressing a

scrambled control or shRNA against human PHIP1 (V2THS_174590, V2THS_174591,

V2THS_174592) were obtained from Open Biosystems, Thermo Fisher Scientific (Huntsville, AL).

The HA-PHIP1-MSCV retroviral vector was previously generated in our laboratory by a post-

doctoral fellow, where the full-length human PHIP1 cDNA with a triple HA tag on the N-terminus

was cloned into the Murine Stem Cell Virus (MSCV) backbone from Clontech. For transfection

reagents, PEI was purchased from Sigma (Cat. # 408727) and Lipofectamine 2000 was purchased

from Invitrogen (Cat. # 11668-027). MG132 proteasomal inhibitor was purchased from Calbiochem

(Cat. # 474790) as part of the Proteasome Inhibitor Set 1 (Calbiochem, Cat. # 539164).

2.2 Cell culture

MCF10A cells stably overexpressing constitutively active CD8-IGF-IR (‘MCF10A-IGFR’) or IRS-1

or IRS-2 (MCF10A-IRS1 or 2) were kindly provided by Dr. Adrian V. Lee at Baylor College of

Medicine. Parental MCF10A or MCF10A-derived stables were all maintained in DMEM-F12

(Invitrogen, Cat. # 11330-032) containing 5% horse serum (Gibco, Cat. # 16050-122), 1%

penicillin/streptomycin (BioWhittaker, Cat. # 17-602E), 20 ng/mL EGF (Peprotech, Cat. # AF-100-

15), 10 μg/mL insulin (Sigma, I-1882), 0.5 μg/mL hydrocortisone (Sigma, Cat. # H-0888) and 100

ng/mL cholera toxin (Sigma, Cat. # C-8052). For MCF10A-IGFR cell line, the medium additionally

Page 29: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

19

contained 5 μg/mL puromycin (Sigma, Cat. # P-8833) to maintain the selective pressure for the

stable expression of the CD8-IGF-IR transgene. For subculturing MCF10A, cells were washed once

with 1x PBS and trypsinized with 1 mL of 0.05% trypsin in 10 cm dish and incubated for 20 minutes

in 37 oC. MCF10A, MCF-7, and NIH/3T3 were obtained from American Type Culture Collection

(ATCC) and cells within passage number 10 were used in experiments. 293T, MDA-MB-231 and

NIH/3T3 cells were cultured in DMEM (Wisent, Cat. # 319-005-CL) supplemented with10% heat-

inactivated fetal bovine serum (FBS; Wisent, Cat. # 090-150) and 1% penicillin/streptomycin. MCF7

cells were grown in DMEM/F12, 10% FBS and 1% antibiotics.

For cell cryopreservation, freezing media containing 90% FBS and 10% DMSO (Sigma, Cat.

# D-2650) was generally used, except for MCF10A, which was prepared by 70% MCF10A growth

medium, 20% horse serum, and 10% DMSO. Approximately 2 million to 5 million cells in 1 mL of

freezing media were stored in cryotubes.

2.3 Virus production and transduction

The 293T cell line was used for both lentivirus and retrovirus production. The cells were plated at

70% confluency the day before transfection on 100 mm culture dishes. For lentivirus production, 9

μg of GIPZ or TRIPZ plasmid (Open Biosystems), 9 μg psPAX.2 (packaging) and 4.5 μg of MD2.G

(envelope) plasmids were added to 5 mLs of serum-free, antibiotic-free DMEM (Wisent) and

incubated at room temperature (RT) for 5 minutes, after which 20 μL of 2 mg/mL polyethylenimine

(PEI; Sigma) was added and allowed to complex with the plasmids for further 20 minutes. For

doxycyline-induced protein knockdown in MCF10A-IGFR or MDA-MB-231 TRIPZ stables,

lentivirally infected and drug selected cells were incubated in 10% FBS DMEM containing 1 µg/mL

doxycyline (Sigma, Cat. # D-9891). The medium was refreshed every 24 hours for 3 days before

being lysed for Western blot analyses.

Similarly, for the retrovirus production, 10 μg MSCV and 10 μg of pCl-Ampho (RetroMax)

plasmids were transfected into 293T cells. 6 hours after transfection, 5 mLs of DMEM containing

20% FBS (no antibiotics) were added per culture dish and incubated at 37 oC and 5% CO2 for 48

hours. The spent medium containing lenti- or retrovirus was filtered through 0.45 μm pore (made of

mixed cellulose esters; Milllipore, Cat. # SLHA033SS), aliquoted, and stored in -80 oC. Before being

used, the virus was rapidly thawed in a 37oC waterbath.

For transduction, the cells to be infected were seeded the day before at 40-60% confluency in

60 mm dishes before for lentiviral infection or at 100,000 cells per well on 6-well plates for retroviral

infection. The transduction mixture comprised a 1:1 ratio of the viral supernatant and the growth

Page 30: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

20

medium, and 4-8 μg/mL polybrene (= hexadimethrine bromide; Sigma, Cat. # H-9268). The final

volumes of the mixture were 3 mLs for 60 mm dishes and 1 mL for 6-well plates. For retroviral

infection, cell 6-well plates were spun at 1,800 rpm for 45 minutes at RT. The media was changed

the next morning to fresh growth medium and drug selection was started 48 hours after. The

concentrations of antibiotics used were 2 μg/mL for puromycin (Sigma, Cat. # P-8833) and 800

μg/mL for G418 (Bioshop, Cat. # GEN418.5). For puromycin, the period of selection was 5-7 days,

while for G418, it was minimum 10 days, or until all of the non-infected control cells died.

2.4 siRNA transfection for transient knockdown experiments

For MDA-MB-231, 100,000 to 200,000 cells were plated per well on a 6-well plate 24 hours before

the transfection. SMARTpool PHIP-siRNA (Dharmacon, Cat. # M-019291-00) and Scrambled-

siRNA (Dharmacon, Cat. # D-001210-02) were resuspended in the siRNA buffer (Dharmacon Cat. #

B-002000-UB-015) at the final concentration of 20 μM. For each well, the cells were washed twice

with serum-free media (SFM) before transfection. 100 nM of siRNA and 0.8% of Lipofectamine

2000 (Invitrogen, Cat. # 11668-027) were added to 1 mL SFM and incubated at RT for 20 minutes to

allow complex formation before being added to the cells. The transfection medium was aspirated 6

hours later and changed to growth medium. The cells were analyzed after being incubated for

minimum 48 hours. For transfection of larger number of cells, the volumes were adjusted by

appropriate proportions.

For transfection of MCF10A-IGFR cells for the proliferation assay, 50,000 cells were seeded

per well on 12-well plates the day before the transfection. 100 nM of siRNA and 0.4%

Lipofectamine2000 were mixed in serum-free DMEM/F12 for 25 minutes and added onto the cells.

During the 25 minutes, the attached cells were washed twice with plain DMEM/F12 with no other

supplements. The transfection mixture was incubated with cells overnight (16 hours) and aspirated

and replaced with MCF10A starvation medium containing 0.5% horse serum, 1%

penicillin/streptomycin, 20 ng/mL EGF, 10 μg/mL insulin, 0.5 μg/mL hydrocortisone and 100 ng/mL

cholera toxin the next morning. The day of the transfection was designated as Day 0 and the serum-

reduced MCF10A media was replaced every 2 days until Day 8. The cells were counted on Days 2, 4,

6 and 8 with Beckman Coulter as explained further in Section 2.5.

For MCF-7 cells, reverse transfection method was used to transiently knockdown PHIP. For

transfections in 6-well plates, 200 nM of siRNA was mixed with 0.5% Lipofectamin 2000 by first

separately incubating each of them in 250 μL of serum-free DMEM/F12 and further incubating for

20 minutes at RT after combining the two components to make a final volume of 0.5 mL (per well;

Page 31: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

21

final siRNA concentration 100 nM). This was mixed with 1 mL of freshly trypsinized and

resuspended MCF7 cells at concentration of 200,000 cells/mL in the growth medium (10% FBS in

DMEM/F12 but without antibiotics) and incubated overnight at 37 °C 5% CO2. The transfection

media was aspirated and changed to growth medium the next day. For the treatment with IGF-I (Cell

Signaling, Cat. # 8917), 10,000 cells were reverse transfected in growth medium overnight in 96-well

plate and washed once with plain DMEM/F12 the next morning. The cells were starved in SFM for

24 hours, after which cells were treated with 100 ng/mL IGF-I in DMEM/F12 for 48 hours and

measured for growth using the MTT assay as mentioned in the next Section.

2.5 Cell proliferation assays

For cell counting on the hemocytometer, cells were trypsinized with 0.05% trypsin (with EDTA) and

blocked with 20% FBS. The volumes were adjusted to maximize the cell concentration per mL. 10

μL of cell suspension was mixed with 10 μL of trypan blue and loaded onto the hemocytometer. The

four 16-squares at each corner were counted and averaged. The total cell number was calculated by:

[averaged cell number x 2 x 104 x Volume of cell resuspension]. When counting cells on the

Beckman Coulter Counter Z2 (Beckman), the cells were prepared similarly and the lower threshold

used was 6.0 μm. Usually, 50 – 100 μL of cell suspension was diluted in 10 mL of isotone solution

before subjecting the measurement. The proliferation assay using the 3-(4,5-dimethylthiazol-2-yl)-

2,5-diphenyl tetrazolium bromide (MTT) reagent (Sigma, Cat. # M-2128) was performed by adding

20 μL of 5 mg/mL filter-sterilized MTT dissolved in PBS per 100 μL cell culture media (overall

MTT concentration 1 mg/mL) on a 96-well plate and incubating at 37 °C 5% CO2 for 3 hours and 30

minutes. The media was carefully removed and the purple formazan precipitates were dissolved in

150 μL MTT solvent (4 mM HCl, 0.1% NP40 in isopropanol) per well. The plate was then covered

with aluminium foil and agitated on an orbital shaker for 15 minutes, after which the absorbance was

read at 595 nm on the spectrophotometer.

2.6 Soft agar anchorage-independent colony formation assay

For the soft agar assay, which looks at anchorage-independent colony formation, 10,000 or 20,000

MCF10A cells infected with AdPHIP virus the day before were suspended in MCF10A growth

media containing 0.4% agarose, low-melting point (Sigma, Cat. # A-9414) plated on top of a basal

layer with 1% agarose in complete MCF10A media. For the specific procedure, 3% agarose in PBS

was autoclaved and re-dissolved in a microwave prior to use. For the basal layer, the agarose was

Page 32: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

22

mixed 1:3 with MCF10A complete medium and 2 mL of 1% agarose was put per well of a 6-well

plate. For the top layer, one set of triplicates was prepared by mixing 1.08 mL of 3% agarose , 2.92

mL MCF10A growth media and 4 mL of cell suspension (40,000 or 80,000 cells total) and then

plated on top of the basal agar at 2 mL per well. The wells were maintained by adding 0.3-0.5 mL of

media every 3-4 days. Colonies were stained with MTT (2 mg/ml in PBS) by incubation for 4 h at 37

°C. Cells were photographed with Nikon ECLIPSE TE2000-U microscope and SimplePCI6

software. Regarding the number of MOI used during the adenovirus infection, viral titre was measured by

immunohistochemistry for AdGFP using AdEasy Viral Titre Kit (Agilent, Cat. # 972500), which detects

only live viral particles, but for AdPHIP, values of titre derived from measuring the absorbance was used,

which does not differentiate between live and dead particles (where OD260nm = 1 x 1012 viral particle/mL).

The actual degree of GFP fluorescence was used to determine the appropriate volume of virus used in the

experiment, which in the study was MOI of 10 for AdGFP and MOI of 650 for AdPHIP.

For the soft agar assay for the MCF10A-PHIP retrovirally infected stable cell line, very

similar procedure was used with minor modification. For the bottom layer, SeaPlaque low-melting

agarose (Lonza, Cat. # 50101) was dissolved in 10 mL 1x PBS to concentrations of 2.1% by heating

in the microwave for approximately 2 minutes. The agarose solutions were quickly mixed with pre-

warmed MCF10A growth medium (in 40 °C waterbath) in 1:2 and plated on to 6-well plates in

minimum of triplicates with 1.5 mL mixture per well. The gel was allowed to solidify in the room

temperature for 30 minutes, during which time the top layer was prepared. Similar to the bottom

layer, 1.7% of agarose was heated in 1x PBS and cooled to 40 °C before being mixed with MCF10A

medium containing 20,000 cells (for triplicates, or (n+1) x 5,000 for n replicates) and plated on top of

the bottom layer in volumes of 1.5 mL per well. 0.5 mL of MCF10A growth medium was added

drop-wise every 3 to 4 days for 14 days. The colonies were stained with MTT as described above.

2.7 SDS-PAGE Electrophoresis and Western Blotting

Cells were lysed in hot 6x SDS reducing buffer (150 mM Tris-HCl pH 6.8, 30% glycerol, 10% SDS,

3% β-mercaptoethanol, and 0.002% bromophenol blue) and scraped off the culture dishes or plates

with a scraper. The lysates were boiled for minimum five minutes at 100 °C and measured for total

protein concentration by the Bradford assay (Sigma, Cat. # B-6916). 30 to 50 μg of cell lysates were

loaded and run on 10 to 12% polyacrylamide gels in 1x SDS running buffer (25 mM Tris-HCl, 190

mM glycine, 0.1% SDS pH 8.6) at 150V for 60 minutes to separate the proteins by size alongside a

pre-stained protein marker (Fermentas, Cat. # SM0671). After the size fractionation by the SDS-

PAGE electrophoresis, the proteins were transferred on to polyvinylidene fluoride (PVDF)

Page 33: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

23

membranes (Millipore, Cat. # IPVH00010) at 110V for 90 minutes in transfer buffer (25mM Tris-

HCl, 192mM glycine, 20% (v/v) methanol, pH 8.3).

Membranes were then blocked to reduce non-specific signals for 30 to 60 minutes on a

rocker with 5% skim milk powder dissolved in 1x TBST (50mM Tris-Cl pH 7.5, 150 mM NaCl,

0.05% Tween-20). After several rinses with 1x TBST, the membranes were cut at appropriate marker

lanes and incubated with primary antibodies at 4°C for 20 hours (overnight). The primary antibodies

were diluted in 5% bovine serum albumin (BSA; Bioshop, Cat. # ALB001.500) in 1x TBST

containing preservative 0.5% NaN3.

The membranes were washed three times with 1x TBST for 10 minutes per wash and

incubated with anti-mouse or anti-rabbit HRP-linked secondary antibody for 1 hour at RT on a rocker.

The secondary antibodies were diluted in 5% skim milk in 1x TBST. For anti-rabbit HRP secondary

antibody (Biorad, Cat. # 170-5046), dilution of 1:10,000 was used in 10 mL of 5% skim milk in 1x

TBST and anti-mouse HRP-conjugate (Biorad, Cat. # 170-5047) was diluted to 1:2,000 in 5% skim

milk in 1x TBST for all the experiments. Following three more washes for 10 minutes each with 1x

TBST, the membranes were subjected to ECL solution (Thermo Scientific, Cat. # PI34080/DDEL)

for 60 seconds before being developed on autoradiography films (Denville Scientific, Cat. # E3018).

Antibody Company Catalog # Source WB Dilution Protein size PHIP (H-302) Santa Cruz sc-68354 Rabbit 1 in 500 206kDa PHIP (CT-7) Rozakis lab - Rabbit 1 in 10,000 206kDa HA-probe (F-7) Santa Cruz sc-7392 Mouse 1 in 500 depends Phospho-p44/42 MAPK Cell Signalling #9101S Rabbit 1 in 1,000 42, 44kDa Phospho-Akt Cell Signalling #4060S Rabbit 1 in 2,000 60kDa Cyclin D1 Thermo RB-212-PO Rabbit 1 in 500 36kDa Cyclin D2 Thermo MS-213-PO Mouse 1 in 500 34kDa β-catenin BD 610153 Mouse 1 in 1,000 92kDa α/β-Tubulin Cell Signalling #2148 Rabbit 1 in 1,000 55kDa IGF-IR b (C-20) Santa Cruz sc-713 Rabbit 1 in 1,000 97kDa, 200kDa E-cadherin BD 610181 Mouse 1 in 5,000 120kDa p53 (FL-393) Santa Cruz sc-6243 Rabbit 1 in 1,000 53kDa

Table 1. Table of antibodies used in the study with their dilutions

2.8 Immunoprecipitations

HA-PHIP1 immunoprecipitations were performed on 293T cells transfected with MSCV-HA-PHIP

or MIEV (MSCV-IRES-EGFP vector) control plasmid on 10 cm culture dishes at least 48 hours prior

Page 34: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

24

to the experiment. The HA-PHIP protein was first enriched through nuclear fractionation, thereby

reducing the cytoplasmic content and decreasing the potential non-specific signals. The cells were

washed twice with ice cold 1x PBS and the buffer was aspirated as much as possible before adding

500 µL of Buffer A (10 mM HEPES pH 7.9, 1.5 mM MgCl2, 10 mM KCL, 0.5 M DTT) containing

protease inhibitors (1x Protease Inhibitor Cocktail with EDTA from Bioshop Cat. # PIC001.1, 1 mM

NaF, 1 mM Na3VO4 and 2 mM PMSF) per 10 cm dish and scraping the cells off the plate on a pan of

ice. The cell lysates were incubated on ice for 15 minutes and added 10% stock solution of NP-40 to

a final concentration of 0.5%, after which they were vortexed for 10 second each. The nuclei were

pelleted by centrifugation at 6500 rpm for 20 seconds on a microcentrifuge. The supernatant, which

contains the cytoplasmic proteins, were usually transferred to fresh eppendorf tubes in case of a

future analysis.

The nuclear pellet was lyzed in 1x RIPA buffer (50 mM Tris-HCl pH 7.4, 1% NP-40 (v/v),

0.25% Na-deoxycholate (w/v), 150 mM NaCl, 2 mM EDTA and 0.1% SDS) containing protease

inhibitors (1x Protease inhibitor cocktail, 1 mM NaF, 1mM Na3VO4 and 2mM PMSF) added

immediately before use. To fully release the nucleic content, the nuclear lysates were sonicated twice

for 10 seconds at Power Level 3 and centrifuged at 10,000 rpm for 10 minutes at 4 °C. The

supernantant was transferred to fresh eppendorf tubes while the cellular debris was discarded. The

nuclear protein lysate in the 1x RIPA buffer was subjected to preclearing with 50 µL of protein-A

50% slurry (Millipore, Cat. # 16-156) per 1 mL for 30 minutes at 4 °C with gentle rocking.

Meanwhile, the IP-antibody-IP-matrix complex was prepared by adding 50 µL of suspended (25%

v/v) IP matrix and 2 µg of IP antibody (10 µL of p53, Santa Cruz Cat. # sc-6243) into 500 µL of

1xPBS and incubating at 4 °C on a rotator for 1.5 hours. The IP-antibody-IP-matrix complex was

subsequently washed twice with 1x PBS, and centrifuged briefly at 13,000 rpm for 1 min at 4 °C.

The precleared lysates were then immunoprecipitated with the complexed HA-antibody and protein

A-matrix for 2 hours at 4 °C on a rotator in a total volume of 1 mL of 1x RIPA. After the incubation,

the eppendorf tubes were microcentrifuged at 13,000 rpm for 30 seconds to pellet the IP matrix and

washed with 1x RIPA (with protease inhibitors) for 3 or 4 times. To release the immunoprecipitated

protein 35 µL of 6xSDS lysis buffer was added to the pellet and the samples were boiled at 100 °C

for 5 minutes, vortexed briefly, and centrifuged at 13,000 rpm for 5 mins and loaded on the SDS-

acrylamide gels for Western blot analysis.

Page 35: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

25

2.9 Propidium iodide (PI) staining and FACS analysis

For the FACS analysis, cells to be analyzed were fixed in 70% ethanol and stained with propidium

iodide (PI). For analyzing the effect of PHIP1 knockdown on cell-cycle progression, 200,000 MDA-

MB-231 cells were seeded per well onto 6-well plates in 10% FBS DMEM in six replicates and

transfected with 100 nM siRNA (siScrambled or siPHIP) the next day as per described in Section 2.4.

The next day, the medium was changed to either 0.5% FBS or 5% FBS and incubated in 37 oC and

5% CO2 for 48 hours. The cells were prepared for FACS by washing once with sterile 1x PBS and

trypsinizing with 0.05% trypsin for 5 minutes. The cells were centrifuged for 5 minutes at 1,500 rpm

in 10% FBS DMEM growth medium and the supernatant was carefully aspirated. The cell pellet was

thoroughly resuspended with 0.5 mL PBS and 10 mLs of 70% ethanol was added and fixed overnight

at -20oC, or until staining. PI staining solution consisted of 1 mg/mL PI (Molecular Probes, Cat. # V-

13242,), 0.2% TritonX-100 and 0.2 mg/mL of RNAseA (DNAse-free; Sigma, Cat. # R-6513) all in

1x PBS. For staining, fixed cells were centrifuged at 3,000 rpm for 3 minutes, after which the 70%

ethanol was removed as much as possible. The cell pellet was washed once with 1x PBS by

suspending and centrifugating briefly at 5 minutes, 1,500 rpm. 1 mL of the PI solution was added to

each 15 mL falcon tube and the thoroughly resuspended cell pellet was incubated at room

temperature for 30 minutes in the dark. For the FACS analysis, the cells in the PI staining solution

were transferred to glass tubes and the data were acquired on the FACS (Yoda) with settings

optimized manually. 16,000 cells were counted per flow measurement. The raw data analyses were

performed using the WinMDI 2.9 software.

2.10 RNA Isolation and real-time quantitative PCR (RT-qPCR)

Total RNA was isolated from cells using the Qiagen RNeasy Mini Kit (Qiagen, Cat. # 74104)

according to the manufacturer’s instructions. Briefly, 200,000 to 500,000 cells seeded per well on 6-

well plates were lysed in 600 µL RLT buffer and centrifuged in QIAshredder spin column for 2 min

at 13000 rpm 4 °C. Then same volume of 70% EtOH (made with RNase-free water) was added to the

flow through and mixed 5-10 times by pipeting. The mixture was re-centrifuged in fresh RNeasy spin

columns at 13000 rpm for 15s at 4 °C twice in 600 µL and the flow-through was discarded. Then

700µl of Buffer RW1 was added and centrifuge for 15s at 13000 rpm at 4 °C, where the flow-

through was again discarded. Afterwards, 500µl Buffer RPE was added to the RNeasy spin column

and spun at 13000 rpm for 15s at 4 °C. The flow-through was discarded and the last step was

repeated once more. For eluting the RNA, the RNeasy spin column was placed in a new 1.5mL

Page 36: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

26

eppendorf tube and 30 µl RNAse-free water was added directly to the spin-column membrane and

centrifuged at 13000 rpm, 1 min, 4 °C. The flow through was measured for RNA concentration on a

spectrophotometer. Following extraction, total RNA was treated with rDNAse I (Ambion, Cat. #

AM1906) to remove any DNA contamination. For reverse transcription, High Capacity cDNA

Reverse Transcriptase Kit was used (Applied Biosystems Cat. # 4368813). Per reaction, 2 µL of 10x

RT Buffer, 0.8 µL of 25x dNTP mix (100 mM), 2 µL 10x RT Random Primers, 1 µL Multiscribe

reverse transcriptase, 4.2 µL RNAse-free H2O and 10 µL of RNA (1 µg total) were mixed on ice and

put into a thermocycler at 25 °C for 10 min, 37 °C for 120 min, 85 °C for 5s and 4 °C for indefinite.

The resulting cDNA (10 ng per reaction or 2.5ng/µl) was used for amplification in quantitative real-

time PCR (qPCR). cDNA (4 µL/well) was added to a qPCR mixture (6 µL/well) containing 3.475 µL

RNAse-free H2O, 1 µL of 10x PCR buffer, 0.6µL of 50 mM MgCl2 , 0.2 µL of 50 µM Primer Mix,

0.2 µL of 10 mM dNTP mixture, 0.2 µL ROX reference dye, 0.3 µL SYBR Green I (stock diluted

1:1000 in RNAse-free water) and 0.025 µl (or 125 U) Platinum Taq Polymerase (Invitrogen, Cat. #

10966-083)). For PCR amplification, the qPCR was set to 95 °C for 3min, 40 cycles of PCR, 95 °C

for 10 s, 65 °C for 15 s, 72 °C for 20 s, 95 °C for 15 s, 60 °C for 15 s, and 95 °C for 15 s and

performed in the ABI Prism 7900 HT Sequence Detection System (Applied Biosystems).

PHIP1 (C-TER) F 5'-CTA GAA ATC AAG GTC GAA GGA CAG C-3' R 5'-TGC CTT TGC TCC TCT TCA GAG TCA-3'

PHIP1 (3'-UTR) F 5'-AAT CGT GGA GGT AGG AAG CCC AAA-3' R 5'-TCT TCG AAC AAC AGC TGC CTT TGC-3'

CYCLIN D1 F 5'-GGC GGA GGA GAA CAA ACA GAT CAT-3' R 5'-TTG ATC ACT CTG GAG AGG AAG CGT-3'

CYCLIN D2 F 5'-AGA CCT TCA TTG CTC TGT GTG CCA-3' R 5'-CAA TCT GCT CCT GGC AAG CTT TGA-3'

p21 F 5'-TTA GCA GCG GAA CAA GGA GTC AGA-3' R 5'-ACA CTA AGC ACT TCA GTG CCT CCA-3'

TBP F 5'-GGG CAT TAT TTG TGC ACT GAG A-3' R 5'-TAG CAG CAC GGT ATG AGC AAC T-3'

Table 2. Sequence of qPCR primers used in the study

Page 37: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

27

Gene specific oligonucleotide primers were designed using the Primer Quest SciTool from Integrated

DNA Technologies (IDT). The expression level of the various transcripts was calculated using the

comparative CT method as described in the ABI Manual in detail. The values were normalized to that

of the TATA-binding protein (TBP), which is a ubiquitous transcription factor, whose gene is

frequently used for comparative qPCR analyses.

Page 38: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

28

Chapter 3. Results

3.1 PHIP1 is overexpressed in primary breast tumor samples of MMTV transgenic mouse models and in MCF10A stable cell lines overexpressing members of the IGF-IR pathway.

One of the common mechanisms of a proto-oncogene-mediated oncogenic transformation is

overexpression of the protein product, and the differential expression profile of PHIP observed in

early screening of human breast cancer cell lines led us to question its potential role in the mammary

gland tumorigenesis. The initial Western blot analysis of human breast cancer cell lines indicated that

PHIP1 was notably upregulated in cancerous cell lines compared to human mammary epithelial cell

line (HMEC) (Janet Farhang-Fallah, unpublished data), which are primary normal cells usually

harvested from tissues removed during breast reduction surgeries (Stampfer et al, 1993). As PHIP1

was previously found to be a downstream nuclear co-activator of the IGF-IR signaling (Podcheko et

al, 2007), this prompted us to look at whether PHIP1 could be also differentially expressed in the

MCF10A stable cell lines overexpressing the constitutively active, truncated IGF-IR (CD8-IGF-IR)

as well as those with IRS-1 or -2 overexpression. Whole cell lysates subjected to the immunoblot

analyses indicated that PHIP1 was indeed distinctively present in the MCF10A cell lines with ample

IGF-IR signaling molecules.

To extend and confirm the finding that PHIP1 was more abundantly present in tumor-derived

cells compared to that of the normal, we performed Western blot analysis on the crude lysates of the

breast tumors derived from transgenic mice of FVB strain overexpressing classical breast oncogenes

Wnt, Neu, and IGF-IR (Figure 6). The Western blot revealed more robust level of PHIP1 in breast

tumor samples of the oncogenic MMTV mice compared to that of the FVB wild-type female mouse.

The results suggested that PHIP protein levels could be elevated in different in vivo breast cancer

models.

3.2 MCF10A-PHIP1 stable cell line spontaneously silences ectopic PHIP1 expression at the protein level.

In order to study the oncogenic property of human PHIP1, we attempted to generate PHIP1-

overexpressing MCF10A stable cell line, which would enable us to do long-term experiments, such

as in vivo orthotopic injections, soft agar anchorage-independence colony formation assay and 3D

Page 39: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

29

A. B.

1 2 3 4 5 6 7 8 9

Figure 6. PHIP is manifested in the primary breast tumor samples of MMTV transgenic mouse

models of classical oncogenes, and in MCF10A stable cell lines with constitutively active IGF-IR

pathway.

(A). PHIP expression profile in mouse mammary gland tissue samples of different breast tumor models. MMTV-

Neu (N) represents a mouse model with overexpressed growth receptor ErbB2 and MMTV-Wnt (W) represents

overexpression of the differentiation marker Wnt, which often results in EMT and metastasis. N85 and N135 refer to

two independent mice of MMTV-Neu strain. Likewise, W204, W225 and W248 are three independent MMTV-Wnt

mice. MMTV-IGFR (RJ) similarly overexpress human IGF-IR in the mammary gland. N and W breast tumor

samples were provided by Dr. Liu from Dr. Zacksenhaus’ lab in University of Toronto, while RJ samples were

obtained from Dr. Moorehead at the University of Guelph. The Western blot was performed only once. (B).

Densitometry analysis of the Western blot for PHIP expression in mouse breast tumors normalized against the

values from actin. The error bars indicate standard deviation of 6 scannings of the one Western blot from (A). (C).

Expression of PHIP in MCF10A stable cell lines overexpressing components of the IGF-IR pathway. MCF10A-

IRS1 (lane 2) and MCF10A-IRS2 (lane 3) cells are MCF10A cells that were infected with retrovirus coding for

C.

Actin

PHIP1

HA

1 2 3 4

Page 40: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

30

wild-type human IRS-1 and IRS-2 genes containing HA tags (as shown in the blot). The Western blot was

performed only once.

growth in Matrigel™. The technical problem with the production of viable retrovirus was troubleshot

at multiple levels. Firstly, three retroviral packaging cell lines, Phoenix, 293T and PT-67, were tested

for transfection efficiency and retroviral titre, with a GFP-positive retroviral vector (MIEV, or

MSCV-IRES-EGFP-vector) for better visibility of the procedures. As recommended by the Nolan

laboratory at Standford University, the NIH/3T3 cell line was used to test the viral titre as it is easily

infected with retrovirus compared to other mammalian cell lines. PT-67, a NIH/3T3-based packaging

cell line, was relatively difficult to transfect with Lipofectamine2000, and required a long-term

selection with a high concentration of G418 (at 500 μg/mL). Generally, stable PT-67 transfectants as

separate colonies are screened and tested for viral titre. Also, upon testing for the presence of

functional virus by using the supernatant from PT-67 cells transfected with MIEV to infect NIH/3T3,

there were very few GFP positive cells and the titre proved to be too low, requiring the laborious

selection of high-titre PT-67 clones. Thus the utilization of PT-67 was discontinued. Phoenix cells,

being 293T-based, had the advantage of easy transfectivity. However, transfecting MSCV alone did

not produce functioning retrovirus. It was later realized that the Phoenix cell line had lost its stable

expression of Gag-Env-Pol, as it required pCl-Eco, a helper plasmid that contained the retrovirus

structural genes, in order to generate retrovirus capable of infecting NIH/3T3 cells. Unfortunately,

pCl-Eco was ecotrophic and could not produce retrovirus compatible with human cells. Therefore,

pCl-Ampho was obtained from University of Montreal as a generous gift and amphotrophic

retrovirus was generated by co-transfecting pCl-Ampho and pMSCV-HA-PHIP or pMIEV into 293T

cells and collecting the culture media 48 hours post-transfection. Transduction protocol was also

optimized by performing spin-infection, minimizing the volume of infection mixture to reduce viral

Brownian motion, testing different virus versus medium ratios, and adding appropriate amount of

polybrene to neutralize surface charge on the cell membrane and enhance viral adsorption (Davis et

al, 2002). MIEV was used in every transfection and infection as a visual marker and a positive

control for the presence of active retrovirus. Upon optimizing the retrovirus production and

transduction protocols, I proceeded to generate both NIH/3T3 and MCF10A stable cell lines by spin-

infecting cells on six-well plates. Cells were infected every second day three times, and drug

selection was started 48-72 hours after the last infection.

Excitingly, Western blot of cells shortly after the antibiotic selection with G418 showed

robust expression of HA in MCF10A (Figure 7). This, however, was unexpectedly diminished with

sub-culturing (Figure 8), rendering the cell line unusable for long-term analysis, such as orthotopical

Page 41: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

31

Figure 7. Retroviral infection of MCF10A leads to robust expression of HA.

(A). MSCV-HA-PHIP retroviral vector expression in 293T cells. 293T cells were transfected with MSCV-HA-

PHIP (lane 1, MSCV-PHIP), MSCV-empty or MIEV-control vector as controls. Cell lysates were probed with

anti-PHIP or anti-tubulin antibodies. The Western blot is a representative of at least 3 independent experiments

(N=3). (B). MCF10A cells were infected with MSCV-PHIP retrovirus three consecutive times (lane 2,

MCF10A-PHIP III) or with MIEV retrovirus as control (lane 1, MCF10A-MIEV) and cells were selected with

G418 for 10 days. Lane 3 represents MCF10A cells infected with MSCV-PHIP retrovirus 6 times but without

drug selection. Cells were collected on day 12 after the last infection and lysates were probed for expression of

recombinant HA-PHIP using anti-HA antibodies. Lane 4 represents Phoenix cells transfected with MSCV-HA-

PHIP, which was used as a Western blot control for HA-PHIP expression levels. The figure shown is a

representative of 2 Western blots (N=2). (C). Fluorescent pictures of MCF10A-MIEV and NIH/3T3-MIEV

control stable cells 48 hours after the third retroviral infection, plated on 6-well plates. Cells were lysed for

Western blot analysis on day 12 after the last infection, and same degree of fluorescence was observed. The

GFP photographs are representative figures from at least 3 independent experiments (N=3). (D). MCF10A

cells infected with either MSCV-PHIP or MIEV-control virus and were selected for 10 days in G418. Selected

A. B.

C. D. MCF10A-PHIP

MCF10A-MIEV

PHIP

293T MSCV-PHIP

293T MSCV-empty

293T MIEV

Tubulin

MCF10A-MIEV

MCF10A-PHIP III

MCF10A-PHIP VI

HA

PHIP

Tubulin

Phoenix/MSCV-PHIP

1 2 3 1 2 3 4

MCF10A-MIEV NIH/3T3-MIEV

Page 42: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

32

cells were expanded and 5,000 cells were then seeded on soft agar for 14 days. Cells were stained with 2

mg/mL MTT and imaged using Nikon digital camera. The experiment was successfully performed only once.

Figure 8. Loss of HA-PHIP expression in the MCF10A stables.

(A). A Western blot showing the significant decrease of HA-PHIP signal in MCF10A-PHIP III that previously

had robust HA (lane 3). Lane 1 and 2 respectively represent non-infected MCF10A-parental and MCF10A-

MIEV as HA negative controls. MCF10A-PHIP VI (lane 4) is MCF10A cells infected with PHIP retrovirus for

six times but not selected with G418. NIH/3T3-PHIP III (lane 6) refers to NIH/3T3 cells infected with PHIP

retrovirus three times under the same conditions as MCF10A cell line, and NIH/3T3-MIEV (lane 5) acts as its

GFP control. MCF10A-CD8-IGFR (lane 7) and MDA-MB-231 (lane 8) were included as PHIP positive

control. Phoenix/MSCV-PHIP (lane 9) indicates Phoenix cells that were transfected with MSCV-PHIP vector

and acts as both PHIP and HA positive control. The figure is a representative of 2 Western blots (N=2). (B).

The earliest passage of MCF10A-PHIP III (lane 2) and MCF10A-vector control (lane 1) that had been

cryopreserved after first subculture following G418 selection for 10 days were thawed and analyzed for HA

signal, which was present. The Western blot was run only once (N=1).

inoculation into SCID or nude mice to see tumor formation in vivo. In another round of infection,

single colonies were isolated, but none gave the HA signal, despite the continued presence of GFP in

the cells infected with GFP-only control virus. At first, the disappearance of HA signal in MCF10A-

PHIP was surmised to arise from the environmental factors that may modulate PHIP expression in

order to maintain the cellular homeostasis, such as the concentration of growth components in the

MCF10A medium, cell density, and the presence of G418. Upon consulting Dr. Senthil Muthuswamy,

A. B.

MCF10A-vecto

r

MCF10A-PHIP III

PHIP

Tubulin

MCF10A parental

MCF10A-MIEV

MCF10A-PHIP III

MCF10A-PHIP VI

170kDa130kDa

95kDa

72kDa

55kDa

40kDa

NIH/3T3-MIEV

NIH/3T3-PHIP III

MCF10A-CD8-IGFR

MDA-MB-231

Phoenix/MSCV-PHIP

HA

PHIP

IGF-IR

Tubulin

HA

1 2

1 2 3 4 5 6 7 8 9

Page 43: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

33

A. B.

Figure 9. PHIP is targeted for protein degradation in MCF10A-PHIP stable cell line.

(A). HA-PHIP mRNA levels in MCF10A-PHIP stable cell line were quantified using RT-qPCR analysis.

Recombinant PHIP cDNA was amplified using primers designed to the C-terminal region of the human PHIP

mRNA and was normalized to an ubiquitous control gene, in this case TATA-binding protein (TBP). The

statistical analysis was done by Student’s t-test. The qPCR was performed once with replicates of 3 indpendent

wells (n=3). The error bars indicate standard deviation. (B). MCF10A-PHIP cells (lanes 4, 5 and 8, 9) that had

previously lost the expression of HA-PHIP were plated in 200,000 cells per well on 6-well plates. On the next

day, the cells were treated with 5 µM MG132 proteasomal inhibitor or its solvent DMSO in MCF10A growth

medium for either 4 or 16 hours and lysed at the indicated time points in 6x SDS. The Western blot

membranes were blotted for HA and p53, which was used as a positive control to MG132 as it is a well-known

target of ubiquitination. The first lane indicates MCF10A-vector cells that had neither DMSO nor MG132 in

the medium during the 4 hours. The Western blot is a representative of 2 independent experiments (N=2).

one of the co-developers of the MCF10A culture system, we were advised that the cells may be

compensating as a feedback response to the probable excessive activation of the insulin pathway

triggered by the ectopic overexpression of the wild-type PHIP. Therefore, it was highly

recommended to reduce insulin levels in the MCF10A media (personal communications).

Unfortunately, changes in those conditions did not seem to influence the PHIP levels. The specificity

of the current antibody was also suspected, despite the unfailing presence of a thick band for the

positive control 293T-MSCV-PHIP lysate, and new WDR-targeting antibody from the crude rabbit

sera was extracted. The antibody gave a clean band, but to our disappointment, the size did not

correspond to PHIP, possibly indicating the presence of another WDR-containing isoform.

Surprisingly, RT-qPCR analysis using human PHIP primers targeting the C-terminal region

No treat

DMSOMG132

DMSOMG132

DMSOMG132

DMSOMG132

vector vectorPHIP PHIP

4 HOURS 16 HOURS

HA

p53

β-actin

1 2 3 4 5 6 7 8 9

Page 44: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

34

revealed that PHIP was in fact being overexpressed in the stable MCF10A cell lines at the transcript

level (Figure 9A). This led us to question whether PHIP was specifically being targeted for protein

degradation. To test this hypothesis, the MCF10A stable cells were treated with MG132, a

proteasomal inhibitor, and lysed at either 4 or 16 hours after treatment. Intriguingly, HA signal was at

least partially restored on the Western blot (Figure 9B), suggesting that PHIP was being

proteolytically degraded, rather than being silenced at the transcriptional level, which sometimes can

occur in retrovirally generated stable cell lines (Swindle et al, 2004). One possible explanation

behind this occurrence may be that PHIP auto-regulates its own protein levels, as a putative binding

partner to the DDB1-Cul4 ubiquitin ligase complex (Angers et al, 2006), which will be discussed

further in the Discussion, Chapter 4. Unfortunately, this aspect could not be investigated further due

to time and reagent limitations.

3.3 AdPHIP1 is not the ideal system for the soft agar colony-forming assay

In favor of our hypothesis that PHIP1 is involved in oncogenic transformation of breast epithelial

cells, initial analysis with the MCF10A-PHIP1 stable cell line resulted in positive colony

formation on soft agar in 2 weeks post-seeding (Figure 7), indicating that the cells could acquire

the ability to grow in the absence of anchorage with PHIP1 expressed approximately twice the

physiological level. However, due to the spontaneous silencing of the ectopic PHIP1 after a

number of rounds of sub-culturing, we attempted to replicate the soft agar colony formation

assay using an alternative means of protein overexpression.

It was previously shown that adenovirus coding for human PHIP1 (AdPHIP) with HA

epitope could successfully overexpress the protein in INS-1 cells (Podcheko et al, 2007). Thus to

circumvent the silencing in the stable cell lines, we attempted using AdPHIP as an alternative

means to examine the effect of full-length PHIP1 overexpression. Although soft agar assays

typically take 3-4 weeks, there have been reported cases of adenovirus used in the long-term

experiment, such as in Lee et al (2003), where transduction of lung cancer cells with adenovirus

expressing truncated (defective) IGF-IR led to a significant suppression of colony formation in a

soft agar clonogenic assay. Encouragingly, the authors had also previously reported successful

colony formation using adenovirus after 3 weeks (Lee et al, 1996). Upon testing the virus on

MCF10A cells for the degree of overexpression, the virus was able to reproducibly produce HA

signal in a dose-dependent manner (Figure 10). Unfortunately, however, despite multiple

attempts, the HA-PHIP1 expression could not be maintained over one week and thus no colony

Page 45: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

35

1 2 3 4 5 6 7 8 1 2 3 4 5 6 7 8 9

Figure 10. Soft agar results with MCF10A cells infected with PHIP1 adenovirus.

(A). MCF10A-cells infected with AdGFP (second panel) or AdPHIP (third panel) were plated on low-melting

agarose (soft agarose) and stained with MTT 16 days after seeding. MCF10A-ERBB2 (first panel panel),

which stably overexpress constitutively active ERBB2 receptor, was kindly donated by Dr. Senthil

Muthuswamy and was used as the positive control in the assay. The figures are a representative of 5

independent experiments (N=5). (B). MCF10A parental cells were infected with different doses of either

AdGFP or AdPHIP and were analyzed for dose-dependent overexpression of HA-PHIP 48 hours post-infection.

The figure is representative of 2 independent Western blots (N=2). (C). MCF10A cells that had been

concurrently infected with those seeded on soft agar were maintained on 6-cm plates and lysed on the

designated dates (Days 4, 9 and 14) for Western blot analysis of HA-PHIP signal. Day 0 was taken as the day

of the adenoviral infection. The Western blot is a representative of 5 independent assays (N=5).

Page 46: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

36

was observed even after 20 days. MCF10A-ERBB2, which has stable amplified gene expression

of constitutively active, truncated ERBB2, was used as the positive control and it produced

visible colonies in every soft agar assay attempted. Likewise, the MCF10A cells infected with

GFP adenovirus, which was used as the negative control, did not show any colony formation,

which validated the soft agar assay technique.

3.4 TRIPZ shRNA-PHIP constructs fail to maintain PHIP knockdown. One of the typical approaches used in cancer research to study the effect of a protein is the generation

of shRNA stables in the cancerous cell line of interest. Stable knockdown of the protein under study

allows one to investigate tumor cell growth in vivo through xenograft implantation or orthotopic

injections, as well as numerous in vitro assays such as anchorage-independent growth in soft agar, 3D

acini formation in Matrigel, and migration assays.

In an attempt to create an optimal model system that will enable us to study the role of PHIP

in multiple aspects of breast cancer, we pursued to obtain the tet-inducible shRNA-TRIPZ plasmids.

One major benefit of employing an inducible system in MCF10A-based cell line would be the ability

to investigate the effect of the knockdown of a protein under study at different time points of the

acinar morphogenesis, as well as being able to show reversibility and the specificity of an

observation associated with the protein that is being knocked down.

For the generation of the stable cell lines to be used in our experiments, MCF10A-IGFR were

infected three times every second day with appropriate TRIPZ-shRNA lentivirus, and were exposed

to 1 µg/mL doxycycline for minimum 72 hours before being FACS-sorted by TurboRed (Figure 11C)

versus 7-AAD, a cell viability marker. Initial analysis indicated that there was some degree of PHIP

knockdown (Figure 11A), which was supported by the proliferation assay (Figure 11B). However,

subsequent Western blot indicated no change in the protein level, and neither the amount of

doxycycline nor the length of the exposure time influenced the outcome (Figure 11D). To validate

that the TRIPZ shRNA-PHIP constructs from the Openbiosystems were indeed able to knockdown

PHIP, and to exclude the possibility that such obliteration of knockdown could be a MCF10A-

specific effect, the TRIPZ lentiviral stable cell lines were re-generated in MDA-MB-231 (Figure 12).

Removal of cells that do not express the shRNA transgene by puromycin selection was considered

important, as transfection and expression of the TRIPZ constructs in 293Ts gave dubious results

multiple times and this observation was suspected to be due to masking of the visible knockdown by

high PHIP levels from the non-transfected cells. Also, in an attempt to mimic the constitution of the

SMARTpool siRNA from Dharmacon where there are multiple siRNA sequences that collectively

Page 47: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

37

lead to efficient PHIP knockdown, a plate of MDA-MB-231 was infected with ‘pooled’ TRIPZ

lentivirus generated by transfecting three different shRNA-PHIP constructs at equal ratios to 293T.

Fortuitously, Western blot analysis of cells straight after puromycin selection indicated a

partial reduction in PHIP levels in shRNA_174591 and shRNA_174592 after 72 hours exposure to 1

µg/mL doxycycline (Figure 12C). To test whether this observation could be associated with a

biological effect, a proliferation assay was performed using two different methods; one by enzymatic

MTT growth assay and the other by directly counting cell number with Beckman Coulter Z2 (Figure

12D). To our astonishment and inconsistent to the previous results where PHIP knockdown led to

inhibition of cell growth, the shRNA stables in fact grew better than the Scrambled control cells.

Western blot of the cells revealed that there was no observable PHIP knockdown at the protein level

(Figure 12E, F). However, though the lentivirally generated shRNA stable cell lines were deemed

unfit to be used in the study, this phenomenon might be a further important and intriguing suggestion

that PHIP is regulated at multiple stages and such potentially intricate regulation might be subjected

to another exciting investigation.

Page 48: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

38

B. A.

Figure 11 Analyses of MCF10A-IGF-IR TRIPZ stable cell lines.

(A). MCF10A-IGFR cells infected with lentivirus generated from TRIPZ-shRNA-Scrambled (lane 1), -GAPDH

(lane 2) or -174592 (lane 3) were treated with 1 µg/mL doxycyline for 72 hours following puromycin selection and

were blotted for PHIP, E-cadherin, IGF-IR and tubulin on the Western blot. The Western blot represents n=1. (B).

MTT assay was performed to assess the growth pattern of newly generated shRNA stables corresponding to (A).

5,000 cells of MCF10A-IGFR-TRIPZ-shRNA-Scrambled or shRNA-174592 were plated onto 96-well plates in four

replicates (n=4) in either starved medium, which consisted of plain DMEM/F12 (serum free), or regular MCF10A

growth medium containing 5% horse serum and other components. The day of seeding was designated as Day 0.

The media were changed every two days and replenished with fresh doxycyline. The cell proliferation was measured

on every second day using the MTT assay, and wells only containing the media without cells were used as MTT

negative control. The proliferation assay was performed once. The statistical analyses were done by two-way

ANOVA. The astrix refers to statistical significance between ‘Scrambled Growth’ and ‘174592 Growth’ groups. **

= P <0.01 *** = P <0.001 (C). 72 hours post-treatment with doxycyline, MCF10A-IGFR stables of TRIPZ-shRNA-

Scrambled or TRIPZ-shRNA-174592 were photographed under the Nikon fluorescence microscope, which show the

C. D.

TRIPZ Scrambled

TRIPZ GAPDH

TRIPZ 174592

E-cad

IGF-IR

Tubulin

Day 1

Day 2

Day 4

Day 6

Day 8

0.0

0.2

0.4

0.6Scrambled Starved174592 StarvedScrambled Growth174592 GrowthBlank (medium only)

**

***

***

Abs

orba

nce

595n

m/6

50nm

PHIP1

1 2 3

shRNA_Scrambled shRNA_174592

Day 3 Day 6

0.0 0.5 1.0 0.0 0.5 1.0 0.0 0.5 1.0 0.0 0.5 1.0Dox (ug/mL)

Scrambled ScrambledshPHIP shPHIP

MCF10A-vector

PHIP1

IGF-IR

Tubulin

Page 49: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

39

robust presence of doxycycline-induced red fluorescence (RFP). The photographs are a representative of at least 3

independent experiments. (D). PHIP1 knockdown in MCF10A-IGFR TRIPZ stables was assessed under different

conditions of amount of doxycyline used (0, 0.5 or 1.0 µg/mL) or the length of treatment (for 3 days or 6 days after

seeding). The cells lysed were blotted with PHIP, IGF-IR or tubulin. Medium was changed every day with fresh

doxycycline, and MCF10A-vector (last lane on the right) was used as the negative control for IGF-IR. The Western

blot was performed only once.

B. A. TRIPZ Scrambled TRIPZ 174591 TRIPZ 174592

293T MSCV-vector

293T MSCV-PHIP

293T PEI only

293T TRIPZ Scrambled

293T TRIPZ pooled

293T TRIPZ 174590

293T TRIPZ 174591

293T TRIPZ 174592

170kDa

55kDa

PHIP1

Tubulin

1 2 3 4 5 6 7 8

C. D.

Figure 12. Validation of TRIPZ constructs in 293T and MDA-MB-231 cells.

E. F.

Scr

ambl

ed

1745

91

1745

92

Scr

ambl

ed

1745

91

1745

92

Scr

ambl

ed

1745

91

1745

92

PHIP

β -actin

PHIP(lower exposure)

Day 2 Day 4 Day 6

0.5% FBS

1 2 3 4 5 6

Day 2 Day 4 Day 6

Scr

ambl

ed

1745

91

1745

92

Scr

ambl

ed

1745

91

1745

92

Scr

ambl

ed

1745

91

1745

925% FBS

PHIP

β -actin

PHIP(lower exposure)

Page 50: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

40

(A). 293T cells were transfected with different shRNA constructs with the TRIPZ backbone plasmid, including

174590 (lane 6), 174591 (lane 7) and 174592 (lane 8). In order to mimic the synergistic effect of having

multiple shRNA as in SMARTpool siRNA, the three TRIPZ-shRNA plasmids were co-transfected in 293T in

1:1:1 ratio (lane 5). These were compared to 293T transfected with TRIPZ-shRNA-Scrambled (lane 4) and

293T treated with only PEI without DNA (lane 3). 293T transfected with MSCV-PHIP (lane 2) was used as a

PHIP positive control as well as for the test of PEI functionality, which was compared to 293T transfected with

MSCV empty vector (lane 1). The Western blot is a representative of at least 3 independent experiments

(N=3). (B). MDA-MB-231 cells were infected with lentiviruses generated from TRIPZ-shRNA-174590,

174591, 174592 or from pooled transfection, and selected with puromycin for 5 days. The treatment of

doxycycline was performed following the drug selection, and pictures show MDA-MB-231 TRIPZ stables

after 72 hours of 1 µg/mL doxycycline treatment. The photographs are a representative of at least 2

independent assays (N=2). (C). The MDA-MB-231 TRIPZ stables that had undergone puromycin selection

and 72 hour-treatment with doxycycline was run on the Western blot to analyze degree of PHIP knockdown.

The stable cell lines were generated by infecting MDA-MB-231 cells with lentivirus mixed with growth

medium in 1:1 ratio and 8 µg/mL of polybrene. Three independent TRIPZ shRNA constructs 174590 (lane 4),

174591 (lane 5), 174592 (lane 6) as well as from pooled infection of all three lentiviral types (lane 3) were

compared to stables of TRIPZ-shRNA-Scrambled (lane 1) and shRNA-GAPDH (lane 2). The Western blot

was performed successfully only once (N=1). (D). 100,000 cells of MDA-MB-231 TRIPZ-shRNA-173491,

174592 and Scrambled were seeded onto 6-well plate in 5% or 0.5% FBS media and counted with Beckman

Coulter every two days for 6 days. Each datapoint represent six replicates (n=6), and the media containing

fresh doxycyline was replaced every 2 days throughout the experiment. The experiment is a representative of

two independent experiments (N=2). (E+F). The MDA-MB-231 TRIPZ stables that were used in the

proliferation assay as in (D) and grown under either 0.5% or 5% FBS were also maintained in 6-well plates

and lysed on Day 2, 4 and 6 for Western blot analyses. The Western blot was performed only once (N=1).

3.5 PHIP1 is implicated in cancer cell proliferation

As PHIP1 is a known downstream regulator of the IGF-IR pathway, we assessed whether PHIP was

implicated in IGF-IR-mediated mitogenesis, and thus the effect of siRNA-mediated knockdown of

PHIP on the growth of MCF10A-CD8-IGF-IR cell line was examined. siRNA-PHIP–treated

MCF10A-IGF-IR cells reproducibly displayed significant growth inhibition compared to the siRNA-

Scrambled-treated counterparts, as shown in Figure 13. More convincingly, as the protein level of

PHIP was partially restored on Day 8 with the time-dependent degradation of the siRNA from the

cells, the cell growth accordingly peaked, suggesting that PHIP is intimately involved in the cell

Page 51: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

41

1 2 3 4

Figure 13. PHIP knockdown inhibits the growth of MCF10A-IGFR cells.

(A). Proliferation assay of MCF10A-IGFR with PHIP knockdown. 50,000 MCF10A-IGFR cells were seeded on 12-

well plate and transfected with either siScrambled or siPHIP on Day 0 with Lipofectamine 2000. The medium was

changed the next day to reduced serum medium containing 0.5% horse serum and other constituents. Cells were

counted using Beckman Coulter counter (n=6 per group) for Days 2, 4, 6, and 8 and Statistical analysis was done by

two-way ANOVA test. *** P<0.0001. The figure shown is a representative of at least 2 independent experiments

(N>2). (B). The cells that had been transfected concurrently with those for the proliferation assay were lysed on each

time points of the experiment and analyzed on Western blot for the presence of siRNA-mediated PHIP knock-down.

The Western blot is a representative of 3 independent analyses (N=3). (C). MCF10A-IGFR were plated on 6-well

plate and transfected with either siScrambled or siPHIP overnight. The next day, the medium was changed to either

0.5% or 5% horse serum DMEM/F12 and the cells were incubated for 48 hours before lysis with 6x SDS lysis

buffer. The lysates were blotted for cyclin D1 and cyclin D2 as well as p-Akt to see the effect of PHIP knockdown

under complete (lanes 3, 4) and reduced (lanes 1, 2) serum conditions. The Western blot was performed only once.

(D). MCF10A-IGFR were similarly treated as (C) but grown in 5% horse serum for 48 hours before lysis in 6x SDS

for Western blot or RLT Buffer for qPCR. Total RNA was extracted from MCF10A-IGFR with no treatment (lane

Page 52: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

42

1), with siScrambled (lane 2) or siPHIP (lane 3) transfection and were reacted with primers against human p21.

Statistical analysis was done by one-way ANOVA where * refers to P < 0.05 and *** represents P < 0.0001. The

qPCR was performed once (N=1) with n=6 per datapoint.

A. B.

+ + +---IGF-I

PHIP1

p-Akt

p-MAPK 42/44

Actin

- - ++--siPHIP- + --+-siScram

1 2 3 4 5 6

050000

100000150000200000

250000300000

350000400000

siScrambled siPHIP

Cel

l num

ber

D.C.

siScrambled

siPHIP1

Actin

PHIP1

Figure 14. PHIP1 knockdown inhibits the growth of both ER positive and ER negative cell lines.

(A). MCF7 cells were reversely transfected with either siScrambled or siPHIP in 96-well plate (10,000 cells/well) in

five replicates and starved overnight, after which (+) cells were treated with 100 ng/mL IGF-I for 48 hours in the

presence of 1% serum. Parental cells (first two lanes) were included as positive control for mitogenic response to

IGF-I. The degree of cell proliferation was measured by the MTT assay, and media-only wells were included as

negative controls for the MTT reagent. Statistical analyses were done by performing one-way ANOVA. ns = not

significant. ***=P <0.0001. The experiment was successfully performed only once (N=1). (B). Using the same ratio,

200,000 MCF7 cells were reversely transfected on to 6-well plates, starved overnight and were either further starved

for 48 hours in 1% serum (-; lanes 1, 3, 5) or treated with 100 ng/mL IGF-I under the same condition (+; lanes 2, 4,

6). Cells were collected on the same time point as when the MTT assay was performed, and lysates were blotted for

PHIP, p-Akt, p-MAPK and actin. The Western blot is a representative of 3 independent experiments (N=3). (C+D).

In the preliminary analysis, 200,000 MDA-MB-231 cells were plated on 6-well plate and transfected with either

Page 53: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

43

siScrambled or siPHIP with Lipofectamine 2000. The transfection mixture was replaced with 10% FBS DMEM 6

hours after transfection. The cells were counted 48 hours after on the hemocytomter and lysed for Western blot,

which was incubated with anti-PHIP and anti-actin antibodies to see the siRNA-mediated knockdown of PHIP. The

figures are a representative of at least two independent experiments (N>2).

A. B.

D.

MDA-231 s

iSCRA 0.5%

FBS

MDA-231 s

iPHIP 0.5%

FBS

MDA-231 s

iSCRA 5% FBS

MDA-231 s

iPHIP 5% FBS

0

50

100

150ApopG1SG2/M

Perc

ent o

f cel

l cou

nted

C.

MDA-231 siScrambled 0.5% FBS vs MDA-231 siPHIP 0.5% FBS

CELL CYCLE PHASE P value Summary Apoptosis P > 0.05 ns G1 P<0.001 *** S P<0.001 *** G2/M P<0.001 ***

MDA-231 siScrambled 0.5% FBS vs MDA-231 siScrambled 5% FB S

CELL CYCLE PHASE P value Summary Apoptosis P > 0.05 ns G1 P<0.001 *** S P<0.001 *** G2/M P > 0.05 ns

MDA-231 siPHIP 0.5% FBS vs MDA-231 siPHIP 5% FBS

CELL CYCLE PHASE P value Summary Apoptosis P > 0.05 ns G1 P<0.001 *** S P<0.001 *** G2/M P<0.001 ***

MDA-231 siScrambled 5% FBS vs MDA-231 siPH IP 5% FBS

CELL CYCLE PHASE P value Summary Apoptosis P > 0.05 ns G1 P<0.001 *** S P<0.001 *** G2/M P<0.001 ***

Figure 15. PHIP knockdown in MDA-MB-231 arrests G1 to S phase transition of cell cycle.

(A) MDA-MB-231 cells were treated with siRNA-Scrambled (=siScram) or siRNA-PHIP (=siPHIP) in six replicates

and starved overnight the next day in 0.5% FBS DMEM. The cells were then incubated in either 0.5% or 5% FBS

media for further 24 hours, after which they were stained with propidium-iodide (PI) and sorted by FACS. 16,000

cells were measured during the flow-through and the values were averaged acrossed the replicates (n=6). The

experiment was performed only once (N=1). (B) Table showing the results of statistical analyses on the processed

FACS data. Two-way ANOVA was performed between the pairs indicated. ns = not significant; *** P<0.0001. (C).

Average values (in %) of proportions of cells in different phases of the cell cycle for the bar graph in (A). (D).

MDA-MB-231 cells were plated on 6-well plates and were transfected with siRNA Scrambled or PHIP as described

Page 54: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

44

previously. The cells were lysed in RLT buffer for qPCR analysis 48 hours post-transfection in the growth medium

and the cDNA reverse transcribed from the extracted RNA were reacted with primers against C-terminal region of

PHIP transcript (PHIP C-TER), 3’UTR of PHIP (PHIP 3’UTR), p21, cyclin D1 or cyclin D2. Each datapoints

represent two independent samples of triplicate values each (n=6 total). The comparative Ct method was used to

calculate the fold difference, using the TBP values as the baseline, and the Student’s t-test was used for the statistical

analyses. The error bars indicate the standard deviation.

proliferation. The level of cyclin D2, but not cyclin D1, was also reduced on the Western blot, and

this effect could not be overridden by the presence of high serum (5% equine serum) (Figure 13C),

confirming that cyclin D2 expression is regulated by PHIP consistent to the previous finding by

(Podcheko et al, 2007). In order to support that downregulation of PHIP inhibits the enhanced

cellular proliferation induced by IGF-I, MCF7 was used as another cell model. MCF7 is an estrogen

receptor (ER)-positive luminal epithelial cell line that responds mitogenically to IGF-I (Dufourny et

al, 1997; Dupont et al, 2001). These cells were reversely transfected with siRNA and were starved in

serum-free medium overnight before being treated with the physiological concentration of IGF-I

(100 ng/mL) for 48 hours. As expected, measurement of the relative live cell number by the MTT

assay demonstrated that the mitogenic effect of IGF-I was curtailed in cells transfected with siRNA-

PHIP (Figure 14A). Changes in the level of phosphor-Akt was checked on the Western blot to

signify the cellular response to the IGF-I treatment (Figure 14B). Interestingly, the first knockdown

experiment using MCF7 seemed to exhibit dampening of the phophorylated Akt levels with PHIP

downregulation. As overexpression of PHIP was previously shown to promote the phosphorylation

of Akt in INS-1 cells (Podcheko et al, 2007), the experiment was repeated twice more in order to

validate whether PHIP knockdown prohibited Akt phosphorylation in MCF7 cells. Unfortunately,

subsequent Western blots failed to reproduce the initial observation.

Preliminary cell counting of MDA-MB-231 48 hours after transfection with siRNA indicated

that there was a significant difference in the cell number between siScrambled and siPHIP (Figure

14C). When there was reasonable knockdown, lower cell number was constantly observed in siPHIP

wells compared to siScrambled. As a noticeable number of floating cells were observed during the

siRNA transfection, we also attempted to address whether the reduced cell number with siPHIP

transfection was due to increased apoptosis or cell cycle arrest. FACS analysis with Propidium Iodide

(PI) staining on MDA-MB-231 with PHIP knockdown grown under two different serum conditions

(0.5% and 5% FBS) indicated that PHIP knockdown induces G1 arrest without significantly affecting

the degree of apoptosis (Figure 15A, B). This was extended further by performing qPCR on cell

cycle regulators to see how the knockdown of PHIP modulated them to prohibit the cell cycle

Page 55: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

45

progression. Upon approximately 90% reduction of PHIP in siRNA-PHIP treated MDA-MB-231

cells compared to siRNA-Scrambled controls, cyclin D2 was again visibly diminished, while p21

was slightly increased (Figure 15). Intriguingly, qPCR analysis on MCF10A-IGFR cells showed a

dramatic increase in p21 that was inversely dose-dependent to the amount of PHIP present in the cell

(Figure 13D).

3.6 PHIP1 may complex with p53

p21 belongs to the Cip/Kip family of cyclin-dependent kinase inhibitor (CKI) that mainly inhibits the

activity of cyclin E/cdk2 (Gartel et al, 2005). p21 is also a well-known transcriptional target of p53

(El-Deiry et al, 1993) and is required for the p53-dependent induction of G1 arrest in human cancer

cells (Waldman et al, 1995). Because knockdown of PHIP1 increased the level of p21, and the

magnitude of change was larger in MCF10A-based cells that contained wild-type p53 compared to

MDA-MB-231, which has mutated p53 (Lacroix et al, 2006), it prompted us to question whether

PHIP1 was directly involved in the transcriptional regulation of p21 by inducing proteasomal

degradation of the protein (for example, by activating MDM-2 through phosphorylation of Akt

(Ogawara et al, 2002; Zhou et al, 2001) or by potentially complexing with p53 and acting as a co-

repressor. Another lab member had also previously shown preliminary data that PHIP interacts with

p53 in MCF7 (Alexey Podcheko, unpublished data). The 293T cell line, which contains abundant

levels of wild-type p53, was chosen as the model system for the co-immunoprecipitation (co-IP)

experiment, in order to avoid co-transfecting the cells with two plasmids to introduce both HA-PHIP

and a differently tagged-p53. Additionally, antibody manufacturing companies commonly use 293T

as a source of the p53 protein to test their antibodies.

Unlike what was expected, transient overexpression of HA-PHIP by transfecting MSCV-HA-

PHIP into 293T did not decrease the level of p53. HA-IP protocol was optimized by performing

nuclear fractionation on the transfected cells to enrich HA-PHIP in the lysate similar to the methods

used in (Waldman et al, 1997), and by strictly limiting the incubation time with the antibody-agarose

bead complex to two hours in 4 oC to minimize protein degradation. After ensuring a successful HA-

IP, the membrane was stripped and re-probed with p53 antibody, and ExactaCruz secondary antibody

was used to eliminate unwanted signals from the IgG fragments. Remarkably, a thicker band on the

HA-IP lane compared to the control was observed (Figure 16B). The reverse co-IP, where p53 was

pulled down first, likewise gave a much stronger HA band in another set of trial (Figure 16C).

However, as later mentioned in the Discussion, this phenomenon was not consistently observed in the

other attempts.

Page 56: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

46

Figure 16. PHIP complexes with p53.

(A). Overexpression of PHIP in 293T does not change p53 levels. 293T cells were transfected with MSCV-empty

vector or MSCV-PHIP with PEI. 48 hours post-transfection, the cells were subjected to nuclear fractionation into

cytosolic (lanes 1, 2) or nuclear compartments (lanes 3, 4). The lysates were blotted for PHIP or p53 on the Western

blot. The Western blot is a representative of three independent experiments (N=3). (B). Inputs (lanes 1-4) equal to

(A), where HA-PHIP from cytosolic and nuclear compartments of 293T cells transfected with either MSCV-control

or MSCV-PHIP were detected with HA antibody. As PHIP was only found to be in the nucleus, the nuclear extract

was subjected to immunoprecipitation with HA antibody, and immunoblotted for both HA and p53 antibodies (lane

5 and 6). The immunoprecipitation experiment was successfully performed only once (N=1). (C). Reciprocal

immunoprecipitation was performed likewise, where nuclear fractionation was performed on 293T cells transfected

with MSCV-vector or MSCV-PHIP to enrich the concentration of PHIP and p53. The inputs are the nuclear lysates

before undergoing immunoprecipitation (lanes 1, 2). The nuclear lysates that were subjected to immunoprecipitation

with p53 antibody and immunoblotted with both p53 and HA antibody are represented in lanes 3 and 4. ExactaCruz

rabbit secondary antibody was used for the detection of immunoprecipitated p53, in order to eliminate the detection

C.

IB: HA

IB: p53

cont

rol

HA

-PH

IP

cont

rol

HA

-PH

IP

INPUTS OUTPUTS

IP: p53

170kDa

HA-IP of nuclear fraction

cont

rol

cont

rol

HA

-PH

IP

HA

-PH

IP

cytosolic nuclear

cont

rol

HA

-PH

IP

INPUTS OUTPUTS

IB: HA

IB: p53

Nuclear

170kDa130kDa95kDa

Cont

rol

PHIP

Cont

rol

PHIP

Cytosolic

PHIP1

p53

1 2 3 4

B. A.

1 2 3 4 5 6 1 2 3 4

Page 57: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

47

of heavy and light chains of the IgG that could conceal the signal for p53 at around 50 kDa. The

immunoprecipitation assay was successfully performed only once (N=1).

Page 58: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

48

Chapter 4. Discussion and Future Directions

4.1 PHIP1 is differentially expressed in cancer cells.

Western blot on the mammary tumor lysates of MMTV mouse models indicated that PHIP1 was

more prominently present in samples containing oncogenes compared to the control wild-type mouse,

which is a compelling evidence that PHIP1 is implicated in breast cancer. However, there needs to be

more data in order to make a more definitive conclusion:

First of all, there should have been more than one age-mate wild-type (WT) samples to take

into account the possible existence of variation among individual animals and between mouse strains.

Unfortunately, the initial analysis which included at least two control samples used refined preps of

mammary epithelial cells isolated using the Mammary Stem Cell Enrichment Kit (Stem Cell) and

lysed in 1x RIPA buffer, which yielded absolutely no PHIP1 band on the Western blot. It was later

realized that due to the nuclear confinement of PHIP, sonication for minimum of 10 seconds was

necessary to release PHIP1 from the cellular nuclei into the solution to enable its detection with the

antibody. Second caveat of this crude approach was that tumors that arise from different oncogenes

have characteristic phenotypes and that the loading control informs us neither of the proportion of the

proteins derived from the stromal versus epithelial cells nor of the degree of contribution from

different cell types within the tumor tissues. Although there are examples in the literature where

crude lysates of the mammary tissues processed with RIPA or PBS/NP-40-based buffers are used

(Romieu-Mourez et al, 2003; Wagner et al, 2003), it would still have been preferable to do epithelial

cell extraction and lyse the cells in 6xSDS lysis buffer or in 1x RIPA and sonicated before running

the Western blot. Along the same line, one should note that while the tumor tissues are mostly

comprised of epithelial mass, the tissues obtained from the WT mouse was the whole mammary

gland, which contains both epithelial and stromal components. Therefore, as well as having more WT

tissue samples, there needs to be a better extraction of mammary epithelial cells for it to be a more

appropriate control.

In addition to what was found in this study, there should be additional analyses of clinical

human breast tissues. Correlation or association studies, such as RT-qPCR analyses for PHIP1

mRNA levels on human breast tissues from various clinical stages (from premalignant to advanced)

may provide better insight into at what stage PHIP1 is more intensely present during the tumor

development, if a trend is present. Also, looking into gene amplification of human PHIP gene using

Southern blot or fluorescent in situ hybridization (FISH) as part of an epidemiological study or

Page 59: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

49

prognostic evaluations may increase its clinical value and potential widespread use as a diagnostic

tool. Combining multiple approaches will better consolidate or confirm the finding that PHIP1 is

specifically upregulated in human breast cancer. For example, an alternative approach to delineate

the expression of PHIP among different breast cancer types includes performing Tumor microarray

(TMA) of breast tumors at various clinical stages. However, there may be a few limitations to this

approach due to the inherent nature of detecting protein, which is the very final product of a gene

expression. For example, even if the intensity of the PHIP staining is more robust and disorganized in

human breast tumor samples per unit area, they still do not inform us of whether the strong staining

was the result of another oncogenic event that led to excessive proliferation and disorganization of

the epithelial cells that already expressed PHIP at high levels before any transformation occurred or

whether the overexpression of PHIP had a role in the tumor initiation. Additionally, the PHIP

antibody has been shown multiple times on Western blot to detect both the 206 kDa isoform and

another around 100 kDa. Therefore, it is possible that the results from the staining do not correspond

to the endogenous, full-length wild-type PHIP1 under investigation. The signal might originate from

mutated, truncated or stabilized PHIP with intact BD domain, which is the target of the antibody.

Screening the established mammary epithelial cell lines that cover characteristic stages of the

disease progression is another option to support the data from the clinical samples. However, it

should be noted that a major limitation in breast cancer research is the lack of multiple control cell

lines. One of the most prominently used non-tumorigenic control cell is the MCF10A, which is a

spontaneously immortalized human mammary epithelial cell line originated from the breast tissue of

a 36-year-old patient with fibrocytic changes (Soule et al, 1990). It displays many physiological

characteristics of the non-diseased breast, such as the requirement of growth factors for survival,

anchorage-dependent growth in culture, and lack of tumorigenicity in nude mice (Soule et al, 1990).

A more significant attribute of the MCF10A line in the study of mammary development and

dysfunction is its ability to form 3D acinar structures in reconstituted basement membrane, or

Matrigel, that resemble the glandular epithelium seen in in vivo (Debnath et al, 2002; Muthuswamy

et al, 2001). Thus, despite a number of cytogenetic abnormalities such as deletion of p16 and p14ARF-

containing locus and amplification of c-myc, and the basal phenotype of MCF10A (being ER

negative) (Debnath et al, 2003), MCF10A is widely used as the “normal” cell line for the functional

analyses of numerous breast cancer-related oncogenes. One recent example would be the generation

and study of stable MCF10A cell lines that stably overexpress constitutively active IGF-IR (Kim et

al, 2007) as mentioned in the Introduction. While it may be feasible to make sound conclusions

on the effect of an oncogene if the expression levels significantly differed between vector and

Page 60: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

50

transgenic cell lines, the expressive pattern of a protein from comparing multiple breast cancer cell

lines to only one or two controls might not be as informative if the levels were not visibly distinct

between normal and diseased samples. For example, we were initially excited to find that PHIP1

levels were significantly higher in the stable cell lines that overexpressed components of the IGF-IR

signaling pathway, when compared to the vector control (Figure 6C). As PHIP1 was previously

found to be a downstream positive effector of the IGF-I signaling pathway, it well supported our

hypothesis that PHIP may be involved in the IGF-IR-induced transformation. However, this

observation could not be replicated, and the PHIP levels between MCF10A parental cells and MCF7

or MDA-231 were also almost indistinguishable on a number of Western blots. Therefore, there

needs to be a larger sample size of currently used breast cancer cell lines as well as data from clinical

human breast cancer samples in order to confidently make the claim that PHIP1 is differentially

expressed in cancer compared to the normal state.

Additionally, it should be noted that cyclin D2, a transcriptional target of PHIP, is

downregulated in breast cancer, evidenced both by Evron et al (2001) and ONCOMINE, which is a

widely used bioinformatic resource among many cancer researchers (Rhodes et al, 2004). In at least

four different studies preceding the analyses by Evron et al, cyclin D2 was found to be abundantly

present in HMECs which had limited life span, whereas it was noted to be absent in almost all breast

cancer cell lines. Comprehensive series of reverse transcription (RT)-PCR confirmed that cyclin D2

mRNA level was lost in breast cancer cell lines as well as primary breast carcinomas when compared

to HMEC, and Western blot demonstrated that cyclin D2 is also lost at the protein level in primary

breast carcinomas (Evron et al, 2001). Interestingly, similar to PHIP, cyclin D2 was detected in both

luminal and myoepithelial cells obtained from normal mammoplasty specimens. The authors, more

importantly, showed that random methylation of the CpG-rich region of the cyclin D2 promoter in

breast cancer cells was associated with the loss of cyclin D2 expression in breast cancer, in contrast

to the expectation that cyclin D2 will likely act as a positive growth regulator by accelerating the G1

to S phase transition, which is the case for gastric or germline tumors (Takano et al, 2000; Schmidt et

al, 2001). Indeed, hypermethylation of the CpG-rich regions on the cyclin D2 promoter occurred in

almost half of the primary breast carcinomas, and the treatment of MCF7 and MDA-MB-231 with

methyltransferase inhibitor 5-aza-dC or histone deacetylase inhibitor trichostatin-A (TSA) restored

the cyclin D2 expression. As cyclin D2 promoter hypermethylation was also observed in ductal

carcinoma in situ (DCIS), the authors suggested that loss of cyclin D2 expression may be an early

event in the transformation event that precede the malignant stage of an invasive carcinoma (Evron et

al, 2001).

Page 61: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

51

Therefore, in order to clarify this apparent discrepancy and to test whether wild-type PHIP

was directly involved in cellular transformation, we attempted to stably overexpress human PHIP in

the non-tumorigenic MCF10A cell line and aimed to study the changes in cell phenotype, which is

discussed further in the next section.

4.2 PHIP1 as a component of the nuclear proteasome and transcriptional machinery

In this study, we attempted to address the potential oncogenic property of PHIP1 by generating cell

lines that stably overexpressed wild-type (WT), full-length protein. Thus MCF10A cells were

infected with retrovirus, which stably incorporates the reversely transcribed DNA from its viral RNA

genome into the host chromatin, and achieved approximately twice the level of protein present in the

GFP control as semi-quantitatively determined by the immunoblot analysis. Despite the initial

promising results where colony formation was observed on the soft agar assay, the protein levels

soon diminished within 2 weeks of G418 selection, in contrast to the durable 3-4 fold expression in

the mRNA transcription indicated by the Real-Time quantitative PCR (RT-qPCR) and the non-fading

presence of GFP in the MIEV-infected control cell line. Upon treatment of the cells with MG132, a

26S-proteasome inhibitor, HA signal was at least partially recovered, suggesting that HA-PHIP1 may

be targeted for ubiquitination and proteasomal degradation. Infection with PHIP adenovirus, which

we used as an alternative, also failed to sustain sufficient levels of the protein for the soft agar colony

formation assay and the initial robust expression of HA-PHIP rapidly declined within a week. There

are a number of areas of consideration that may explain this phenomenon, including possible

existence of PHIP autoregulation and protein stabilization of mutated or defective PHIP1.

An unexplored area of PHIP1 function in important cellular processes is in the

interconnection between the ubiquitin-proteasome pathway (UPP) and transcriptional complexes.

PHIP1 is also known as the DDB1 and CUL4-associated factor 14 (DCAF14), where DCAF denotes

a general group of WD40-containing proteins that act as substrate receptors within various DDB1-

CUL4 ubiquitin ligases (Lee and Zhou, 2007). The role of PHIP1 as a component of the E3 RING-

finger ubiquitin ligase is currently unknown. However, a number of speculations may be made based

on previous findings and proposed models that support the existence of nuclear proteasomes and the

involvement of proteasomal complexes in transcription activation and regulation.

Several studies involving immunolocalization and biochemical fractionation indicated that

proteasomes and their subunits were not only localized in the cytoplasm but also in the nucleus

(Adori et al, 2006; Lafarga et al, 2002; Brooks et al, 2000). Although the exact nuclear proteasomal

Page 62: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

52

content varied according to cell type, cell density and growth conditions, the nuclear ubiquitin-

proteasome system (nUPS) has been reported to degrade cell-cycle regulators, transcriptional factors

and tumor suppressors, such as p53 (Mikecz, 2006). The nUPS also plays important roles in nuclear

gene expression and quality-control. In 2003, Muratani and Tansey proposed a model for how the

Ub-proteasome system may control the transcriptional machinery. In their unified model, the

regulation of transcription by Ub-proteasome system occurs in a step-wise manner in a very short

period of time to inhibit any excess gene expression by an activator or the RNA polymerase II

complex (pol II). The authors proposed that the Ub-proteasome is also recruited to the promoter site,

probably mediated by direct binding to an activator or those of the basal transcriptional machinery.

The Ub-proteasome acts to ubiquitinate the transcriptional composite, such as the activator, pol II,

histones and other members, which subsequently attracts the components of the proteasome. Thus,

the interaction between pol II and proteasome only occurs when the activator and the transcriptional

machinery are bound to the promoter, and the degradation of the activator by the recruited

proteasome prohibits transcription re-initiation. It is possible that PHIP1 is involved in the process, as

summarized in Figure 17. The BD domain of PHIP may aid correct positioning of the co-activator by

binding to specific acetylated histone residues, while WD40 repeats act as protein scaffold. One

study validating this model has demonstrated that the steroid receptor coactivator (SRC) directly

interacts with LMP2 (Low Molecular mass Polypeptide 2), which is a component of catalytic β

subunit of the 20S proteasome, and that LMP2 is required throughout the estrogen receptor-mediated

gene transcription. Through chromatin immunoprecipitation (ChIP) assays using primers spanning

the whole genomic region of the ER promoter and the coding region of an ER target, the authors of

the study showed that LMP2 was recruited on the entire sequence of the target gene when

transcriptionally activated by the estradiol (E2) treatment. More intriguingly, sequential knockdown

of the transcription initiation complex and the time-course analyses showed the cyclic nature of

component recruitment and supported the requirement of SRC-mediated LMP2 recruitment in ER-

mediated gene transcription (Zhang et al, 2006).

Similar to the failure of stable protein expression of ectopic PHIP1 observed in retrovirally

infected MCF10A cells, lentivirally-infected GIPZ or TRIPZ-shRNA-PHIP1 knockdown in

Page 63: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

53

A.

C.B.

Figure 17. A model for role of PHIP1 as part of the ubiquitin-proteasome complex and in the

regulation of transcription initiation.

(A). The CUL4-DDB1 E3 Ubiquitin ligase ubiquitinates the activator, RNA pol II, histones and other TFs to

recruit the proteasomal complex. PHIP, or DCAF14, would act as a substrate receptor to aid correct

positioning of the complex subunits and specificity of the substrate. (B).The proteasomal complex degrades

activator, so that re-initiation of transcription does not occur. (C). The proteasomal complex moves along with

pol to modify chromatin structure and also disenages RNA pol II at the end of transcription or at the DNA

damage sites. Pictures modified from (Muratani and Tansey, 2003), and (Jackson and Xiong, 2009).

Page 64: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

54

MCF10A-CD8-IGF-IR cell line could not be sustained. Because we have no knowledge of how PHIP

is regulated, it is difficult to interpret why the overexpression or silencing could not be maintained,

especially when we had been able to successfully knockdown other proteins of interest studied in the

laboratory using the same GIPZ backbone vectors. Currently, there is no information on any existing

compensatory mechanism that responds to long-term reduction in PHIP1 levels. However, it could be

possible that PHIP1 is very tightly regulated similar to the autoregulative network of p53. The

autoregulation of PHIP1 might include a feedback loop where a protein under the control of PHIP1

acts as a sensor and leads to PHIP1 activation or degradation. Also, PHIP1 may directly or indirectly

transcriptionally activate the ubiquitin ligase complexes to its own destruction. Because PHIP1 does

not harbor a DNA-binding region, it probably would co-operate with an activator or repressor

transcription factors to regulate its target genes.

In this study, PHIP1 was found to complex with p53 in 293T cell model. The co-IP

experiment was repeated five times, but because the results were not consistent in all of the attempts,

despite the fact that the pull down of p53 never failed, identification and optimization of conditions

that are conducive to PHIP binding with p53 are needed to better understand the biological function

of PHIP within the context of cell cycle regulation and tumor progression. It could be that when the

co-IP did work, the cells were under an unknown stress (for example, excessive confluency and

serum-starvation). Also, the fact that HA-IP was not as easy to reproduce as p53 may be attributed to:

the relative abundance of the available epitope; the protein’s different sensitivity to proteases (as

changing the manufacturer for Protease Inhibitors Cocktail seemed to decrease the HA-IP

efficiency); or the different specificity and the affinity of the HA antibody compared to that of p53.

Other approaches to demonstrate that PHIP participates in transcriptional downregulation of p21,

would include luciferase assay with co-transfection of pGL3 luciferase reporter vector with p21-

responsive promoter or chromatin-immunoprecipitation (ChIP) using primers that span the p21

promoter site along with PHIP overexpression via adenovirus or transfection. These are subjected to

more in-depth analyses, which could not be performed in this study due to time limitations. However,

from the fact that PHIP1 can bind to p53, the PHIP1 regulatory network might include overlapping

components involved in the p53 autoregulation (reviewed in Lu, 2009), which could be another area

for investigation.

Moreover, data from large-scale screening such as mass spectrometry and cDNA microarray

analyses in the future would greatly aid in identifying gene targets of PHIP1 as well as suggesting its

binding partners, which could include proteasomal subunits that may play a role in the transcriptional

initiation and control of those target genes. The findings should be confirmed by

Page 65: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

55

immunoprecipitation assay and by the use of specific inhibitors such as lactacystin that blocks 20S

activity or silencing reagent such as siRNA for a particular protein component for the functional

analyses.

Interestingly, PHIP1 mutations in non-BRCA1/2 breast cancer have been reported in a Ph.D.

thesis from Karolinska Institutet, Stockholm, Sweden (Maguire, 2005, unpublished data). BRCA1/2

is the major genetic predisposition to breast cancer so far known. In the study, data from genome

wide linkage scan and cDNA microarray were screened and a 2.8Mb region of shared haplotype on

chromosome 6q14.1 was identified in a total of 31 families analyzed. The 2.8 Mb region contained

six known genes, one of which was PHIP. Of the 53 sequence alterations discovered in the six genes,

there were no frameshift or nonsense mutations, and PHIP gene was specifically found to contain a

missense mutation, Leu1093Pro. In support of PHIP1 mutation in breast cancer, Arason and

colleagues very recently performed genome-wide searches for linkage on 9 Icelandic and 13

additional Nordic multiple-case non-BRCA1/2 families to map highly penetrant loci. The three

highest signals were reported to be located at chromosomes 6q, 2p and 14q at respective

heterogeneity multipoint logarithm of odds (HLOD) scores in all families at 3.27, 1.66 and 1.24

(Arason et al, 2010). Thus, the authors concluded that Chromosomes 2p, 6q and 14q were candidate

sites for genes indicative of high risk for breast cancer and suggested a polygenic model of additive

probability with joint effect of the risk-factor genes (Arason et al, 2010).

Based on these results, it is possible that in a normal cell, such as MCF10A, the ectopically

expressed PHIP1 is also subjected to the autoregulatory mechanism as for the endogenous protein,

thus leading to its downregulation. Analogously, in cancerous state, this regulatory function may be

disrupted or evaded through PHIP1 mutation, leading to accumulation of potentially oncogenic

product that leads to uncontrolled cell proliferation. This may occur at the coding region or at the

post-translational level that results in the stabilization of protein at an active state. This prospect of

the existence of PHIP1 mutation in cancer is akin to p53, which was first discovered in SV40-

transformed cells in the late 1970s and was originally identified as an oncogene because it could

transform NIH/3T3 mouse fibroblasts similar to ras or myc (Weisz et al, 2007). However, p53 was

later re-classified as a tumor suppressor when only the mutated form was found to be transformative

(Finlay et al, 1989; Weisz et al, 2007). There is currently limited knowledge on PHIP mutational

status in cancer and it is another research area needing more thorough investigations.

Page 66: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

56

4.3 Role of PHIP1 in cell-cycle progression

The G1 to S phase transition in cell cycle progression is regulated by the D- and E-type cyclins with

their cyclin-dependent kinases CDK2, 3, and 4/6. The cyclin D-CDK4/6 complex leads to the

phosphorylation and inactivation of tumor suppressor Retinoblastoma (RB), which releases the E2F

transcription factors for the subsequent transcription of cyclin E and other genes that promote G1

progression and drive the DNA replication in the S phase. There are three types of the D-type

cyclins, cyclin D1, D2 and D3. Unlike the first two D-type cyclins, Cyclin D3 is strongly expressed

in benign tumors of steroid hormone secreting endocrine cells and is also implicated in pathogenesis

of malignant lymphoma (Doglioni et al, 1998; Bartkova et al, 1998). Additionally, cyclin D3 may

have a dual role in cell proliferation and terminal differentiation (Bartkova et al, 1994).

The current knowledge on the role of PHIP1 in cell cycle regulation is its ability to directly

and specifically induce cyclin D2 expression in INS-1 cells, an effect which was enhanced with IGF-

I stimulation (Podcheko et al, 2007). However, adenoviral overexpression of PHIP1 had no effect on

cellular cyclin D1 levels (Podcheko et al, 2007) and neither did the knockdown of PHIP1 in

MCF10A-CD8-IGF-IR or MDA-MB-231 as presented in this study. The prominent form of cyclin D

involved in breast cancer is cyclin D1, which has been long established to be a putative proto-

oncogene (Bartcova et al, 1994; Wang et al, 1994; Weinstat-Saslow et al, 1995; Steeg and Zhou,

1998). It has been previously shown that ablation of cyclin D1 in vivo leads to specific protection

against cancers induced by neu and ras oncogenes while having no influence on the tumorigenesis

driven by c-myc or Wnt-1 (Yu et al, 2001). In the same study, cyclin D2 was upregulated in the

mammary tumors of MMTV-Myc and MMTV-Wnt females (Yu et al, 2001). The fact that PHIP1

downregulation had no effect on cyclin D1 levels unlike cyclin D2 in MCF10A-IGFR cell model

might suggest that PHIP1 works more distinctively with pathways that activate cyclin D2, such as the

Myc- or Wnt-1-mediated signaling.

Cyclin D2 is transcriptionally regulated by the Myc/Max/Mad network (Bouchard et al,

2001), a family of basic helix-loop-helix-leucine zipper (bHLH-LZ) proteins that activates cyclin E-

Cdk2 complex by sequestering p27Kip1 into cyclin D2-Cdk4 complexes (Bouchard et al, 1999). As

demonstrated by the oligonucleotide microarray analysis of Myc-induced genes in primary human

fibroblasts (Coller et al, 2000), as well as lack of proliferative response to Myc in cyclin D2

knockout cells, cyclin D2 is a crucial downstream effector of Myc (Bouchard et al, 1999). Myc

promotes cell cycle progression by activating the expression of cdc25A, Cdk4 and cyclins D2, -E,

and –A, while downregulating the expression of growth arrest genes such as p21, p27, p15 and gas1

Page 67: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

57

(Gartel and Shchors, 2003). Induction of cyclin D2 expression by Myc involves MycBoxII (within

the transactivation domain, or TAD)-mediated recruitment of TRRAP, a component of the

PCAF/GCN5 and TIP50 HAT complexes, and preferential histone H4 acetylation at a single

nucleosome (Bouchard et al, 2001). It could be that PHIP1, being also a positive transcriptional

regulator of cyclin D2, directly or indirectly contributes to this Myc-dependent control of cyclin D2

expression. PHIP1 might facilitate the recruitment or correct positioning of transcriptional machinery

after the acetylation of histone residues by the HAT complexes via the interaction with

bromodomains. Or, PHIP might be involved in the transcriptional activation by ubiquitinating the

Myc nuclear factor to enable the transcription of its target genes by Pol II as per model proposed by

Muratani and Tansey. Also, it has been previously shown that activation of PI3K/Akt and MAPK

pathways regulate the Myc-mediated transcription through the phosphorylation and degradation of its

alternative heterodimer binding partner Mad1 (Zhu et al, 2008). Mad1 competes with Myc for

binding with Max for heterodimerization, thus suppressing the Myc transcriptional activity, which

necessitates the obligatory binding partner Max (Zhu et al, 2008).

In MDA-MB-231, FACS analysis with PI staining indicated that the growth inhibition

observed was mainly due to growth arrest rather than increased apoptosis. In this study, PHIP1

knockdown was shown to significantly increase p21 mRNA level in two different cell lines, in

MDA-MB-231 and MCF10A-CD8-IGF-IR. This could indicate that PHIP1 acts to repress p21

expression in normal state through unknown mechanism. Although the main activation of p21

expression is p53, p21 can be transcriptionally regulated in either a p53-dependent or p53-

independent manner, when stimulated by different agents, such as phorbol ester (PMA), transforming

growth factor- β (TGF-β) or nerve growth factor (NGF) (Abbas and Dutta, 2009). Interestingly, p21

is also a direct repressive target of c-Myc, as demonstrated by the oligonucleotide microarray

analysis in primary human fibroblasts by Coller et al (Coller et al, 2000). Additionally, Gartel and

colleagues showed that c-Myc repressed p21 at the promoter region 119 bp upstream of the

transcription start site, in common with the Sp1 binding sites (Gartel et al, 2001). Because Myc was

shown to interact with the C-terminal zinc finger domain of Sp1, which is required in the

transcription of p21, Myc likely downregulates p21 expression in part by sequestering Sp1 (Gartel et

al, 2001). Intriguingly, c-Myc and p21 has also been shown to reciprocally regulate each other

through protein-protein interaction (Kitaura et al, 2000). p21 can block c-Myc-Max complex

formation on the E-box by binding to the Myc Box II region, while binding of c-Myc to the C-

terminal region of p21 partially rescued the DNA polymerase δ-PCNA activity that is inhibited by

p21 (Kitaura et al, 2000). Therefore, in the context of breast cancer, it may be that upregulation of

Page 68: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

58

p21 from knockdown of PHIP1 leads to inhibition of c-Myc activity and thus the downregulation of

cyclin D2. Given the overlapping gene targets of c-Myc with that of PHIP1, it will be interesting to

see whether c-Myc requires the presence of PHIP for its normal function.

The intricate regulation of Myc by the proteasomal degradation is another area that may

involve PHIP1. It has long been recognized that the two highly conserved regions on the N-terminal

region of Myc protein family, called the Myc Boxes I and II, provide the signals for protein

degradation via the proteasome (Flinn et al, 1998). It has been previously shown that when treated

with proteasome inhibitors, c-Myc stably associates and accumulates in the nucleoli, which might be

the site for sequestrating and degrading excess c-Myc (Arabi et al, 2003). Similarly, there are a

number of ubiquitin ligases known to regulate c-Myc activity such as SCFSkp2 (von der Lehr et al,

2003; Kim et al, 2003), SCFFbw7 (Moberg et al, 2004; Yada et al, 2004; Welcker et al, 2004), and

HectH9 (Adhikary et al, 2005). CUL4-DDB1 E3 ubiquitin ligase has also emerged as a novel

regulatory mechanism of Myc activity in 2010 by Choi et al. The authors identified a new substrate

receptor in a mass spectrometry of Flag-tagged Myc in HeLa cells called TRPC4AP (transient

receptor potential cation channel, subfamily C, member 4-associated protein)/TRUSS (tumor

necrosis factor receptor-associated ubiquitous scaffolding and signaling protein), which complexed

with CUL4-DDB1 E3 ligase to selectively degrade Myc through the proteasome (Choi et al, 2010).

However, though TRPC4AP/TRUSS negated Myc function in this case, one should not rush to

conclude that the same would apply for PHIP1. The mechanisms of Myc regulation are complex and

vary depending on the cellular context. There are cases of dual roles of the same protein, such as

p300/CBP, which usually acts as a co-activator of Myc transcriptional targets but under certain

conditions, such as inhibition of cellular HDAC, leads to Myc degradation via acetylation of several

lysine residues between TAD and DNA-binding domain (Faiola et al, 2005).

Along with the idea that PHIP1 may cooperate with Myc for its function, another area that

needs further investigation is the role of PHIP1 in three dimensional (3D) culture system, which

more closely mimics the physiological microenvironment of the mammary gland than the plastic dish

(Vargo-Gogola and Rosen, 2007). As mentioned previously, single MCF10A cells undergo acinar

morphogenesis into a 3D structure when embedded in Matrigel. The cells initially proliferate (Day 1-

8) and then organize into apico-basally polarized epithelial cells that encircle a central hollow lumen

formed by apoptosis due to the differential action of serine threonine kinase Akt in the two

populations (Day 6-9). This can well be performed using the MCF10A cells cultured on Matrigel

(Debnath et al, 2002; Debnath et al, 2003). There have been examples where the activity of a protein

differs drastically when cells are embedded in extracellular matrix compared to a monolayer. For

Page 69: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

59

example, while long-term activation of Myc in MCF10A did not induce apoptosis when grown in a

monolayer, more than 60% cell death was observed within acini (Zhan et al, 2008). Also, a subset of

genes was found to be alternatively spliced in cells growth under 3D culture conditions compared to

2D on oligonucleotide microarray, suggesting that the cell geometry can influence the

posttranscriptional gene regulation (Li et al, 2006). Therefore, it would be interesting to see the

function of PHIP1 in 3D contexts which more closely resemble the physiological environment.

Additionally, because the repressive function of Myc may be more dominant in some cellular context

(Zhan et al, 2008; Evan et al, 1992, Nilsson et al, 2003), a concurrent activation of anti-apoptosis

response may be required for the Myc-driven transformation (Zhan et al, 2008). It could be this cell

survival signaling PHIP1 is responsible for during the tumor initiation.

4.4 Putting PHIP1 in the context of breast cancer and IGF-IR signaling

As mentioned in the Introduction, insulin-like growth factor 1 receptor (IGF-IR) is highly implicated

in breast cancer, and is involved in virtually all stages of tumor development by providing the

proliferative, anti-apoptotic and pro-metatastatic signals. For example, early studies indicated that in

malignant breast cancer, IGF-IRs are overexpressed approximately 14-fold, with 2-4 fold higher

levels of autophosphorylation compared to the normal tissue controls. When combining the increased

receptor function with increased protein expression, the overall IGF-IR activity could mount up to

40-fold elevation in cancer (Resnik et al, 1998). However, increasing evidence has suggested that

IGF-IR can have different cellular effects at various cancer stages. Schnarr et al (2000), for example,

observed that IGF-IR and IRS-1 were downregulated in cells that were poorly differentiated, which

represent cells of advanced stages of human breast cancer. In fact, the authors found that IGF-IR and

IRS-1 expressions correlated better with tumor progression than loss of the ER expression (Schnarr

et al, 2000). Similarly, as demonstrated by Bartucci and co-workers (2001), IGF-IR plays different

roles between ER positive and ER negative cell lines. While IGF-I could act as a chemoattractant for

both cell types and induce cell motility, it was only found to be mitogenic and anti-apoptotic in ER

positive cell lines (Bartucci et al, 2001). Currently, the consensus is that IGF-IR can have different

cellular effects depending on its signaling through IRS-1 or IRS-2, where IRS-1 has a proliferative

function and IRS-2 has a more metastatic role (Denley et al, 2007; Deepali, 2008). This is supported

by a number of studies which demonstrated that overexpression of IRS-1 or IGF-IR reduced estrogen

growth requirements and enhanced survival (Surmacz, 1995; Guvakova et al, 1997) and that in ER

negative cell line, IRS-1 overexpression is not sufficient to enhance IGF-I mediated cell proliferation

(Jackson et al, 1999). One of reports supporting the role of IRS-2 in cell motility is by Zhang et al

Page 70: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

60

(2004), which revealed greater phosphorylation of IRS-2 than IRS-1 during IGF-I-mediated cell

motility, and co-localization of IRS-2 with focal adhesion kinase (FAK) in the leading edge of

filopodia of motile cells.

The key finding in this study was that PHIP1 knockdown could significantly inhibit the

growth of MCF10A-CD8-IGF-IR. CD8-IGF-IR is constitutively active regardless of the presence of

the IGF-I ligand, and is composed of human T-cell CD8α extracellular domain fused with the IGF-IR

β cytoplasmic subunit (Carboni et al, 2005; Kim et al, 2007). It has been previously demonstrated

that CD8-IGF-IR is highly oncogenic, where MMTV transgenic mice overexpressing CD8-IGF-IR

develop salivary and mammary tumors as early as 8 weeks of age (Carboni et al, 2005). Similarly,

stable CD8-IGF-IR overexpression in MCF10A was sufficient to cause growth factor-independent

growth, colony formation on the soft agar, and tumor formation in immunocompromised mice at Day

15 when injected orthotopically to the mouse mammary gland (Kim et al, 2007). More importantly,

overexpression of CD8-IGF-IR disrupted the formation of normal MCF10A cell mammary acini

when grown in 3D Matrigel, resulting in the filling of the luminal space due to cell hyperproliferation

and inhibited apoptosis. Additionally, the authors provided the evidence that upregulation of

constitutively active CD8-IGF-IR could lead to direct downregulation of epithelial cell markers such

as E-cadherin and α-catenin and upregulation mesenchymal markers such as vimentin, fibronectin

and α-SMA, thus inducing epithelial-to-mesenchymal transition (EMT) (Kim et al, 2007), which

supported the fact that IGF-IR could be involved in every stage of breast tumor development, from

the initial oncogenic transformation to tumor dissemination. The fact that PHIP knockdown inhibited

the growth of highly aggressive cell line is a very exciting finding, as it indicates that IGF-IR

necessitates the presence of PHIP1 for its cell proliferative function. This notion would have been

strengthened if the growth characteristic of MCF10A-vector cell transfected with siScrambled had

been also included in the same growth curve, as this will validate the CD8-IGF-IR-mediated

enhanced cell proliferation under reduced serum conditions as previously reported by Kim and

colleagues, as well as showing whether PHIP1 knockdown alone could completely rescue the

hyperproliferative phenotype of CD8-IGF-IR overexpression into vector-like growth pattern. As a

downstream regulator of IGF-IR pathway, it is possible that the role of PHIP1 also differs depending

on the growth factor receptor status and the stage of tumor development metastatic stage.

Additionally, performing the proliferation assay with MCF10A stable cell lines which overexpress

IRS-1 or IRS-2 and comparing the data between the two cell lines may provide an insight into

whether PHIP works downstream of any particular IRS and if so, what specific role it plays when

activated by the IGF-IR.

Page 71: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

61

Lastly, from the fact that PHIP1 is involved in CD8-IGF-IR-mediated cell proliferation and

transcriptional regulation of cyclin D2, PHIP1 may also function to potentiate the synergistic effect

of different mitogens, such as estrogen, which enhances the IGF-I-mediated upregulation of type D

cyclins and cyclin E (Dupont et al, 2000). Moreover, there have been a number of studies that

illustrate how different mitotic signaling pathways can cooperate, despite recruiting and activating

different docking proteins. Fleming and colleagues, for instance, showed that although the degrees of

overlap vary in different cell lines, the distinct upstream signaling molecules recruited by IGF-IR and

EGFR are recruited to both enhance Akt activity (Fleming et al, 2006). Because PHIP1 was

previously shown to induce Akt phosphorylation via unknown mechanism (Podcheko et al, 2007), an

indirect modulation of Akt activity by PHIP1 might be another point of cross-talk between two

growth factor pathways. It has been well established that the estrogen pathway synergistically elicits

mitogenic responses by interacting with the insulin/IGF-I pathway in breast epithelial cells at various

crossing points such as induction of IRS1 expression of estrogen and binding of the ER with p85

subunit of the PI3K and tyrosine kinase Src (Zhang et al, 2005; Bernard et al, 2006) or the more

aggressive tumor formation through the synergist effect of cyclin D1 and c-Myc (Yong et al, 2007).

Page 72: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

62

References

Abbas, T., & Dutta, A. (2009). P21 in cancer: Intricate networks and multiple activities. Nature Reviews Cancer, 9(6), 400-414.

Adhikary, S., Marinoni, F., Hock, A., Hulleman, E., Popov, N., Beier, R., et al. (2005). The ubiquitin ligase HectH9 regulates transcriptional activation by myc and is essential for tumor cell proliferation. Cell, 123(3), 409-421.

Ádori, C., Low, P., Moszkovkin, G., Bagdy, G., László, L., & Kovács, G. G. (2006). Subcellular distribution of components of the ubiquitin-proteasome system in non-diseased human and rat brain. Journal of Histochemistry and Cytochemistry, 54(2), 263-267.

Aktas, H., Cai, H., & Cooper, G. M. (1997). Ras links growth factor signaling to the cell cycle machinery via regulation of cyclin D1 and the cdk inhibitor p27(KIP1). Molecular and Cellular Biology, 17(7), 3850-3857.

Allred, D. C., Mohsin, S. K., & Fuqua, S. A. W. (2001). Histological and biological evolution of human premalignant breast disease. Endocrine-Related Cancer, 8(1), 47-61.

Ande, S. R., Chen, J., & Maddika, S. (2009). The ubiquitin pathway: An emerging drug target in cancer therapy. European Journal of Pharmacology, 625(1-3), 199-205.

Andre, F., & Pusztai, L. (2006). Molecular classification of breast cancer: Implications for selection of adjuvant chemotherapy.3(11), 632.

Angers, S., Li, T., Yi, X., MacCoss, M. J., Moon, R. T., & Zheng, N. (2006). Molecular architecture and assembly of the DDB1-CUL4A ubiquitin ligase machinery. Nature, 443(7111), 590-593.

Arabi, A., Rustum, C., Hallberg, E., & Wright, A. P. H. (2003). Accumulation of c-myc and proteasomes at the nucleoli of cells containing elevated c-myc protein levels. Journal of Cell Science, 116(9), 1707-1717.

Aranda, V., Haire, T., Nolan, M. E., Calarco, J. P., Rosenberg, A. Z., Fawcett, J. P., et al. (2006). Par6-aPKC uncouples ErbB2 induced disruption of polarized epithelial organization from proliferation control. Nature Cell Biology, 8(11), 1235-1245.

Arason, A., Gunnarsson, H., Johannesdottir, G., Jonasson, K., Bendahl, P., Gillanders, E., et al. (2010). Genome-wide search for breast cancer linkage in large icelandic non-BRCA1/2 families. Breast Cancer Research, 12(4), R50.

Bargmann, C. I., Hung, M., & Weinberg, R. A. (1986). Multiple independent activations of the neu oncogene by a point mutation altering the transmembrane domain of p185. Cell, 45(5), 649-657.

Bartkova, J., Lukas, J., Strauss, M., & Bartek, J. (1998). Cyclin D3: Requirement for G1/S transition and high abundance in quiescent tissues suggest a dual role in proliferation and differentiation. Oncogene, 17(8), 1027-1037.

Bartucci, M., Morelli, C., Mauro, L., Ando', S., & Surmacz, E. (2001). Differential insulin-like growth factor I receptor signaling and function in estrogen receptor (ER)-positive MCF-7 and ER-negative MDA-MB-231 breast cancer cells. Cancer Research, 61(18), 6747-6754.

Bernard, L., Legay, C., Adriaenssens, E., Mougel, A., & Ricort, J. (2006). Estradiol regulates the insulin-like growth factor-I (IGF-I) signalling pathway: A crucial role of phosphatidylinositol 3-kinase (PI 3-kinase) in estrogens requirement for growth of MCF-7 human breast carcinoma cells. Biochemical and Biophysical Research Communications, 350(4), 916-921.

Page 73: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

63

Bissell, M. J., Radisky, D. C., Rizki, A., Weaver, V. M., & Petersen, O. W. (2002). The organizing principle: Microenvironmental influences in the normal and malignant breast. Differentiation, 70(9-10), 537-546.

Bouchard, C., Dittrich, O., Kiermaier, A., Dohmann, K., Menkel, A., Eilers, M., et al. (2001). Regulation of cyclin D2 gene expression by the Myc/Max/Mad network: Myc-dependent TRRAP recruitment and histone acetylation at the cyclin D2 promoter. Genes and Development, 15(16), 2042-2047.

Bouchard, C., Thieke, K., Maier, A., Saffrich, R., Hanley-Hyde, J., Ansorge, W., et al. (1999). Direct induction of cyclin D2 by myc contributes to cell cycle progression and sequestration of p27. EMBO Journal, 18(19), 5321-5333.

Bouchard, L., Lamarre, L., Tremblay, P. J., & Jolicoeur, P. (1989). Stochastic appearance of mammary tumors in transgenic mice carrying the MMTV/c-neu oncogene. Cell, 57(6), 931-936.

Brooks, A. R., Shiftman, D., Chan, C. S., Brooks, E. E., & Milner, P. G. (1996). Functional analysis of the human cyclin D2 and cyclin D3 promoters. Journal of Biological Chemistry, 271(15), 9090-9099.

Brooks, P., Fuertes, G., Murray, R. Z., Bose, S., Knecht, E., Rechsteiner, M. C., et al. (2000). Subcellular localization of proteasomes and their regulatory complexes in mammalian cells. Biochemical Journal, 346(1), 155-161.

Cardoso, R., Durbecq, V., Sotiriou, C., & Ross, J. S. (2005). HER-2/neu Gene and protein in breast cancer. In J. S. Ross, & G. N. Hortobagyi (Eds.), Molecular oncology of breast cancer (pp. 232-255). Canada: Jones and Bartlett Publishers.

Chen, C., Seth, A. K., & Aplin, A. E. (2006). Genetic and expression aberrations of E3 ubiquitin ligases in human breast cancer. Molecular Cancer Research, 4(10), 695-707.

Chen, L., Manjeshwar, S., Lu, Y., Moore, D., Ljung, B., Kuo, W., et al. (1998). The human homologue for the caenorhabditis elegans cul-4 gene is amplified and overexpressed in primary breast cancers. Cancer Research, 58(16), 3677-3683.

Chitnis, M. M., Yuen, J. S. P., Protheroe, A. S., Pollak, M., & Macaulay, V. M. (2008). The type 1 insulin-like growth factor receptor pathway. Clinical Cancer Research, 14(20), 6364-6370.

Choi, S. H., Wright, J. B., Gerber, S. A., & Cole, M. D. (2010). Myc protein is stabilized by suppression of a novel E3 ligase complex in cancer cells. Genes and Development, 24(12), 1236-1241.

Coller, H. A., Grandori, C., Tamayo, P., Colbert, T., Lander, E. S., Eisenman, R. N., et al. (2000). Expression analysis with oligonucleotide microarrays reveals that MYC regulates genes involved in growth, cell cycle, signaling, and adhesion. Proceedings of the National Academy of Sciences of the United States of America, 97(7), 3260-3265.

Dai, M., Jin, Y., Gallegos, J. R., & Lu, H. (2006). Balance of yin and yang: Ubiquitylation-mediated regulation of p53 and c-myc. Neoplasia, 8(8), 630-644.

Dankort, D., Maslikowski, B., Warner, N., Kanno, N., Kim, H., Wang, Z., et al. (2001). Grb2 and shc adapter proteins play distinct roles in neu (ErbB-2)-induced mammary tumorigenesis: Implications for human breast cancer. Molecular and Cellular Biology, 21(5), 1540-1551.

Davis, H. E., Morgan, J. R., & Yarmush, M. L. (2002). Polybrene increases retrovirus gene transfer efficiency by enhancing receptor-independent virus adsorption on target cell membranes. Biophysical Chemistry, 97(2-3), 159-172.

Page 74: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

64

D'Cruz, C. M., Gunther, E. J., Boxer, R. B., Hartman, J. L., Sintasath, L., Moody, S. E., et al. (2001). c-MYC induces mammary tumorigenesis by means of a preferred pathway involving spontaneous Kras2 mutations. Nature Medicine, 7(2), 235-239.

Debnath, J., Mills, K. R., Collins, N. L., Reginato, M. J., Muthuswamy, S. K., & Brugge, J. S. (2002). The role of apoptosis in creating and maintaining luminal space within normal and oncogene-expressing mammary acini. Cell, 111(1), 29-40.

Debnath, J., Muthuswamy, S. K., & Brugge, J. S. (2003). Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods, 30(3), 256-268.

Deepali, S. (2008). Regulation of breast cancer metastasis by IGF signaling. Journal of Mammary Gland Biology and Neoplasia, 13, 431-441.

Denley, A., Carroll, J. M., Brierley, G. V., Cosgrove, L., Wallace, J., Forbes, B., et al. (2007). Differential activation of insulin receptor substrates 1 and 2 by insulin-like growth factor-activated insulin receptors. Molecular and Cellular Biology, 27(10), 3569-3577.

Dhalluin, C., Carlson, J. E., Zeng, L., He, C., Aggarwal, A. K., & Zhou, M. (1999). Structure and ligand of a histone acetyltransferase bromodomain. Nature, 399(6735), 491-496.

Dickson, C., Creer, A., & Fantl, V. (2000). Mammary gland oncogenes as indicators of pathways important in mammary gland development. Oncogene, 19(8), 1097-1101.

Doglioni, C., Chiarelli, C., Macrí, E., Dei Tos, A. P., Meggiolaro, E., Palma, P. D., et al. (1998). Cyclin D3 expression in normal, reactive and neoplastic tissues. Journal of Pathology, 185(2), 159-166.

Dufourny, B., Alblas, J., van Teeffelen, H. A. A. M., van Schaik, F. M. A., van der Burg, B., Steenbergh, P. H., et al. (1997). Mitogenic signaling of insulin-like growth factor I in MCF-7 human breast cancer cells requires phosphatidylinositol 3-kinase and is independent of mitogen-activated protein kinase. Journal of Biological Chemistry, 272(49), 31163-31171.

Dupont, J., Karas, M., & LeRoith, D. (2003). The cyclin-dependent kinase inhibitor p21CIP/WAF is a positive regulator of insulin-like growth factor I-induced cell proliferation in MCF-7 human breast cancer cells. Journal of Biological Chemistry, 278(39), 37256-37264.

Dupont, J., & Le Roith, D. (2001). Insulin-like growth factor 1 and oestradiol promote cell proliferation of MCF-7 breast cancer cells: New insights into their synergistic effects. Molecular Pathology, 54(3), 149-154.

Dupont, J., Dunn, S. E., Barrett, J. C., & LeRoith, D. (2003). Microarray analysis and identification of novel molecules involved in insulin-like growth factor-1 receptor signaling and gene expression. Recent Progress in Hormone Research, 58(1), 325-342.

Dupont, J., Karas, M., LeRoith, D. (2000). The potentiation of estrogen on insulin-like growth factor I action in MCF-7 human breast cancer cells includes cell cycle components. Journal of Biochemical Chemistry, 275(46), 35893-35901.

Evan, G. I., Wyllie, A. H., Gilbert, C. S., Littlewood, T. D., Land, H., Brooks, M., et al. (1992). Induction of apoptosis in fibroblasts by c-myc protein. Cell, 69(1), 119-128.

Evron, E., Umbricht, C. B., Korz, D., Raman, V., Loeb, D. M., Niranjan, B., et al. (2001). Loss of cyclin D2 expression in the majority of breast cancers is associated with promoter hypermethylation. Cancer Research, 61(6), 2782-2787.

Page 75: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

65

Faiola, F., Liu, X., Lo, S., Pan, S., Zhang, K., Lymar, E., et al. (2005). Dual regulation of c-myc by p300 via acetylation-dependent control of myc protein turnover and coactivation of myc-induced transcription. Molecular and Cellular Biology, 25(23), 10220-10234.

Fantozzi, A., & Christofori, G. (2006). Mouse models of breast cancer metastasis. Breast Cancer Research, 8(4)

Farhang-Fallah, J., Yin, X., Trentin, G., Cheng, A. M., & Rozakis-Adcock, M. (2000). Cloning and characterization of PHIP, a novel insulin receptor substrate-1 pleckstrin homology domain interacting protein. Journal of Biological Chemistry, 275(51), 40492-40497.

Farhang-Fallah, J., Randhawa, V. K., Nimnual, A., Klip, A., Bar-Sagi, D., & Rozakis-Adcock, M. (2002). The pleckstrin homology (PH) domain-interacting protein couples the insulin receptor substrate 1 PH domain to insulin signaling pathways leading to mitogenesis and GLUT4 translocation. Molecular and Cellular Biology, 22(20), 7325-7336.

Finlay, C. A., Hinds, P. W., & Levine, A. J. (1989). The p53 proto-oncogene can act as a suppressor of transformation. Cell, 57(7), 1083-1093.

Flinn, E. M., Busch, C. M. C., & Wright, A. P. H. (1998). Myc boxes, which are conserved in myc family proteins, are signals for protein degradation via the proteasome. Molecular and Cellular Biology, 18(10), 5961-5969.

Gartel, A. L., & Shchors, K. (2003). Mechanisms of c-myc-mediated transcriptional repression of growth arrest genes. Experimental Cell Research, 283(1), 17-21.

Gartel, A. L., Ye, X., Goufman, E., Shianov, P., Hay, N., Najmabadi, F., et al. (2001). Myc represses the p21 (WAF1/CIP1) promoter and interacts with Sp1/Sp3. Proceedings of the National Academy of Sciences of the United States of America, 98(8), 4510-4515.

Gartel, A. L., & Radhakrishnan, S. K. (2005). Lost in transcription: P21 repression, mechanisms, and consequences. Cancer Research, 65(10), 3980-3985.

Ghosh, P., Wu, M., Zhang, H., & Sun, H. (2008). mTORC1 signaling requires proteasomal function and the involvement of CUL4-DDB1 ubiquitin E3 ligase. Cell Cycle, 7(3), 373-381.

Guvakova, M. A., & Surmacz, E. (1997). Overexpressed IGF-I receptors reduce estrogen growth requirements, enhance survival, and promote e-cadherin-mediated cell-cell adhesion in human breast cancer cells. Experimental Cell Research, 231(1), 149-162.

Harris, L., Fritsche, H., Mennel, R., Norton, L., Ravdin, P., Taube, S., et al. (2007). American society of clinical oncology 2007 update of recommendations for the use of tumor markers in breast cancer. Journal of Clinical Oncology, 25(33), 5287-5312.

Hashemolhosseini, S., Nagamine, Y., Morley, S. J., Desrivières, S., Mercep, L., & Ferrari, S. (1998). Rapamycin inhibition of the G1 to S transition is mediated by effects on cyclin D1 mRNA and protein stability. Journal of Biological Chemistry, 273(23), 14424-14429.

Haynes, S. R., Dollard, C., Winston, F., Beck, S., Trowsdale, J., & Dawid, I. B. (1992). The bromodomain: A conserved sequence found in human, drosophila and yeast proteins. Nucleic Acids Research, 20(10), 2603.

Hennighausen, L., & Robinson, G. W. (2005). Information networks in the mammary gland. Nature Reviews Molecular Cell Biology, 6(9), 715-725.

Higa, L., & Zhang, H. (2007). Stealing the spotlight: CUL4-DDB1 ubiquitin ligase docks WD40-repeat proteins to destroy. Cell Division, 2

Page 76: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

66

Higa, L. A., Wu, M., Ye, T., Kobayashi, R., Sun, H., & Zhang, H. (2006). CUL4-DDB1 ubiquitin ligase interacts with multiple WD40-repeat proteins and regulates histone methylation. Nature Cell Biology, 8(11), 1277-1283.

Huang, W., Chang, H. Y., Fei, T., Wu, H., & Chen, Y. (2007). GSK3β mediates suppression of cyclin D2 expression by tumor suppressor PTEN. Oncogene, 26(17), 2471-2482.

Hynes, N. E., & Lane, H. A. (2001). Myc and mammary cancer: Myc is a downstream effector of the ErbB2 receptor tyrosine kinase. Journal of Mammary Gland Biology and Neoplasia, 6(1), 141-150.

Jackson, S., & Xiong, Y. (2009). CRL4s: The CUL4-RING E3 ubiquitin ligases. Trends in Biochemical Sciences, 34(11), 562-570.

Jackson, J. G., & Yee, D. (1999). IRS-1 expression and activation are not sufficient to activate downstream pathways and enable IGF-I growth response in estrogen receptor negative breast cancer cells. Growth Hormone and IGF Research, 9(5), 280-289.

Jia, S., Kobayashi, R., & Grewal, S. I. S. (2005). Ubiquitin ligase component Cul4 associates with Clr4 histone methyltransferase to assemble heterochromatin. Nature Cell Biology, 7(10), 1007-1013.

Kim, H., Litzenburger, B. C., Cui, X., Delgado, D. A., Grabiner, B. C., Lin, X., et al. (2007). Constitutively active type I insulin-like growth factor receptor causes transformation and xenograft growth of immortalized mammary epithelial cells and is accompanied by an epithelial-to-mesenchymal transition mediated by NF-{kappa}B and snail. Molecular and Cellular Biology, 27(8), 3165-3175.

Kim, S. Y., Herbst, A., Tworkowski, K. A., Salghetti, S. E., & Tansey, W. P. (2003). Skp2 regulates myc protein stability and activity. Molecular Cell, 11(5), 1177-1188.

Kitaura, H., Shinshi, M., Uchikoshi, Y., Ono, T., Tsurimoto, T., Yoshikawa, H., et al. (2000). Reciprocal regulation via protein-protein interaction between c-myc and p21(cip1/waf1/sdi1) in DNA replication and transcription. Journal of Biological Chemistry, 275(14), 10477-10483.

Kroman, N., & Mouridsen, H. T. (2003). Prognostic influence of pregnancy before, around, and after diagnosis of breast cancer. Breast, 12(6), 516-521.

Lacroix, M., Toillon, R., & Leclercq, G. (2006). p53 and breast cancer, an update. Endocrine-Related Cancer, 13(2), 293-325.

Lafarga, M., Berciano, M. T., Pena, E., Mayo, I., Castaño, J. G., Bohmann, D., et al. (2002). Clastosome: A subtype of nuclear body enriched in 19S and 20S proteasomes, ubiquitin, and protein substrates of proteasome. Molecular Biology of the Cell, 13(8), 2771-2782.

Lavoie, J. N., L'Allemain, G., Brunei, A., Müller, R., & Pouysségur, J. (1996). Cyclin D1 expression is regulated positively by the p42/p44(MAPK) and negatively by the p38/HOG(MAPK) pathway. Journal of Biological Chemistry, 271(34), 20608-20616.

Lee, C., Park, K., Adachi, Y., Seol, J. Y., Yoo, C., Kim, Y. W., et al. (2003). Recombinant adenoviruses expressing dominant negative insulin-like growth factor-I receptor demonstrate antitumor effects on lung cancer. Cancer Gene Therapy, 10(1), 57-63.

Lee, C., Wu, S., Gabrilovich, D., Chen, H., Nadaf-Rahrov, S., Ciernik, I. F., et al. (1996). Antitumor effects of an adenovirus expressing antisense insulin-like growth: Factor I receptor on human lung cancer cell lines. Cancer Research, 56(13), 3038-3041.

Lee, J., & Zhou, P. (2007). DCAFs, the missing link of the CUL4-DDB1 ubiquitin ligase. Molecular Cell, 26(6), 775-780.

Page 77: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

67

Li, C., Kato, M., Shiue, L., Shively, J. E., Ares Jr., M., & Lin, R. (2006). Cell type and culture condition-dependent alternative splicing in human breast cancer cells revealed by splicing-sensitive microarrays. Cancer Research, 66(4), 1990-1999.

Li, T., Chen, X., Garbutt, K. C., Zhou, P., & Zheng, N. (2006). Structure of DDB1 in complex with a paramyxovirus V protein: Viral hijack of a propeller cluster in ubiquitin ligase. Cell, 124(1), 105-117.

Li, W., You, L., Cooper, J., Schiavon, G., Pepe-Caprio, A., Zhou, L., et al. (2010). Merlin/NF2 suppresses tumorigenesis by inhibiting the E3 ubiquitin ligase CRL4DCAF1 in the nucleus. Cell, 140(4), 477-490.

Liao, D. J., & Dickson, R. B. (2000). c-myc in breast cancer. Endocrine-Related Cancer, 7(3), 143-164.

Liao, G., Zhang, M., Harhaj, E. W., & Sun, S. (2004). Regulation of the NF-{kappa}B-inducing kinase by tumor necrosis factor receptor-associated factor 3-induced degradation. Journal of Biological Chemistry, 279(25), 26243-26250.

Loyola, A., & Almouzni, G. (2004). Bromodomains in living cells participate in deciphering the histone code. Trends in Cell Biology, 14(6), 279-281.

Maor, S., Yosepovich, A., Papa, M. Z., Yarden, R. I., Mayer, D., Friedman, E., et al. (2007). Elevated insulin-like growth factor-I receptor (IGF-IR) levels in primary breast tumors associated with BRCA1 mutations. Cancer Letters, 257(2), 236-243.

Marmorstein, R., & Berger, S. L. (2001). Structure and function of bromodomains in chromatin-regulating complexes. Gene, 272(1-2), 1-9.

Mawson, A., Lai, A., Carroll, J. S., Sergio, C. M., Mitchell, C. J., & Sarcevic, B. (2005). Estrogen and insulin/IGF-1 cooperatively stimulate cell cycle progression in MCF-7 breast cancer cells through differential regulation of c-myc and cyclin D1. Molecular and Cellular Endocrinology, 229(1-2), 161-173.

McCall, C. M., Hu, J., & Xiong, Y. (2005). Recruiting substrates to cullin 4-dependent ubiquitin ligases by DDB1. Cell Cycle, 4(1), 27-29.

McCall, C. M., Miliani de Marval, P. L., Chastain, P. D.,II, Jackson, S. C., He, Y. J., Kotake, Y., et al. (2008). Human immunodeficiency virus type 1 vpr-binding protein VprBP, a WD40 protein associated with the DDB1-CUL4 E3 ubiquitin ligase, is essential for DNA replication and embryonic development. Molecular and Cellular Biology, 28(18), 5621-5633.

Mikecz, A. (2006). The nuclear ubiquitin-proteasome system. Journal of Cell Science, 119(10), 1977-1984.

Mittnacht, S., Paterson, H., Olson, M. F., & Marshall, C. J. (1997). Ras signalling is required for inactivation of the tumour suppressor pRb cell-cycle control protein. Current Biology, 7(3), 219-221.

Moberg, K. H., Mukherjee, A., Veraksa, A., Artavanis-Tsakonas, S., & Hariharan, I. K. (2004). The drosophila F box protein archipelago regulates dMyc protein levels in vivo. Current Biology, 14(11), 965-974.

Moody, S. E., Perez, D., Pan, T., Sarkisian, C. J., Portocarrero, C. P., Sterner, C. J., et al. (2005). The transcriptional repressor snail promotes mammary tumor recurrence. Cancer Cell, 8(3), 197-209.

Mukherjee, S., & Conrad, S. E. (2005). C-myc suppresses p21WAF1/CIP1 expression during estrogen signaling and antiestrogen resistance in human breast cancer cells. Journal of Biological Chemistry, 280(18), 17617-17625.

Muller, W. J., Sinn, E., Pattengale, P. K., Wallace, R., & Leder, P. (1988). Single-step induction of mammary adenocarcinoma in transgenic mice bearing the activated c-neu oncogene. Cell, 54(1), 105-115.

Muratani, M., & Tansey, W. P. (2003). How the ubiquitin-proteasome system controls transcription.4(3), 201.

Page 78: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

68

Nelson, W. J. (2003). Adaptation of core mechanisms to generate cell polarity. Nature, 422(6933), 766-774.

Nilsson, J. A., & Cleveland, J. L. (2003). Myc pathways provoking cell suicide and cancer. Oncogene, 22(56 REV. ISS. 8), 9007-9021.

O'Connell, B. C., & Harper, J. W. (2007). Ubiquitin proteasome system (UPS): What can chromatin do for you? Current Opinion in Cell Biology, 19(2), 206-214.

Ogawara, Y., Kishishita, S., Obata, T., Isazawa, Y., Suzuki, T., Tanaka, K., et al. (2002). Akt enhances Mdm2-mediated ubiquitination and degradation of p53. Journal of Biological Chemistry, 277(24), 21843-21850.

Ohuchi, N., Abe, R., Takahashi, T., & Tezuka, F. (1984). Origin and extension of intraductal papillomas of the breast: A three-dimensional reconstruction study. Breast Cancer Research and Treatment, 4(2), 117-128.

Oliveira, A. M., & Fletcher, J. A. (2005). Oncogenes. In J. S. Ross, & G. N. Hortobagyi (Eds.), Molecular oncology of breast cancer (pp. 348-357). Canada: Jones and Bartlett Publishers, Inc.

Peeper, D. S., Upton, T. M., Ladha, M. H., Neuman, E., Zalvide, J., Bernards, R., et al. (1997). Ras signalling linked to the cell-cycle machinery by the retinoblastoma protein. Nature, 386(6621), 177-181.

Peyrat, J., Bonneterre, J., Beuscart, R., Djiane, J., & Demaille, A. (1988). Insulin-like growth factor 1 receptors in human breast cancer and their relation to estradiol and progesterone receptors. Cancer Research, 48(22), 6429-6433.

Podcheko, A., Northcott, P., Bikopoulos, G., Lee, A., Bommareddi, S. R., Kushner, J. A., et al. (2007). Identification of a WD40 repeat-containing isoform of PHIP as a novel regulator of {beta}-cell growth and survival. Molecular and Cellular Biology, 27(18), 6484-6496.

Qin, Y., Capaldo, C., Gumbiner, B. M., & Macara, I. G. (2005). The mammalian scribble polarity protein regulates epithelial cell adhesion and migration through E-cadherin. Journal of Cell Biology, 171(6), 1061-1071.

Reese, D. M., & Slamon, D. J. (1997). HER-2/neu signal transduction in human breast and ovarian cancer. Stem Cells, 15(1), 1-8.

Rennstam, K., Baldetorp, B., Kytölä, S., Tanner, M., & Isola, J. (2001). Chromosomal rearrangements and oncogene amplification precede aneuploidization in the genetic evolution of breast cancer. Cancer Research, 61(3), 1214-1219.

Resnik, J. L., Reichart, D. B., Huey, K., Webster, N. J. G., & Seely, B. L. (1998). Elevated insulin-like growth factor I receptor autophosphorylation and kinase activity in human breast cancer. Cancer Research, 58(6), 1159-1164

Rhodes, D. R., Yu, J., Shanker, K., Deshpande, N., Varambally, R., Ghosh, D., et al. (2004). ONCOMINE: A cancer microarray database and integrated data-mining platform. Neoplasia, 6(1), 1-6.

Riedemann, J., Sohail, M., & Macaulay, V. M. (2007). Dual silencing of the EGF and type 1 IGF receptors suggests dominance of IGF signaling in human breast cancer cells. Biochemical and Biophysical Research Communications, 355(3), 700-706.

Rosner, A., Miyoshi, K., Landesman-Bollag, E., Xu, X., Seldin, D. C., Moser, A. R., et al. (2002). Pathway pathology: Histological differences between ErbB/Ras and wnt pathway transgenic mammary tumors. American Journal of Pathology, 161(3), 1087-1097.

Samani, A. A., Yakar, S., LeRoith, D., & Brodt, P. (2007). The role of the IGF system in cancer growth and metastasis: Overview and recent insights. Endocrine Reviews, 28(1), 20-47.

Page 79: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

69

Schindl, M., Gnant, M., Schoppmann, S. F., Horvat, R., & Birner, P. (2007). Overexpression of the human homologue for caenorhabditis elegans cul-4 gene is associated with poor outcome in node-negative breast cancer. Anticancer Research, 27(2), 949-952.

Schnarr, B., Strunz, K., Ohsam, J., Benner, A., Wacker, J., & Mayer, D. (2000). Down-regulation of insulin-like growth factor-I receptor and insulin receptor substrate-1 expression in advanced human breast cancer. International Journal of Cancer, 89(6), 506-513.

Schoenenberger, C. A., Andres, A. C., Groner, B., van der Valk, M., LeMeur, M., & Gerlinger, P. (1988). Targeted c-myc gene expression in mammary glands of transgenic mice induces mammary tumours with constitutive milk protein gene transcription. EMBO Journal, 7(1), 169-175.

Shi, D., & Grossman, S. R. (2010). Ubiquitin becomes ubiquitous in cancer: Emerging roles of ubiquitin ligases and deubiquitinases in tumorigenesis and as therapeutic targets. Cancer Biology and Therapy, 10(8), 737-747.

Skowyra, D., Craig, K. L., Tyers, M., Elledge, S. J., & Harper, J. W. (1997). F-box proteins are receptors that recruit phosphorylated substrates to the SCF ubiquitin-ligase complex. Cell, 91(2), 209-219.

Slamon, D. J., Godolphin, W., Jones, L. A., Holt, J. A., Wong, S. G., Keith, D. E., et al. (1989). Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science, 244(4905), 707-712.

Sondek, J., Bohm, A., Lambright, D. G., Hamm, H. E., & Sigier, P. B. (1996). Crystal structure of a GA protein βγ dimer at 2.1Å resolution. Nature, 379(6563), 369-374.

Soule, H. D., Maloney, T. M., Wolman, S. R., Peterson, W. D., Jr., Brenz, R., McGrath, C. M., et al. (1990). Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Research, 50(18), 6075-6086.

Stampfer, M. R., & Yaswen, P. (1993). Culture systems for study of human mammary epithelial cell proliferation, differentiation and transformation. Cancer Surveys, 18, 7-34.

Steeg, P. S., & Zhou, Q. (1998). Cyclins and breast cancer. Breast Cancer Research and Treatment, 52(1-3), 17-28.

Stewart, T. A., Pattengale, P. K., & Leder, P. (1984). Spontaneous mammary adenocarcinomas in transgenic mice that carry and express MTV/myc fusion genes. Cell, 38(3), 627-637.

Surmacz, E. (2000). Function of the IGF-I receptor in breast cancer. Journal of Mammary Gland Biology and Neoplasia, 5(1), 95-105.

Surmacz, E., Guvakova, M. A., Nolan, M. K., Nicosia, R. F., & Sciacca, L. (1998). Type I insulin-like growth factor receptor function in breast cancer. Breast Cancer Research and Treatment, 47(3), 255-267.

Surmacz, E., & Burgaud, J. -. (1995). Overexpression of insulin receptor substrate 1 (IRS-1) in the human breast cancer cell line MCF-7 induces loss of estrogen requirements for growth and transformation. Clinical Cancer Research, 1(11), 1429-1436.

Sutherland, R. L., & Musgrove, E. A. (2004). Cyclins and breast cancer. Journal of Mammary Gland Biology and Neoplasia, 9(1), 95-104.

Swindle, C. S., Kim, H. G., & Klug, C. A. (2004). Mutation of CpGs in the murine stem cell virus retroviral vector long terminal repeat represses silencing in embryonic stem cells. Journal of Biological Chemistry, 279(1), 34-41.

Takano, Y., Kato, Y., van Diest, P. J., Masuda, M., Mitomi, H., & Okayasu, I. (2000). Cyclin D2 overexpression and lack of p27 correlate positively and cyclin E inversely with a poor prognosis in gastric cancer cases. American Journal of Pathology, 156(2), 585-594.

Page 80: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

70

Tamkun, J. W., Deuring, R., Scott, M. P., Kissinger, M., Pattatucci, A. M., Kaufman, T. C., et al. (1992). Brahma: A regulator of drosophila homeotic genes structurally related to the yeast transcriptional activator SNF2/SWI2. Cell, 68(3), 561-572.

Tao, Y., Pinzi, V., Bourhis, J., & Deutsch, E. (2007). Mechanisms of disease: Signaling of the insulin-like growth factor 1 receptor pathway - therapeutic perspectives in cancer.4(10), 602.

Tortora, G. J., & Derrickson, B. (2006). Principles of anatomy and physiology. p1083-1084. USA: John Wiley & Sons, Inc.

Valentinis, B., & Baserga, R. (2001). IGF-I receptor signalling in transformation and differentiation. Molecular Pathology, 54(3), 133-137.

Vargo-Gogola, T., & Rosen, J. M. (2007). Modelling breast cancer: One size does not fit all. Nature Reviews Cancer, 7(9), 659-672.

Varlakhanova, N. V., & Knoepfler, P. S. (2009). Acting locally and globally: Myc's ever-expanding roles on chromatin. Cancer Research, 69(19), 7487-7490.

von der Lehr, N., Johansson, S., & Larsson, L. G. (2003). Implication of the ubiquitin/proteasome system in myc-regulated transcription. Cell Cycle (Georgetown, Tex.), 2(5), 403-407.

Von Der Lehr, N., Johansson, S., Wu, S., Bahram, F., Castell, A., Cetinkaya, C., et al. (2003). The F-box protein Skp2 participates in c-myc proteosomal degradation and acts as a cofactor for c-myc-regulated transcription. Molecular Cell, 11(5), 1189-1200.

Wadman, I. A., Osada, H., Grütz, G. G., Agulnick, A. D., Westphal, H., Forster, A., et al. (1997). The LIM-only protein Lmo2 is a bridging molecule assembling an erythroid, DNA-binding complex which includes the TAL1, E47, GATA-1 and Ldb1/NLI proteins. EMBO Journal, 16(11), 3145-3157.

Waldman, T., Kinzler, K. W., & Vogelstein, B. (1995). p21 is necessary for the p53-mediated G1 arrest in human cancer cells. Cancer Research, 55(22), 5187-5190.

Wang, H., Zhai, L., Xu, J., Joo, H., Jackson, S., Erdjument-Bromage, H., et al. (2006). Histone H3 and H4 ubiquitylation by the CUL4-DDB-ROC1 ubiquitin ligase facilitates cellular response to DNA damage. Molecular Cell, 22(3), 383-394.

Wang, T. C., Cardiff, R. D., Zukerberg, L., Lees, E., Arnold, A., & Schmidt, E. V. (1994). Mammary hyperplasia and carcinoma in MMTV-cyclin D1 transgenic mice. Nature, 369(6482), 669-671.

Wang, Y., Thakur, A., Sun, Y., Wu, J., Biliran, H., Bollig, A., et al. (2007). Synergistic effect of cyclin D1 and c-myc leads to more aggressive and invasive mammary tumors in severe combined immunodeficient mice. Cancer Research, 67(8), 3698-3707.

Weigelt, B., & Bissell, M. J. (2008). Unraveling the microenvironmental influences on the normal mammary gland and breast cancer. Seminars in Cancer Biology, 18(5), 311-321.

Weinstat-Saslow, D., Merino, M. J., Manrow, R. E., Lawrence, J. A., Bluth, R. F., Wittenbel, K. D., et al. (1995). Overexpression of cyclin D mRNA distinguishes invasive and in situ breast carcinomas from non-malignant lesions. Nature Medicine, 1(12), 1257-1260.

Weisz, L., Oren, M., & Rotter, V. (2007). Transcription regulation by mutant p53. Oncogene, 26(15), 2202-2211.

Welcker, M., Orian, A., Grim, J. A., Eisenman, R. N., & Clurman, B. E. (2004). A nucleolar isoform of the Fbw7 ubiquitin ligase regulates c-myc and cell size. Current Biology, 14(20), 1852-1857.

Page 81: Investigating the Role of PHIP1 in Breast Cancer...ii Investigating the Role of PHIP1 in Breast Cancer Chan Mi Lee Master of Science Laboratory Medicine & Pathobiology University of

71

Welcker, M., Orian, A., Jin, J., Grim, J. A., Harper, J. W., Eisenman, R. N., et al. (2004). The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-myc protein degradation. Proceedings of the National Academy of Sciences of the United States of America, 101(24), 9085-9090.

Winston, J. T., Coats, S. R., Wang, Y., & Pledger, W. J. (1996). Regulation of the cell cycle machinery by oncogenic ras. Oncogene, 12(1), 127-134.

Wiseman, B. S., & Werb, Z. (2002). Development: Stromal effects on mammary gland development and breast cancer. Science, 296(5570), 1046-1049.

Xiao, Q., Ciaassen, G., Shi, J., Adachi, S., Sedivy, J., & Hann, S. R. (1998). Transactivation-defective c-MycS retains the ability to regulate proliferation and apoptosis. Genes and Development, 12(24), 3803-3808.

Yada, M., Hatakeyama, S., Kamura, T., Nishiyama, M., Tsunematsu, R., Imaki, H., et al. (2004). Phosphorylation-dependent degradation of c-myc is mediated by the F-box protein Fbw7. EMBO Journal, 23(10), 2116-2125.

Yu, Q., Geng, Y., & Sicinski, P. (2001). Specific protection against breast cancers by cyclin D1 ablation. Nature, 411(6841), 1017-1021.

Zhan, L., Rosenberg, A., Bergami, K. C., Yu, M., Xuan, Z., Jaffe, A. B., et al. (2008). Deregulation of scribble promotes mammary tumorigenesis and reveals a role for cell polarity in carcinoma. Cell, 135(5), 865-878.

Zhang, H., Sun, L., Liang, J., Yu, W., Zhang, Y., Wang, Y., et al. (2006). The catalytic subunit of the proteasome is engaged in the entire process of estrogen receptor-regulated transcription. EMBO Journal, 25(18), 4223-4233.

Zhang, S., Li, X., Burghardt, R., Smith III, R., & Safe, S. H. (2005). Role of estrogen receptor (ER) α in insulin-like growth factor (IGF)-I-induced responses in MCF-7 breast cancer cells. Journal of Molecular Endocrinology, 35(3), 433-447.

Zhang, X., Kamaraju, S., Hakuno, F., Kabuta, T., Takahashi, S., Sachdev, D., et al. (2004). Motility response to insulin-like growth factor-I (IGF-I) in MCF-7 cells is associated with IRS-2 activation and integrin expression. Breast Cancer Research and Treatment, 83(2), 161-170.

Zheng, N., Schulman, B. A., Song, L., Miller, J. J., Jeffrey, P. D., Wang, P., et al. (2002). Structure of the Cul1-Rbx1-Skp1-F boxSkp2 SCF ubiquitin ligase complex. Nature, 416(6882), 703-709.

Zhou, B. P., Liao, Y., Xia, W., Zou, Y., Spohn, B., & Hung, M. (2001). HER-2/neu induces p53 ubiquitination via akt-mediated MDM2 phosphorylation. Nature Cell Biology, 3(11), 973-982.

Zhu, J., Blenis, J., & Yuan, J. (2008). Activation of PI3K/Akt and MAPK pathways regulates myc-mediated transcription by phosphorylating and promoting the degradation of Mad1. Proceedings of the National Academy of Sciences of the United States of America, 105(18), 6584-6589.

Zou, Y., Mi, J., Cui, J., Lu, D., Zhang, X., Guo, C., et al. (2009). Characterization of nuclear localization signal in the N terminus of CUL4B and its essential role in cyclin E degradation and cell cycle progression. Journal of Biological Chemistry, 284(48), 33320-33332.