187
Loyola University Chicago Loyola University Chicago Loyola eCommons Loyola eCommons Dissertations Theses and Dissertations 2010 Polyomavirus Enhancer Activator 3 (PEA3), a Member of the Ets Polyomavirus Enhancer Activator 3 (PEA3), a Member of the Ets Family of Transcription Factors, Is a Transcriptional Activator of Family of Transcription Factors, Is a Transcriptional Activator of Notch-1 and Notch-4 in Breast Cancer: An Opportunity for Novel Notch-1 and Notch-4 in Breast Cancer: An Opportunity for Novel Combinational Therapy Combinational Therapy Anthony George Clementz Loyola University Chicago Follow this and additional works at: https://ecommons.luc.edu/luc_diss Part of the Biochemistry Commons Recommended Citation Recommended Citation Clementz, Anthony George, "Polyomavirus Enhancer Activator 3 (PEA3), a Member of the Ets Family of Transcription Factors, Is a Transcriptional Activator of Notch-1 and Notch-4 in Breast Cancer: An Opportunity for Novel Combinational Therapy" (2010). Dissertations. 81. https://ecommons.luc.edu/luc_diss/81 This Dissertation is brought to you for free and open access by the Theses and Dissertations at Loyola eCommons. It has been accepted for inclusion in Dissertations by an authorized administrator of Loyola eCommons. For more information, please contact [email protected]. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 3.0 License. Copyright © 2010 Anthony George Clementz

Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

Loyola University Chicago Loyola University Chicago

Loyola eCommons Loyola eCommons

Dissertations Theses and Dissertations

2010

Polyomavirus Enhancer Activator 3 (PEA3), a Member of the Ets Polyomavirus Enhancer Activator 3 (PEA3), a Member of the Ets

Family of Transcription Factors, Is a Transcriptional Activator of Family of Transcription Factors, Is a Transcriptional Activator of

Notch-1 and Notch-4 in Breast Cancer: An Opportunity for Novel Notch-1 and Notch-4 in Breast Cancer: An Opportunity for Novel

Combinational Therapy Combinational Therapy

Anthony George Clementz Loyola University Chicago

Follow this and additional works at: https://ecommons.luc.edu/luc_diss

Part of the Biochemistry Commons

Recommended Citation Recommended Citation Clementz, Anthony George, "Polyomavirus Enhancer Activator 3 (PEA3), a Member of the Ets Family of Transcription Factors, Is a Transcriptional Activator of Notch-1 and Notch-4 in Breast Cancer: An Opportunity for Novel Combinational Therapy" (2010). Dissertations. 81. https://ecommons.luc.edu/luc_diss/81

This Dissertation is brought to you for free and open access by the Theses and Dissertations at Loyola eCommons. It has been accepted for inclusion in Dissertations by an authorized administrator of Loyola eCommons. For more information, please contact [email protected].

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 3.0 License. Copyright © 2010 Anthony George Clementz

Page 2: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

LOYOLA UNIVERSITY CHICAGO

POLYOMAVIRUS ENHANCER ACTIVATOR 3 (PEA3),

A MEMBER OF THE ETS FAMILY OF TRANSCRIPTION FACTORS,

IS A TRANSCRIPTIONAL ACTIVATOR OF NOTCH-1 AND NOTCH-4

IN BREAST CANCER: AN OPPORTUNITY

FOR NOVEL COMBINATIONAL THERAPY

A DISSERTATION SUBMITTED TO

THE FACULTY OF THE GRADUATE SCHOOL

IN CANDIDACY FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY

PROGRAM IN MOLECULAR AND CELLULAR BIOCHEMISTRY

BY

ANTHONY G. CLEMENTZ

CHICAGO, ILLINOIS

DECEMBER 2010

Page 3: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

Copyright by Anthony G. Clementz, 2010 All rights reserved.

Page 4: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

ACKNOWLEDGEMENTS

I am grateful to Dr. Clodia Osipo for her continual support, guidance, and

excellent mentorship. She has taught me how to be a strong scientist and person. When I

entered the laboratory, one of the first things she said to me was, “I am going to teach you

to be a good scientist, and above all else, as a young researcher, I will teach you

patience.” Not only have I learned patience, but the importance of integrity,

communication, and dedication. I must say that Dr. Osipo has gone above and beyond in

her mentorship and is herself an amazing scientist. I am very honored to be her first Ph.D.

graduate student.

I would like to thank my committee members, Dr. Mary Druse-Manteuffel, Dr.

Richard Schultz, Dr. Maurizio Bocchetta, and Dr. Paola Rizzo, for their support and

direction during my doctoral research studies. I would like to recognize Dr. Paola Rizzo,

a dear friend that has guided me not only in science, but also in many areas of my life; I

am ever grateful for our long talks and many laughs. I also would like to thank Dr.

Osipo’s lab personnel, former master’s student Parul Patel, Kinnari Pandya, Allison

Rogowski, and Kathleen Meeke, who are all excellent scientists, great friends, and made

my experience within the lab fun and unforgettable.

I would like to thank the Department of Cell Biology, Neurobiology, and

Anatomy: Division of Molecular and Cellular Biochemistry for educating and assisting

iii

Page 5: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

me throughout their program. I would also like to thank Loyola University, the many

faculty, and friends both at Loyola and outside Loyola. I have met many friends and

colleagues that have made my experience here memorable.

Most importantly, I must recognize and thank my dearest parents, Peter and Toni

Clementz, and family, Gina, Peter, Mia, and Andria, who stuck with me through it all,

listened to my stories, and continues to give endless support and love. I have been

blessed to have learned from the greatest people and to them I am truly grateful. I

attribute my gifts to four amazing people in my life and I have carried with me a part of

each of them: among so many other things, my father has taught me determination,

caring, and problem solving; my mother, respect, openness, and consideration. To my

incomparable grandfather, who taught me strength, passion, art, and always saw my

potential before I could even see it in myself; and my grandmother, for her wisdom,

prayer, and hope.

As I evolve into a young scientist, I find that the more I study concerning the vast

molecular infrastructure of the human body and life in general, I find myself humbled by

the intelligent design innate in us. It is within all of us –the beauty and blessings of life.

iv

Page 6: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

To my Father and Mother, And for Dorothy and Sebastian

Page 7: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

Considering the wonderful frame of the human body, this infinitely complicated engine, in which, to the due performance of the several functions and offices of life, so many strings and springs, so many receptacles and channels are necessary, and all to be in their right frame and order; and in which, besides the infinite, imperceptible and secret ways of mortality, there are so many sluices and flood-gates to let death in, and life out, it is next to a miracle we survived the day we were born.

J. Puckle, 1798

I awoke to the reality that I did not choose science; it chose me.

A.G.Clementz, 2010

Page 8: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

vii

PREFACE

Breast cancer continues to be the second leading cause of cancer-related deaths

among women. It is a disease that transcends gender boundaries, arising in both women

and men. Currently, according to the statistics acquired by the National Institute of

Health, there are approximately 200,000 female and 2,000 male new breast cancer cases

in the United States of America. Roughly 41,000 females and 450 males will parish this

year from the disease. However, with the advancement of current research, these

numbers are substantially lower than the past, and the percentage of patients reaching

their five year disease free survival is increasing. The advent of new surgical and

oncological techniques enables patients to receive a lumpectomy, radiation treatment,

targeted chemotherapeutics, and reconstruction or breast-conserving surgery, which not

only leads to increased survival rate, but also aims to regain physically and

psychologically the patient’s former quality of life. Nevertheless, breast cancer is an

aggressive and devastating illness. With continued support from global governments,

commercial industries, and personal conquests, the prospect for a cure is on the horizon.

Eradication of breast cancer is no longer a hopeful wish, but an emerging and tangible

promise.

Page 9: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

viii

TABLE OF CONTENTS

ACKNOWLEDGEMENTS………………………………………………………………iii

PREFACE………………………………………………………………………………..vii

LIST OF TABLES…………………………………………………………..……..……xi

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

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

ABSTRACT…………………………………………………………………………….xix

CHAPTER I: AN INTRODUCTION TO BREAST CANCER.…………………….1 Introduction……………………………………………………......………………1 Categorization and Features of Breast Cancers..………………….………….….3

1. Luminal A Breast Cancers…………………………………………….4 2. Luminal B Breast Cancers…………………………………………….5 3. HER2+ Breast Cancers………………………………...……………...6 4. Basal-like, Triple Negative Breast Cancers……………………….…7 5. Normal-like Breast Cancers……………………………………..…..9 6. Claudin-low Breast Cancers………………………………………....10 7. Molecular Apocrine Breast Cancers…………………………………10

Risk Factors for Breast Cancer………………………………………………….11 Treatment Strategies for Breast Cancer…………………………..……………..11

1. Surgery…………………………………………………………….…12 2. Radiation Therapy……………………………………………………13 3. Systemic Therapy……………………………………………….……13

A. Hormone Therapy……………………………………………14 B. Targeted Biological Therapy…………………………...…....16 C. Cytotoxic Chemotherapy……………………………………..17

Conclusions: Breast Cancer Now and the Future……………………………….18

CHAPTER II: NOTCH, PEA3, AND AP-1: A LITERATURE REVIEW……………...21 Notch Signaling……………………………………………………………..21

1. An Overview of Notch Signaling………………………………...….22 2. Notch Receptors…………………………………………………...…25

A. Structural Analysis of the Notch Receptors……….…….25 B. Notch Receptors and Breast Cancer……………………..….27

3. Notch Ligands……………………………………………….…….29 4. Inhibitors of Notch Signaling……………………………..……...29 5. Regulation of Notch Signaling…………………………….…….32

Page 10: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

ix

A. Post-transcriptional Regulation of Notch...........................32 B. Transcriptional Regulation of Notch…………….……...33

Polyomavirus Enhancer Activator 3 (PEA3)…………………………………….34 1. Ets Transcription Factors…………………………………………….34 2. Polyomavirus Enhancer Activator 3 (PEA3)………………………...35

A. The Structure of PEA3……………………………………….35 B. Activity and Regulation of PEA3…………………………….36

3. PEA3 and Breast Cancer……………………………………………..36 Activating Protein 1 (AP-1)……………………………………….……...40

1. The c-JUN Family………………………………………………42 A. c-JUN……………………………………………..……...42 B. JunB……………………………………………………….45 C. JunD……………………………………………………….45

2. The c-FOS Family………………………………………………46 A. c-FOS……………………………………………………...46 B. Fra-1………………………………………………………49 C. Fra-2………………………………………………………50 D. FosB……………………………………………………….50

3. AP-1 and PEA3…………………………………………………51 A Systems Approach: Linking Notch, PEA3, & AP-1……………………….…52

CHAPTER III: MATERIALS AND METHODS……..………………………...53

Cell Culture……………………………………………………………….…53 Drugs and Chemicals……………………………………………………….53 Expression Vectors………………………………………………………….54 RNA Interference and Reagents…………………………………………….54 Antibodies………………………………………………………………..…..54 Real-Time RT-PCR……………………………………………………….…56 Western Blot Analysis…………………………………………………….…57 Luciferase Constructs and Assay…………………………………………...59 Chromatin Immunoprecipitation (ChIP)…………………………………...60 Co-Immunoprecipitation (Co-IP)…………………………………………..62 Cell Cycle Analysis………………………………………………………..64 Annexin-V Apoptosis Assay……………………………………………....65 Cell Viability Assay…………………………………………………………66 Colony Formation Assay…………………………………………………..67 Statistical Analysis………………………………………………………….68

CHAPTER IV: RESULTS……………..………………………….……………..69

Hypothesis…………………………………………………….…………….69 Specific AIMS………………………………………………………………70 Specific AIM-1: To Determine Whether PEA3 is an Upstream

Mediator of the Notch Signaling Pathway………………..…..…72 1. PEA3 Regulates Notch-1 and Notch-4 Transcripts…………...75 2. PEA3 Regulates Notch-1Protein Expression…………..........75

Page 11: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

x

3. Classic Gene Targets of PEA3 and Notch are Decreased by PEA3 Knockdown………..…………………………………...79

Specific AIM-2: To Elucidate Whether PEA3 Acts as a Direct Positive Regulator of Notch-1 and Notch-4 Transcription………81 1. PEA3 is Enriched on the Notch-1 and Notch-4 Promoters…………..83 2. PEA3 and AP-1 Regulate the Notch-4 Promoter…………………....89 3. PEA3 and AP-1 Dynamically Regulate Notch-4 Transcription……101

Specific AIM-3: To Establish a Biological Significance by Determining Whether Dual Inhibition of Notch and PEA3 Leads to Reduced Cell Proliferation, Tumorigenicity, Migration, and Invasion in vitro………………………………………………107 1. Dual inhibition of Notch and PEA3 Inhibits Cell Proliferation……109 2. Dual Inhibition of Notch and PEA3 Induces Apoptosis……………109 3. Dual Inhibition of Notch and PEA3 Reduces Cell Viability……….111 4. Notch-1 and Notch-4 Overexpression Rescues the Effect of

Dual Notch and PEA3 Inhibition…………….…………….…..111 5. PEA3 Regulates Notch-1 and Notch-4 Expression in other

Subtypes of Breast Cancer………………………………….….117 CHAPTER V: DISCUSSION……………………………………………….….124

The Analysis………………………………………………………………125 The Model………………………………………………………………….135

1. Production…………………………………………………....135 2. Activation…………………………………………………....137

Future Investigations………………………………………………….....138

CHAPTER VI: CLOSING REMARKS…………………..………………….…140

BIBLIOGRAPHY……………………………………………………………...…142

VITA………………………………………………………………………………163

Page 12: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

xi

LIST OF TABLES

TABLE PAGE

1. Properties of the Classic Subtypes of Breast Cancer…………………………….20

2. Relative Levels of AP-1 Family Members in Breast Cancer…………………….41

3. Sequences of siRNA(s)…………………………………………………………..55

4. Sequences of PCR Primers………………………………………………………55

5. Chromatin Immunoprecipitation Primers………………………………………..63

Page 13: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

LIST OF FIGURES

FIGURE PAGE

1. Anatomy of the Breast…………………………………………………………….2

2. Chemical Structures of Common Estrogenic and Aromatase Inhibitors………...15

3. The Notch Signaling Pathway…………………………………………………...24

4. The Four Mammalian Notch Receptors, Their Ligands, and Their Domains…...26

5. Chemical Structures of Common GSI(s)…………………………………….…..31

6. Protein Structure of Polyomavirus Enhancer Activator 3 (PEA3)………………37

7. AP-1 Members and Dynamic Partnerships………………………………………41

8. Protein Structure of c-JUN and its Dominant Negative Form, TAM-67………...43

9. Protein Structure of c-FOS and its Dominant Negative Form, A-FOS………….47

10. Schematic Representation of Notch-1 and Notch-4 Promoters Labeled to Designate Areas of Primer Design……………………………………….63

11. Hypothesis of the Investigation……………………………………………….…71

12. PEA3 Transcripts Upon RNA Interference……………………………………...73

13. Endogenous PEA3 Protein Upon RNA Interference…………………………….74

14. Effects of PEA3 siRNA on Notch Transcripts…………………………………..76

15. Protein Levels of Notch-1 Upon PEA3 RNA Interference………………………77

16. Protein Levels of Notch-4 Upon PEA3 RNA Interference………………………78

xii

Page 14: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

17. Transcript Levels of Classic PEA3 and Notch Targets Upon PEA3 siRNA…….80

18. Schematic Representation of Notch-1 and Notch-4 Promoters………………….82

19. Exogenous Expression of PEA3 and Its Effects on Target Genes……………….84

20. Immunoprecipitation of Endogenous and Exogenous PEA3…………………….85

21. Chromatin Immunoprecipitation of Endogenous and Exogenous PEA3 on the Notch-1 and Notch-4 Promoter…………………………………...86

22. Chromatin Immunoprecipitation of PEA3 on the Notch-1 Promoter……………87

23. Chromatin Immunoprecipitation of PEA3 on the Notch-4 Promoter……………88

24. Western Blot Analysis of PEA3 Expression……………………………………..90

25. PEA3 Enrichment Specificity on the Notch-1 and Notch-4 Promoters as Measured by Chromatin Immunoprecipitation…………………….…….91

26. Wildtype and Mutant Notch-4 Promoter Luciferase Constructs……………...92

27. Luciferase Assay of Wildtype and AP-1 Mutant Notch-4 Promoter………….94

28. PEA3 Does Not Regulate AP-1 Activity………………………………………...95

29. Dominant Negative c-JUN Ablates PEA3 Recruitment on Notch-4 and Has No Effect on PEA3 Recruitment on Notch-1……………………….96

30. Dominant Negative c-JUN (TAM-67) Reduces AP-1 Activity………………….98

31. c-JUN is Required for PEA3 Enrichment on the Notch-4 Promoter…………...99

32. Co-Immunprecipitation of PEA3 in Complex with c-JUN……...……………...100

33. Dominant Negative c-FOS (A-FOS) Has No Effect on PEA3 Recruitment on the Notch-1 and Notch-4 Promoters………………………………...102

34. Dominant Negative c-FOS (A-FOS) Reduces AP-1 Activity………………….103

35. PEA3 and AP-1 Dynamically Regulate Notch-4 Transcription……………….105

36. Notch-1 Transcripts Are Not Effected by AP-1 siRNA Interference………….106

xiii

Page 15: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

37. RNA Interference Knockdown of PEA3 and AP-1 Members…………………108

38. Effects of Dual Notch and PEA3 on Cell Proliferation………………………..110

39. Dual Inhibition of Notch and PEA3 Increases Apoptosis……………………...112

40. Dual Inhibition of Notch and PEA3 Decreases Cell Viability…………………113

41. Notch-1 and Notch-4 Overexpression Reduces the Effect of Dual Notch and PEA3 Inhibition …………………………………………….115

42. Quantification of Notch-1 and Notch-4 Rescue on Colony Formation....……...116

43. Relative PEA3 Levels and Knockdown Efficiency in Breast Cancer Subtypes………………………………………………...120

44. PEA3 Regulates Notch in Other Breast Cancer Subtypes ……………………..121

45. Relative AP-1 Activity in Different Breast Cancer Subtypes…………………..122

46. PEA3 Regulates Notch-4 in BT474 and SKBr3 Breast Cancer Subtypes……...123

47. Model of the Transcriptional Regulation of Notch-1 and Notch-4 by PEA3…..136

xiv

Page 16: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

xv

LIST OF ABBREVIATIONS

ADAM A disintegrin and metalloprotease

A-FOS Dominant negative c-FOS

AI Aromatase inhibitor

ANK Ankyrin repeats

AP-1 Activator protein 1

APBI Accelerated partial breast irradiation

bZIP Basic leucine zipper domain

ChIP Chromatin immunoprecipitation

CoA Co-activator

Co-IP Co-immunoprecipitation

CoR Co-repressor

COX2 Cyclooxygenase 2

CR Cysteine rich region

Ct Cycle number at threshold

DBD DNA binding domain

DCIS Breast ductal carcinoma in situ

DMEM Dulbecco’s Modified Eagle Medium

DMEM/F12 Dulbecco’s Modified Eagle Medium: Nutrient Mixture F12

Page 17: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

xvi

DMSO Dimethyl sulfoxide

EGF-like Epidermal growth factor like repeats

EGFR Epidermal growth factor receptor

ER Estrogen receptor alpha

FBS Fetal bovine serum

FL Notch full length

GSI Gamma-secretase inhibitor

HC Heavy chain IgG isotype control

HER2 Epidermal growth factor receptor 2

HPRT Hypoxanthine-guanine phosphoribosyl transferase

IC Notch intracellular domain

IL-8 Interleukin 8

IMEM Iscove’s Minimal Essential Media

IP Immunoprecipitation

LC Long chain IgG isotype control

LNR Lin12 repeats

MAML-1 Mastermind-like 1

MAPK Mitogen activating pathway kinase

MMP Matrix metalloprotease

MMTV Mouse mammary tumor virus

Mut Mutation

NCR Cytokine response element

Page 18: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

xvii

NEAA Non-essential amino acids

NEC Notch extracellular domain

NIC Notch intracellular domain

NLS Nuclear localization signal

NTM Notch transmembrane domain

O-fut O-fucosyl transferase

PARP Poly-ADP-ribose polymerase

PBS Phosphate buffer saline

PCR Polymerase chain reaction

PEA3 Polyomavirus Enhancer Activator 3

PEA3i PEA3 siRNA

PEST Proline glutamic acid serine threonine

PI Propidium iodide

PR Progestrone receptor

PVDF Polyvinylidene fluoride

RFU Relative fluorescence units

RIPA Radioimmunoprecipitation assay buffer

RT-PCR Realtime polymerase chain reaction

SCRBi Scrambled control random siRNA

SDS Sodium dodecyl sulfate

SDS-PAGE Sodium dodecyl sulfate polyacrylamide gel electrophoresis

SERM Selective estrogen receptor modulator

Page 19: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

xviii

siRNA Small interfering RNA

SUMO Sumoylation

TACE TNF-alpha converting enzyme

TAD Transactivation domain

TAM-67 Dominant negative c-JUN

TBST Tris buffer saline tween

TM Notch transmembrane portion

uPA Urokinase-type plasminogen activator

VEGF Vascular endothelial growth factor

VEGFR Vascular endothelial growth factor receptor

Wt Wildtype

Page 20: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

xix

ABSTRACT

Women diagnosed with triple-negative breast cancer have the worst overall

prognosis and frequently present with metastatic tumors. To date, there are no targeted

therapies available to combat this aggressive form of breast cancer due to the lack of

expression of well-known targets such as ERα, PR, or HER2/neu. Therefore, there is an

immediate need to identify novel targets that are responsible for the proliferation,

survival, and invasive phenotype. Notch-1 and Notch-4, both potent breast oncogenes,

are overexpressed in triple-negative breast cancers-associated with the poorest overall

survival. PEA3 (polyomavirus enhancer activator 3), a member of the Ets family of

transcription factors, is overexpressed in triple-negative breast cancer and also correlates

with aggressive behavior and poor overall survival. Here, we provide new evidence for

transcriptional regulation of Notch in triple negative breast cancer and other subtypes of

breast cancer. Our results showed that PEA3 is a transcriptional activator of both Notch-

1 and Notch-4 genes using a PEA3 siRNA and measuring both transcripts and proteins in

MDA-MB-231 cells where PEA3 levels are endogenously high. In SKBr3 and BT474

breast cancer cells where PEA3 levels are low, exogenous overexpression of PEA3

significantly increased Notch-4 transcripts. Chromatin immunoprecipitation confirmed

enrichment of PEA3 within the promoter regions of both Notch-1 and Notch-4. Notch-1

Page 21: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

xx

recruitment appears to be AP-1 independent, whereas PEA3 recruitment on the Notch-4

promoter is dependent upon c-JUN. Furthermore, results showed that either c-Jun or Fra-

1 are required for Notch-4 transcription while c-FOS was a repressor. Importantly, the

combined inhibition of Notch signaling via a gamma-secretase inhibitor (GSI) and

knockdown of PEA3 (via siRNA) significantly growth arrested breast cancer cells in G1

and decreased both anchorage dependent and independent growth in vitro. In correlation,

a significant decrease in cell viability and tumorigenicity, as well as an increase in

apoptotic cells were observed when a GSI was combined with PEA3 siRNA.

Interestingly, a combined inhibition of Notch signaling and PEA3 showed no significant

changes in invasion and migration. Taken together, results from this study suggest for

the first time that Notch-1 and Notch-4 are novel transcriptional targets of PEA3 in breast

cancer cells. PEA3-mediated Notch-1 transcription is AP-1 independent while Notch-4

transcription requires both PEA3 and AP-1 most probably composed of the c-Jun:Fra-1

complex. Moreover, both PEA3 and Notch signaling are essential for proliferation and

survival of MDA-MB-231 breast cancer cells. Thus, dual targeting of both PEA3 and

Notch pathways might provide a new therapeutic strategy for triple-negative breast

cancer as well as additional therapeutic targeting in other breast cancer subtypes.

Page 22: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

CHAPTER I

AN INTRODUCTION TO BREAST CANCER

Introduction

Breast cancers are thought to originate from a small number of cancer cells that

are capable of self renewal and pluripotency. These breast cancer “stem cells” are capable

of initiating and sustaining breast tumor growth. The cancer-initiating cells or cancer

stem cells were originally identified in hematological malignancies but is now being

recognized in breast and several other solid tumors (Subramaniam et al., 2010). The

normal breast anatomy contains lobular glands that produce milk, the luminal ducts that

carry the milk to the nipple, and the surrounding environment consisting of adipose,

connective, and lymphatic tissue (Figure 1). Transformed stem cells lose their internal

proliferation checkpoints and develop into a benign palpable mass or neoplastic tumor.

When tumor cells further transform into migratory cells or under epithelial-to-

mesenchymal transition (EMT), they become invasive or infiltrating breast cancers

distinguishing them from benign or breast cancer in situ. The breast undergoes multiple

remodeling events similar to EMT throughout a woman’s lifetime and most notably

during pregnancy. Multiple steps during development and pregnancy could be targets of

mutation and subsequent transformation where cancer may develop. Consequently,

1

Page 23: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

2

DuctsAreolaNipple

Connective tissueAdipose tissue

● ● ● ●●● ●●●●●

●●●●●

● ● ● ●●●●

●● ● ● ● ● ● ●

●●

●●

●●●●

●●●

●●●

Duct Cells

Duct

LobuleLobularCells

Tumor

AdipocyteMacrophage

Stroma

Lumen

BasementMembrane

MyoepithelialCell

Luminal Cell

DuctsAreolaNipple

Connective tissueAdipose tissue

● ● ● ●●● ●●●●●

●●●●●

● ● ● ●●●●

●● ● ● ● ● ● ●

●●

●●

●●●●

●●●

●●●

● ● ● ●●● ●●●●●

●●●●●

● ● ● ●●●●

●● ● ● ● ● ● ●

●●

●●

●●●●

●●●

●●●

●● ●● ●● ●●●●●● ●●●●●●●●●●

●●●●●●●●●●

●●

●● ●● ●● ●●●●●●●●

●●

●●●● ●● ●● ●● ●● ●● ●●

●●●●

●●●●

●●●●●●●●

●●●●●●

●●●●●●

Duct Cells

Duct

LobuleLobularCells

Tumor

AdipocyteMacrophage

Stroma

Lumen

BasementMembrane

MyoepithelialCell

Luminal Cell

AdipocyteMacrophage

Stroma

Lumen

BasementMembrane

MyoepithelialCell

Luminal Cell

Figure 1: Anatomy of the Breast.

Page 24: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

3

breast cancer assumes diverse intrinsic gene patterns leading to the formation,

identification, and classification of particular molecular subtypes (Micalizzi et al., 2010)

Categorization and Features of Breast Cancers

Breast cancer is a heterogeneous disease. Molecular and pathological portraits of breast

cancer provide insight into the vast genetic and phenotypic varieties that may present in

the clinic. A hierarchical division among breast cancer using a both pathological and

genetic analysis of expression have been formulated (Perou et al., 2000). They

subdivided breast cancer into four minimal subtypes: luminal, HER2, normal breast-like,

and basal-like. Soon after, further investigations divided the luminal subtype into luminal

A and luminal B based on gene profiling and clinical outcomes (Sorlie et al., 2001). The

classification of tumors stemming from the lumen or the myoepithelial (basal) layer was

determined by immunohistochemistry on sections of patient samples using antibodies to

keratin 5,8,17, and 18 (Perou et al., 2000). Those tumors that showed positive keratin 8

and 18 staining were considered of luminal origin and those that were positive for keratin

5 and 17 were derived from the basal-epithelial layer. Additionally, whether or not there

was presence of gene expression patterns of the estrogen receptor and its corresponding

hormone responsive genes, aided in the categorization of luminal versus basal-like

cancers, respectively. Further classifications are continuing to emerge such as the

molecular apocrine and claudin-low breast cancer subtypes. Although segregated into

different categories, sometimes the classification of breast cancer subtypes share common

gene expression and tend to overlap. However, four classic subtypes have

Page 25: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

4

withstood the test of time, and are commonly referenced: luminal A, luminal B, HER2,

and basal-like (including triple negative) (Table 1) (Kao et al., 2009).

1. Luminal A Breast Cancers

The luminal A subtype makes up approximately 60% of breast cancers and

originates from the cells that nest within the lumen of the breast that have undergone

oncogenic transformation. They are classically hormone receptor positive, that is

estrogen receptor alpha positive (ERα+) and progesterone receptor positive (PR+) (Sorlie

et al., 2001). They are negative for the overexpression or gene amplification of HER2, a

member of the EGFR family, and tend to express low levels of genes responsible for

proliferation. They lack expression of basal markers such as cytokeratin 5/6, but usually

express GATA3 (Chou et al., 2010; Usary et al., 2004), a 13% rate of TP53 mutations

(Langerod et al., 2007; Sorlie et al., 2001), and have a distinct characteristic gene

expression profile (Perou et al., 2000). Luminal A tumors usually present with a low

histological grade and have overall good prognostics when detected early. Patients with

luminal A breast cancer are six-times less likely to die from the disease when compared

to the aggressive basal-like subtype (Langerod et al., 2007). This overall good survival

may be due in part to the availability of targeted therapy directed at ERα. Common

hormonal treatments given to these patients to treat early and advanced breast cancer

include tamoxifen, a selective estrogen receptor modulator (SERM), often given to pre-

menopausal women, and/or letrozole, an aromatase inhibitor (AI), often a better choice

for post-menopausal women (Swaby and Jordan, 2008). Luminal A breast cancer tends

Page 26: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

5

to have a 29% rate of relapse 15 years after initial diagnosis and have the longest survival

rates when compared to the other subtypes (Kennecke et al., 2010; Peppercorn et al.,

2008). However, intrinsic and acquired resistance to adjuvant anti-hormonal therapy is

still a major problem showing a 63% rate of recurrence (Peppercorn et al., 2008).

2. Luminal B Breast Cancers

Like luminal A, the luminal B subtype makes up 55% of breast cancers and

originates from luminal cells nested in the breast. However, the luminal B gene profile

differs from the luminal A, thus creating its own subcategory in breast cancer. They are

classically hormone receptor positive (ERα+ / PR+), however, a key aspect of luminal B

tumors is the presence of ErbB-2/HER2/neu (HER2+) (Sorlie et al., 2001). These tumors

are more aggressive in nature than luminal A tumors and often co-express EGFR and/or

cyclin E1 (Sorlie et al., 2003). They tend to have high expression of proliferation-

associated genes and high histological grade. They too lack expression of basal markers

such as cytokeratin 5/6, but express GATA3 (Usary et al., 2004), and have a 40%-70%

rate of TP53 mutation status (Langerod et al., 2007; Peppercorn et al., 2008). Much of

current research is devoted to attacking this aggressive form of luminal B breast cancer.

If presented in the clinic as a non-metastatic lesion, these patients have overall good

survival due to combinatory treatment strategies involving pharmacological drugs

targeted at the estrogen receptor (tamoxifen or AI), and the humanized monoclonal

antibody directed against HER2 (trastuzumab) or a small molecule tyrosine kinase

inhibitor against the EGFR/HER2 receptor (lapatinib) (Amar et al., 2008; Bedard et al.,

Page 27: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

6

2009; Dean-Colomb and Esteva, 2008). Compared to luminal A, HER2+, or basal-like

subtypes, 50% of patients with luminal B breast cancer have a five year disease-free

survival (Sorlie et al., 2001). However, intrinsic or acquired resistance to anti-hormonal

or anti-HER2 therapy remains problematic as evidenced by a 81% rate of recurrence

(Peppercorn et al., 2008).

3. HER2+ Breast Cancers

Approximately 15-25% of breast cancers have acquired an overexpression or gene

amplification for HER2 on chromosome 17q12 (Rouzier et al., 2005), whose transcripts

can be elevated by two-fold to greater than twenty-fold (Slamon et al., 1989).

Interestingly, the HER2+ subtype is hormone receptor negative showing no positive

expression of the estrogen or progesterone receptors, and low levels of their hormone

responsive genes (Sorlie et al., 2001). HER2+ breast cancers tend to have aggressive

clinical behavior acquiring 4.2 fold increase in expression of proliferation-associated

genes when compared to luminal A tumors (Peppercorn et al., 2008). They also have

high histological grade and harbor a 71% rate of TP53 mutations (Peppercorn et al.,

2008; Weigelt et al., 2009). Although they exhibit luminal features, they are associated

with only a 30% rate of disease-free survival at 5 years (Cheang et al., 2008; Sorlie et al.,

2001). HER2+ tumors are associated with early relapse, high risk of recurrence, and

more likely to involve metastasis to the axillary lymph nodes before diagnosis (Sorlie et

al., 2001). However, current anti-HER2 targeted therapy, such as the monoclonal

antibody, trastuzumab, or the EGFR/HER2 tyrosine kinase inhibitor, lapatinib, has

Page 28: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

7

reduced the risk of relapse and dramatically improved survival outcomes (Romond et al.,

2005). In the adjuvant setting, the overall response rate of 26% to lapatinib treatment

was increased to 90% when lapatinib was combined with chemotherapeutic agents in

HER2+ breast cancers (Amar et al., 2008; Bedard et al., 2009; Dean-Colomb and Esteva,

2008). Although responsive to chemotherapy and regimented targeted therapy, 10-15%

of women acquire resistance, particularly those that present with metastatic tumors (Amar

et al., 2008; Romond et al., 2005).

4. Basal-like, Triple Negative Breast Cancers

The basal-like subtype makes up approximately 15-20% of breast cancer cases.

They are defined as those tumors which lack ER, PR, and HER2 expression (Sorlie et al.,

2001). They are prevalent among pre-menopausal, African American women: 20.8% in

African American women versus 10.4% other races including those harboring a BRCA1

mutation (Morris et al., 2007; Peppercorn et al., 2008). Basal-like tumors are very

aggressive in nature, have high rates of metastasis (Nielsen et al., 2004), and lead to poor

overall survival (van de Rijn et al., 2002). They are considered basal-like because the

tumor expresses genes usually associated with the normal breast basal/myoepithelial cells

(Jones et al., 2004), such as cytokeratin 5 and 17 (Perou et al., 2000). These tumors are

highly proliferative, exhibit elevated expression of proliferation-associated genes, have a

high histological grade, up to 85% tend to harbor TP53 mutations (Sorlie et al., 2001;

Weigelt et al., 2009), and approximately 60% overexpress EGFR (Nielsen et al., 2004;

Reis-Filho et al., 2006). Pathology of basal-like tumors demonstrates high central

Page 29: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

8

necrotic zones, typical and atypical medullary features, and metaplastic areas (Fulford et

al., 2006; Livasy et al., 2006). It is important to note, that not all basal-like tumors are

triple negative. In fact, a small percentage (<5%) of basal-like tumors contain ER, PR, or

HER2 gene expression, implying that the idea of “triple-negative breast cancer” may not

be synonymous with basal-like (Reis-Filho and Tutt, 2008). Reis-Filho made it quite

clear the importance of this separation in the clinic:

Patients with triple negative cancers expressing a basal phenotype had a

significantly shorter disease-free survival than those with triple-negative

cancers lacking the expression of basal markers. Therefore, caution

should be exercised not to equate a triple negative phenotype with a basal-

like profile. Triple-negative cancer is not a synonym for basal-like cancer

(Reis-Filho and Tutt, 2008).

This is a clear testament of the diverse signaling pathways present within the cell, the

intrinsic differences between breast cancers, and the need to better comprehend and target

aberrant factors controlling this transformation.

The interest in basal-like, triple-negative breast cancer is in part due its highly

aggressive nature and the fact that currently there is no known targeted therapy.

Cytotoxic chemotherapeutics such as taxenes (62% initial response rate), epidermal

growth factor receptor (EGFR) inhibitors (49% initial response rate), vascular endothelial

growth factor (VEGF) inhibitors (63% initial response rate), and poly-ADP-ribose

polymerase (PARP) inhibitors (48% initial response rate) are currently under

investigation for the treatment of triple-negative breast cancers (Bartsch et al., 2010).

Page 30: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

9

Although the percent response rates appear high, unfortunately the risk of resistance and

recurrence is also high usually developing between the first and third years following

therapy (Dent et al., 2007). The majority or patients never make their five year survival

mark. The standard of care is a combination of cytotoxic drugs: cyclophosphamide,

doxorubicin, and fluorouracil (Hortobagyi, 1998) or recently shown to be more effective

in triple negative breast cancer was the combination of cyclophosphamide, methotrexate,

and fluorouracil (Colleoni et al., 2010). Their rate of survival is shorter than those who

present with luminal type tumors (Sorlie et al., 2001). Thus, there is an immediate need

for the elucidation of novel targets to treat these patients and increase their survival.

5. Normal-like Breast Cancers

Normal-like breast cancer is still in its infancy regarding lucid categorization. It

is known to be ER positive or negative, PR status undetermined (most likely negative),

and HER2 negative. It does show signs of basal markers, leading some to believe it

stems from the branch of the basal-like subtype. The grade, presence of TP53 mutations,

and relative levels of genes responsible for proliferation are low. Interestingly, Normal-

like breast tumors have been shown to consistently cluster with fibroadenoma and normal

breast samples. These Normal-like breast tumors contain genes normally associated with

adipose tissue (Peppercorn et al., 2008).

Page 31: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

10

6. Claudin-low Breast Cancers

Claudin-low breast cancer acquires its name because of the reduced expression of

genes involved in tight junctions and cell-cell adhesion. It could be considered a branch

of the triple negative subtype in that it lacks expression of ER, PR, or HER2+. The

histological grade and relative levels of genes responsible for proliferation are high. The

TP53 status is unknown. However, what differentiates this type from the triple negative

is that the transcriptomic features of the tumors are suggestive of ‘cancer stem cell-like’

(Hennessy et al., 2009). Investigations into this subtype may prove to be advantageous

and interesting to the field of cancer stem cell research.

7. Molecular Apocrine Breast Cancers

Molecular apocrine breast cancer has recently surfaced as a category, which is

believed to be close to the HER2 positive, ER negative, and PR negative subtype. What

distinguishes it from the HER2+ subtype and all other ER-negative tumors is the

presence of an active androgen receptor (AR) and its subsequent target genes (Farmer et

al., 2005). However, the relative levels of genes responsible for proliferation are high.

With these gene signatures, apocrine tumors represent 8–14% of breast tumors. The

knowledge of elevated AR may provide a novel strategy for targeted therapy in this type

of breast cancer.

Page 32: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

11

Risk Factors for Breast Cancer

Age, race, family history, breast density, early menarche, age of first pregnancy,

and late menopause can all contribute to the risk of breast cancer (Peppercorn et al.,

2008). These are factors over which there is limited personal control. However, certain

hormonal therapies such as hormone replacement therapy, obesity, alcohol consumption,

inactivity, diet, smoking, and radiation exposure are other risk factors that can be

monitored and/or avoided. Similarly, increase instances in ER positive over ER negative

breast cancers have been associated with an increase in alcohol consumption (Suzuki et

al., 2005). Long-term aspirin use has been associated with decreased development of ER

positive tumors, but increased ER negative tumors (Marshall et al., 2005). Poor diets,

even ones primarily rich in vegetables, and inactivity has been associated with the

development of ER negative breast cancers (Fung et al., 2006). Along with a healthy

balanced diet, folate (Vitamin B9) has been linked to a decreased risk of ER negative

breast cancer (Zhang et al., 2005). Even though correlations have been observed, it is

important to understand that there are multiple factors that are required to develop breast

cancer and that none of these alone can be solely attributed to breast cancer development.

Nevertheless, there is no doubt that overall psychological and physiological well being

helps to provide a better defense against breast cancer development.

Treatment Strategies for Breast Cancer

Breast cancer is a diverse and aggressive disease. When presented at time of

diagnosis, each patient is unique and their breast cancer varies according to their grade,

Page 33: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

12

type, and response to treatment. With such individualization, the ultimate decision of

proper treatment lies with the patient, their doctors, and their families. Many times,

multiple treatment strategies are administered to help eradicate the disease. Currently,

the most common strategies to treat breast cancer are surgery, radiation therapy, systemic

therapy, and a low-fat/low-alcohol diet.

1. Surgery

When a patient presents in the clinic, a biopsy is taken, and surgery usually

follows after diagnosis. Surgery can be minimal such as a lumpectomy, or as extensive

as radical mastectomy (Maughan et al., 2010). A lumpectomy is the first choice of

surgery for early stage breast cancers, where the surgeon will remove the tumor and a

layer of lining stroma. After surgery, there is usually a five to seven week follow-up with

radiation therapy. A second surgical choice is simple or total mastectomy, where there is

removal of the entire breast. Later stage breast cancers may have infiltrated to the

sentinel and/or axillary lymph nodes. A biopsy of the sentinel and then possibly the

axillary lymph nodes is taken to determine the extent of metastasis. A modified radical

mastectomy may be administered in which the total breast is removed as well as the

lymph nodes. An extremely aggressive breast cancer that has infiltrated to the lymph

nodes and chest wall requires a total radical mastectomy, where the breast, lymph nodes,

and chest wall muscle are all removed (Fisher et al., 2002a; Fisher et al., 2002b; Veronesi

et al., 2002). Normally, surgery is not a sole means of treatment. As the grade and stage

Page 34: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

13

of the disease increases, the more combinatory strategies are taken (e.g. surgery with

chemotherapy and radiation).

2. Radiation Therapy

Radiation therapy is either given before surgery to help reduce the tumor burden,

or after surgery to insure the annihilation of remaining cancer cells. There are two

methods of administering radiation therapy: external or internal. In external radiation, a

biophysical machine directs a beam of a radioactive isotope towards the affected breast

and/or tumor. The external radiation is administered across an average five to seven

weeks and is effective at eliminating and/or reducing tumorigenic cells. In internal

radiation, the radioactive isotope is dispensed directly into the tumor through thin

capillary wires or needle injection. This method is also termed brachytherapy and is

increasingly becoming more effective. Some patients undergo both forms of radiation

treatment, but the type and stage of the cancer may favor one method over the other.

Currently, accelerated partial breast irradiation (APBI) is a method being developed to

reduce doses, area, and duration of treatment (Dirbas, 2009). Although reduced tumor

burden has been observed, radiation is rarely used as a sole form of treatment. Its

effectiveness relies on both surgery and systemic therapy.

3. Systemic Therapy

Systemic therapy is a means to treat breast cancer through oral administration or

injection, which consequently circulates through the entire body. Treatment may be

Page 35: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

14

given before surgery (neoadjuvant), after surgery (adjuvant), or when surgery is not an

option. Currently, hormone therapy, targeted biological therapy, and cytotoxic

chemotherapy are the most common forms of systemic therapy.

A. Hormone Therapy

The estrogen receptor has emerged as a critical hormonal target to modulate in ER

positive breast cancers and has been shown to promote tumor growth. Tamoxifen, a

selective estrogen receptor modulator (SERM) and partial agonist, is a leading triphenyl-

ethylene compound and medicinally modified moiety that binds and competitively

inhibits the estrogen receptor, dampens the estrogenic signaling, and consequently

reduces the estrogenic responsive effects (Figure 2) (Jordan et al., 1981). Tamoxifen is

given orally to both pre- and post-menopausal women for a period of five years (Swaby

and Jordan, 2008). It has been demonstrated that following treatment, there is an

approximately 33% reduction in death rate and 41% reduction in recurrence (Coates et al.,

2007). Tamoxifen has undesirable thromboembolism events and increased endometrial

cancer risk as major side effects (Hind et al., 2007). Another anti-estrogen is Fulvestrant

(Faslodex), a complete antagonist and competitive inhibitor of the estrogen receptor

which is given as an intramuscular injection (Diagram 2) (Buzdar, 2008). It binds to the

estrogen receptor and destabilizes the three dimensional structure thus facilitating E3

ubiquitin ligase/proteasome-mediated receptor degradation (Berry et al., 2008; Howell,

2006). Consequently, estrogenic signaling and tumor growth is significantly reduced.

Patients taking Faslodex have reported gastrointestinal side effects.

Page 36: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

15

CH3

ON

CH3

CH3

S

OH

OH

CH3

O

F

FF

F

F

NN

N

NN

N

NN

CH3

CH3CH3

CH3

CH3CH3

CH3

CH3

CH2

O

O

Tamoxifen Fulvestrant

Letrozole Anastrozole Exemestane

Aromatase Inhibitors (AI)s

Estrogenic Inhibitors

Figure 2: Chemical Structures of Common Estrogenic and Aromatase Inhibitors

Page 37: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

16

Aromatase Inhibitors (AIs) are a class of drugs that inhibit the aromatase enzyme

that converts either androstenedione into 17β-estradiol, or directly testosterone into 17β-

estradiol. Current drugs include letrozole (Femara), anastrozole (Arimidex), and

exemestane (Aromasin) (Figure 2). They are effective in post-menopausal women where

small amounts of estrogen are produced in the adipose tissue surrounding the tumor.

Current research has shown that AIs alone or interspersed with tamoxifen treatments is

more advantageous than tamoxifen alone (Buzdar et al., 2006). AIs are relatively more

expensive and possess undesirable side-effects such as osteopenia and bone pain (Hind et

al., 2007).

B. Targeted Biological Therapy

Since approximately 30% of breast cancers contain a gene amplification or

overexpression of the HER2 receptor linked to tumor survival, HER2 has become a

critical target to inhibit. In 2006, trastuzumab (Herceptin) was approved by the FDA to

treat patients with HER2 positive breast cancer (O'Mahony and Bishop, 2006).

Trastuzumab is a humanized, monoclonal antibody that binds to the extracellular portion

of the HER2 receptor (Carter et al., 1992). HER2 dimerization is disrupted and its ability

to promote survival is dampened. Studies have shown that treatment with trastuzumab in

combination with cytotoxic chemotherapy such as a taxane have reduced patient death

rate by 33% and risk of recurrence by 52% (Romond et al., 2005). Lapatinib, a small

molecule tyrosine kinase inhibitor targeted against EGFR and HER2, is also used to

target the signaling cascade from the EGFR family of receptors (McArthur, 2009).

Page 38: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

17

Another current form of targeted biological therapy is bevaxizumab (Avastin), which is a

humanized monoclonal antibody against the vascular endothelial growth factor (VEGF)

(Sachdev and Jahanzeb, 2008). VEGF is responsible for angiogenesis, and since a tumor

needs an enriched blood supply, a VEGF inhibitor prevents tumor vascularization and

angiogenesis. Avastin was approved by the FDA in 2008 as an added chemotherapeutic

strategy for advanced breast cancer (Spalding, 2008). Other small molecules or

antibodies are being developed, tested, and currently are in clinical trials. Targeted

biological therapy shows much promise particularly when combined with surgery and

cytotoxic chemotherapy.

C. Cytotoxic Chemotherapy

Some breast cancers require a more general systemic treatment. Cytotoxic

chemotherapy is designed to target rapidly dividing cells, which is a hallmark of a highly

proliferative tumor. Since cytotoxic chemotherapy target all rapidly dividing cells,

several non-cancerous cells may unfortunately be targets as well. Rapidly dividing

benign cells are found in the hair shaft, intestines, and in the bone marrow, which are

why many patients experience side effects such as hair loss, gastrointestinal disturbances,

and anemia (Chan and Giaccia, 2008). Some examples of cytotoxic drugs include

taxanes (paclitaxel, Taxol), cyclophosphamide, fluorouracil, and cisplatin (Mouridsen,

1990; Rowinsky et al., 1992; Sledge, 1992). Taken together, systemic therapy when

combined with other targeted drugs, surgery, and radiation have been shown to be more

advantageous than with one form of treatment (Hortobagyi, 1998; Mauri et al., 2008). As

Page 39: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

18

medicines are being developed and studied, future research needs to understand and

elucidate novel aspects of breast cancer biology.

Conclusions: Breast Cancer Now and the Future

Breast cancer continues to be one of the major causes of cancer-related death in

women. Presently, there are four classic subtypes of breast cancer: luminal A, luminal B,

HER2/neu, and basal-like, but more novel categorizations are being developed as the

systems biology and gene arrays uncover new patterns of expression. Currently,

tamoxifen still remains a leading chemotherapeutic choice for hormone positive breast

cancers. In addition, the advent of novel aromatase inhibitors and more potent

medicinally modified estrogenic inhibitors are becoming an additional choice for

treatment. However, approximately half of the women treated will develop resistance to

the anti-estrogens, driving current research to develop targeted combinational strategies.

Today, trastuzumab (Herceptin) and lapatinib, are chemotherapeutic strategies to treat

patients with HER2+ breast cancer. Unfortunately, the triple negative basal-like subtype

still manifests high mortality, high recurrence, and no known targeted treatment to date.

Therefore, there is an immediate need to identify novel targets that are responsible for the

proliferation and invasive phenotype for these cancers so as to increase overall patient

survival. Taken together, the future of breast cancer lies within the very biochemical

infrastructure of the transformed cells, their stem-likeness, and their microenvironment,

which still remains relatively elusive. As each compromised cell responds to its extrinsic

signaling cascades and its downstream aberrant intrinsic transcriptional effects,

Page 40: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

19

progressive research will uncover and exploit the molecular mechanisms and novel

targets that drive its progression, aggression, and survival.

Page 41: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

SUBTYPE EXAMPLE CELL LINE ER PR HER2 TP53

(MUT) GRADE PROLIFERATIONGENE PROFILE

CLINICAL FEATURES

TREATMENT STRATEGIES

LUMINAL A

MCF-7

TYPE: Metastatic

Adenocarcinoma

+ + – LOW LOW LOW

Most common form of breast cancer

Lower risk of reoccurrence

Endocrine therapy responsive

Less responsive to chemotherapy

Tamoxifen SERM(s)

Letrozole

Exemestane AI(s)

LUMINAL B

BT474

TYPE: Invasive Ductal

Carcinoma

+ + + MODERATE MODERATE HIGH

Endocrine therapy responsive

Moderate response to chemotherapy

Tamoxifen SERM(s)

Letrozole

Exemestane AI(s)

Trastuzumab

Lapatinib

ERBB2/HER2+

SKBR3

TYPE: Metastatic

Adenocarcinoma

– – + HIGH HIGH HIGH

Likely involvement of axillary lymph nodes upon diagnosis

High risk of reoccurrence

Responsive to chemotherapy

Trastuzumab

Lapatinib

BASAL-LIKE

MDAMB231

TYPE: Adenocarcinoma

– – – HIGH HIGH HIGH

Association with BRCA-1 mutations

Common in pre-menopausal African American women

High risk of reoccurrence

Responsive to chemotherapy

No known targeted treatment

No targeted treatment

PARP inhibitors

VEGF inhibitors

Taxanes

Cytotoxic

chemotherapy Abbreviations: ER, estrogen receptor; PR, progesterone receptor; MUT, mutations. References: (Peppercorn et al. 2008, Kao 2009)

Table 1: Properties of the Classic Subtypes of Breast Cancer.

20

Page 42: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

CHAPTER II

NOTCH, PEA3, AND AP-1: A LITERATURE REVIEW

The molecular profile of breast cancer involves a series of interconnected

pathways that when perturbed, disrupts normal cellular homeostasis and drives the cell

towards transformation. The cell discovers new ways of utilizing signaling cascades and

transcriptional events to become hyper-sensitive through amplification or enhanced

activity. In addition, elimination of genes, often termed “tumor suppressors,” is exploited

so the cell can acquire resistance to apoptosis, immortality, self-sufficiency, and

migratory capabilities. Oncogenic pathways that increase proliferation and prevent

apoptosis have emerged as potential targets for the treatment of breast cancer. For

example, Notch signaling, PEA3 transcriptional effects, and AP-1 dynamic partnership

all have been associated with aggressive breast cancers, particularly the triple negative

subtype, and are pathways the cancer cell has manipulated to drive its survival and

progression.

Notch Signaling

In the early twentieth century, the discovery of “Notch” and its name came after

an investigation of genetic gene mutants in Drosophila. Upon exploration of the

mutations, 21

Page 43: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

22

one particular mutant resulted in a “notch” of missing tissue at the tip of the fly wing

blades (Morgan, 1917). This identification eventually led to the discovery of the Notch

pathway. As years passed, Notch signaling became very interesting to scientists both in

development –implicated in insulin-secreting pancreatic beta-cells (Apelqvist et al.,

1999), the inner ear hair cell development (Lanford et al., 1999), and intestinal crypt and

goblet cells (van Es et al., 2005) –and eventually cancer. Notch has emerged as a potent

oncogene and is aberrantly expressed in breast cancer (Stylianou et al., 2006). Elevated

levels of Notch-1 and one of its ligands, Jagged-1, confer poor prognosis and overall

diminished survival (Dickson et al., 2007; Reedijk et al., 2005). Currently, Notch is

becoming an advantageous pathway to explore and exploit in neoplastic cells as well as

potential cancer stem cells having been implicated in proliferation of tumor initiating or

mammary cancer stem cells (Harrison et al., 2010). Currently, Notch is becoming an

advantageous pathway to explore and exploit in neoplastic cells as well as potential

cancer stem cells having been implicated in proliferation of tumor initiating or mammary

cancer stem cells (Harrison et al., 2010).

1. An Overview of Notch Signaling

Notch signaling has emerged as a target for the treatment of breast cancer. In the

mammalian system, there are four Notch receptors (Notch-1,2,3,4) which are all type-I

transmembrane proteins (Blaumueller and Artavanis-Tsakonas, 1997) and five known

ligands (Delta-like 1,2,4, and Jagged 1,2) (Dunwoodie et al., 1997; Lindsell et al., 1995;

Shawber et al., 1996). Cell-to-cell contact activates Notch signaling, which subsequently

Page 44: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

23

enables the pathway to modulate genes involved in cell fate, proliferation and

differentiation (Callahan and Raafat, 2001). Notch is processed in the trans-Golgi

apparatus, where it undergoes the first of three proteolytic cleavages (Figure 3)

responsible for transcriptional activation. The first cleavage (S1) takes place in the trans-

Golgi by furin-like convertase converting the single polypeptide into the mature

heterodimeric Notch receptor held together by di-cations (Ca2+). The receptor is

trafficked to the cellular membrane where it awaits physical contact with its ligand

(Kopan and Ilagan, 2009). When ligand binds through EGF-like repeats and electrostatic

interactions, the extracellular portion of Notch is co-endocytosed with ligand into the

neighboring, ligand-expressing cell thus exposing a second cleavage site (S2) on the

transmembrane portion of the Notch (NTM) which is then cleaved by TNF-alpha

converting enzyme (TACE), a disintegrin and metalloproteinase (ADAM10/17) (Brou et

al., 2000). Consequently, a third cleavage site (S3) within the transmembrane domain of

Notch is now susceptible to proteolysis by the membrane associated aspartyl proteinase,

the γ-secretase complex (presenilin, nicastrin, APH1, PEN2) (Kopan and Ilagan, 2004),

liberating the intracellular portion of Notch (NIC) (Saxena et al., 2001). Notch (NIC)

translocates to the nucleus and binds to CBF-1/Serrate/Lag-1, a constitutive repressor on

target genes that binds the core sequence CGTGGGAA and other related sequences. NIC

then displaces co-repressors (SMRT, N-coR, SKIP, and histone deacetylases) (Hsieh et

al., 1999), and recruits co-activators such as p300, histone acetyl transferases, and

mastermind-like 1 (MAML1-3) (Wu et al., 2000). Notch is known to activate many

genes including, the pro-differentiation transcription factors, HES and HEY family of

Page 45: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

CBF-1

CoR

CoACoA

Golgi

Furin-likeConvertase

HEYHES

Cyclin D1

++

ADAM

Gamma-secretase

Jagged-1

NIC

1

2 3

Notch-1

Endocytosis

PEN2

NicastrinPresenilin

APH-1

MAML-1

NCoRSMRT

CBF-1

CoR

CoACoA

Golgi

Furin-likeConvertase

HEYHES

Cyclin D1

++

ADAM

Gamma-secretase

Jagged-1

NIC

1

2 3

Notch-1

Endocytosis

PEN2

NicastrinPresenilin

APH-1

MAML-1

NCoRSMRT

24

RECEPTORS: LIGANDS: Notch-1 Jagged-1 Notch-2 Jagged-2 Notch-3 Delta-like 1 Notch-4 Delta-like 2

Delta-like 4

Figure 3: The Notch Signaling Pathway. Abbreviations- a disintegrin and metalloproteases, ADAM; Notch intracellular protein, NIC; co-activators, CoA; co-repressors, CoR; mastermind-like 1, MAML-1.

Page 46: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

25

basic helix-loop-helix transcription factors, genes implicated in angiogenesis (Fischer et

al., 2004) and in epithelial-to-mesenchymal transition (Iso et al., 2003; Maier and Gessler,

2000; Zavadil et al., 2004), cyclin D1 (Ronchini and Capobianco, 2001), c-Myc (Weng et

al., 2006), NF-κB (Cheng et al., 2001), and the IGF-1R (Eliasz et al., 2010).

2. Notch Receptors

A. Structural Analysis of Notch Receptors

In the mammalian system, there are four Notch receptors (Notch-1,2,3,4)

(Blaumueller and Artavanis-Tsakonas, 1997). The structure of the receptor is conserved,

but emerging evidence demonstrates distinct properties among the different receptors

(Figure 4). The region of Notch that extends into the intercellular space (NEC) consists of

a series of epidermal growth factor-like (EGF-like) repeats. These EGF-like repeats

make up the ligand binding surface. The number of EGF-like repeats varies among the

receptors: Notch-1 having the most and Notch-4 the least. The EGF-like repeats contain

sites for O-linked glycosylation by the O-fucosyl transferase (O-fut) enzyme (Haines and

Irvine, 2003). Glycosylation plays a vital role in the function, folding, and ligand binding

preference of Notch (Sasamura et al., 2003; Shi and Stanley, 2003). Juxtaposed to the

cellular membrane, there are three Lin12 Notch repeats (LNR) that help stabilize and

direct appropriate ligand binding.

The next portion of Notch is the transmembrane region (NTM), which transverses

the lipid bilayer and contains the cleavage site for the γ-secretase complex. The

intracellular portion (NIC) contains a RAM domain (a domain involved in protein

Page 47: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

26 RECEPTORS

RAMANKNCRTAD

PEST

NLS

NLS

LNR

EGFR

NOTCH-1 NOTCH-2 NOTCH-3 NOTCH-4

EXTRACELLULARNOTCH (NEC)

INTRACELLULARNOTCH (NIC)

RAMANKNCRTAD

PEST

NLS

NLS

LNR

EGFR

NOTCH-1 NOTCH-2 NOTCH-3 NOTCH-4

EXTRACELLULARNOTCH (NEC)

INTRACELLULARNOTCH (NIC)

LIGANDS

EGF

JAGGED FAMILY DELTA-LIKE FAMILY

Jagged-1Jagged-2

Delta-like 1Delta-like 2Delta-like 4

CR

DSL

EGF

JAGGED FAMILY DELTA-LIKE FAMILY

Jagged-1Jagged-2

Delta-like 1Delta-like 2Delta-like 4

CR

DSL

JAGGED FAMILY DELTA-LIKE FAMILY

Jagged-1Jagged-2

Delta-like 1Delta-like 2Delta-like 4

JAGGED FAMILY DELTA-LIKE FAMILY

Jagged-1Jagged-2

Delta-like 1Delta-like 2Delta-like 4

CR

DSL

Figure 4: The Four Mammalian Notch Receptors, Their Ligands, and Their Domains.

Abbreviations- epidermal growth factor repeats, EGF; Lin12, LNR; ankyrin repeats, ANK; nuclear localization signal, NLS; cytokine response element, NCR; transactivation domain, TAD; proline glutamic acid serine threonine, PEST; Notch extracellular region, NEC; Notch intracellular region, NIC; cysteine rich region, CR.

Page 48: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

27

interactions), ankyrin repeats (site of protein-protein interactions), two nuclear

localization sequence (NLS; Notch-1 contains three), and a C-terminal PEST domain (a

signal region for protein degradation). Notch-1 and Notch-2 contain transactivation

domains (TAD) and cytokine response elements (NCR), where as Notch-3 only contains

a NCR region (Kopan and Ilagan, 2009). All these elements together are vital to the

successful activity of Notch signaling.

B. Notch Receptors and Breast Cancer

The Notch receptors have been linked to many types of cancers and are found to

play a vital role in the development and progression of mammary tumorigenesis (Miele et

al., 2006). For example, elevated expression of Notch-1 and its ligand Jagged-1 in

ERalpha negative, basal-like, triple negative, or HER2 positive cells are associated with

poorest overall patient survival (Dickson et al., 2007; Reedijk et al., 2005).

Using a xenograft MDA-MB-231 model expressing active Notch-2 (N2IC), there

was overall decreased tumor take and increased apoptosis (O'Neill et al., 2007). In

agreement in endothelial cells, expression Notch-2 (N2IC) stimulated pro-apoptotic genes

such as caspases 3 and 7, as well as PARP cleavage (Quillard et al., 2009). These studies

imply that unlike Notch-1, Notch-3, or Notch-4, Notch-2 has a pro-apoptotic and possibly

tumor suppressor function. This brings to question whether pan-pharmacological Notch

inhibitors should be used, or should more specific targeting be explored.

Notch-3 has also been implicated in mammary tumor formation as well as

mammary gland development. Similar to studies performed using Notch-1, MMTV-

Page 49: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

28

driven Notch-3 (N3IC) transgenic mice showed similar exacerbation of mammary

tumorigenesis (Hu et al., 2006). In HER2 negative breast cancers, downregulation of

Notch-3 resulted in decreased proliferation and increased apoptosis even more than

Notch-1 (Yamaguchi et al., 2008). Similar to Notch-4, Notch-3 promotes self-renewal

and expansion of mammary gland stem and progenitor cells (Sansone et al., 2007). Thus,

Notch-3 like Notch-1 and Notch-4 plays a critical role in the transformation and

progression of breast cancer.

The Notch-4 gene contains a site of integration (int-3) for retroviral insertion such

as mouse mammary tumor virus (MMTV) (Uyttendaele et al., 1996). This site occurs

downstream of the Lin12 portion, yet upstream of the transmembrane portion of Notch

(see Figure 4). Loss of the extracellular portion from Lin12 to the N-terminus results in a

truncated C-terminus form of Notch-4 that is constitutively active. Notch-1 has similar

integration sites and is found to also be constitutively active (Dievart et al., 1999). In a

separate study, active intracellular Notch-1 or Notch-4 (N1IC and N4IC) was demonstrated

to form spontaneous mammary tumors in mice (Callahan and Raafat, 2001). Recently,

Notch-4 has been implicated in survival and proliferation of tumor initiating or mammary

cancer stem cells (Harrison et al., 2010). Aberrant Notch signaling has become evident

not only in tumor initiation, but also progression playing a role in undifferentiated stem-

like and more terminally differentiated neoplastic cells.

Page 50: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

29

3. Notch Ligands

In the mammalian system, there are five known ligands (Delta-like 1,2,4, and

Jagged 1,2) (Dunwoodie et al., 1997; Lindsell et al., 1995; Shawber et al., 1996). The

ligands are divided into two groups depending upon whether or not they contain a

cysteine rich region (Delta-class or Jagged, respectively) (see Figure 4). In common,

both classes have a DSL domain and a series of EGF-like repeats, which are essential for

Notch receptor binding. Notch receptor binding to its ligand on an adjacent cell is critical

for proteolysis and signaling events. However in Drosophila, cleaved and/or soluble

ligands have been found to activate or downregulate the signaling cascade independent of

cell to cell contact depending upon context (Klueg et al., 1998; Sun and Artavanis-

Tsakonas, 1997). Interestingly during hematopoesis, these “decoy” ligands provide an

exciting new strategy to modulate the Notch pathway, which could be exploited in other

biological systems (Masuya et al., 2002). High Jagged-1 expression has been correlated

with the poorest overall patient survival in ERalpha negative breast cancer subtypes

including the basal-like, triple negative, and HER2 positive tumors (Dickson et al., 2007;

Reedijk et al., 2005).

4. Inhibitors of Notch Signaling

With the understanding that ligand engagement is critical for Notch activation,

receptor decoys could be a means to interrupt the ligand-receptor mediated activation

(Nickoloff et al., 2002). Disruption of the O-linked glycosylation required for mature

receptor formation could also be used to inhibit the activity of Notch. Specific Notch

Page 51: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

30

inhibition could be achieved by using siRNA, antisense, or monoclonal antibodies

directed at specific receptors (Miele et al., 2006). Other methods could be mastermind

decoys, RAS inhibitors, or even transcription factor-stapled peptides that disrupt the co-

activation complex in the nucleus (Moellering et al., 2009).

Enzymatic inhibition has shown promise in dampening Notch signaling. Anyone

of the three enzymatic cleavage steps that process Notch (furin-like convertase [S1],

ADAM10/17 [S2], or γ-secretase [S1]) (see Figure 3 for more detail) may be used as a

means to attenuate signaling. Of great interest is the final cleavage step required for the

liberation of the active, intracellular form of Notch (NIC) to activate Notch signaling

(Kopan and Ilagan, 2004; Kopan and Ilagan, 2009). Its inhibition can be exploited

through emerging pharmacological drugs identified as γ-secretase inhibitors (GSIs),

which inhibit signaling from all four receptors. These can be either peptide-mimics or

small molecule inhibitors. One such GSI is the compound developed by Merck

Pharmaceuticals known as MRK-003 (Figure 5) and used in this investigation (Lewis et

al., 2007). This compound is a small medicinally modified chemical that binds to the

ATP-dependent binding pocket of presenilin, the catalytic subunit of the γ-secretase

complex. Since this is an experimental drug designed by Merck Pharmaceuticals, the

rates, pharmacokinetics, and pharmacodynamics are confidential information secured by

the company. Other GSI compounds include LY 411,575, a reversible competitive

inhibitor, Calbiochem, an irreversible covalent suicide inhibitor (see Figure 5 for

chemical structures), and Z-Leu-Leu-Nle-CHO, a tripeptide inhibitor (Curry et al., 2005).

Recent studies demonstrate that GSI treatment in combination with tamoxifen inhibits

Page 52: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

31

F

F

OHOON

NH

NH

CH3 O

CH3

LY 411,575

OO NH

NHNH

O

O

O

CH3

CH3

(CH2)3CH3CH3

CH3

Calbiochem (C26H41N3O5)

S

N

NH

N

O

O

F

FF

F

FF

MRK-003

Figure 5: Chemical Structures of Common GSI(s).

Page 53: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

32

ER+ breast tumor growth in athymic, nude mice (Rizzo et al., 2008) providing evidence

of novel therapeutic approaches to enhancing current treatments. Certain GSI(s) are

currently in clinical trials for the treatment of breast cancer.

5. Regulation of Notch Signaling

A. Post-transcriptional Regulation of Notch

Notch signaling is regulated post-transcriptionally by several different methods.

Fringe glycosylation of the extracellular portion of Notch determines the relative affinity

for contact with its ligands (Okajima and Irvine, 2002). Phosphorylation events on Notch

also aid its signaling sustainability such as phosphorylation by GSK-3beta (Foltz et al.,

2002). Ubiquitination by E3 ligases such as Neuralized and Mindbomb are important for

endocytosis of Notch ligands and subsequent activation events (Le Borgne et al., 2005;

Wang and Struhl, 2005). Alpha-adaptin mediated endocytosis by negative regulator

Numb also plays a critical role in the regulation of Notch signaling (Berdnik et al., 2002).

Numb is shown to interact with the intracellular Notch domain and recruit the Nedd4

family of E3-ubiquitin ligases. This leads to the ubiquitination and degradation of Notch

IC (McGill et al., 2009; McGill and McGlade, 2003). Other post-transcriptional events

are currently being investigated and could be manipulated as a means to dampen Notch

signaling in cancer. However, Notch is already processed and reaching maturity by this

time. A better option could be to understand the transcriptional regulation of Notch and

terminate its transcription prior to protein synthesis.

Page 54: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

33

B. Transcriptional Regulation of Notch

Notch begins its life in the nucleus as a gene awaiting transcription. The factors

that are transcriptional regulators of the Notch receptors are still widely unknown. It was

shown in human keratinocytes that p53 regulates the Notch-1 receptor (Alimirah et al.,

2007). In vascular endothelial cells, AP-1 was shown to be important in the regulation of

Notch-4 transcription (Wu et al., 2005). Demonstrated by the same group, AP-1 was

shown to stabilize the glucocorticoid receptor on the Notch-4 promoter in vascular

endothelial cells and aid in its expression (Wu and Bresnick, 2007). During T-cell

development, NKAP was shown to be a Notch transcriptional repressor (Pajerowski et al.,

2009). During hair shaft differentiation, both Msx2 and Foxn1 were demonstrated to be

responsible for Notch-1 expression (Cai et al., 2009). However, more detailed

transcriptional regulators are left widely understudied, particularly in breast cancer. Thus,

novel regulators of the Notch pathway will prove to be significant in the fight against

breast cancer.

Promoter analysis of Notch genes reveals evidence of p53, AP-1, SP-1, Ets, as

well as several other consensus sites that may all play a role in the transcriptional

regulation of Notch which is most likely cell context-dependent. In breast cancer,

particularly the basal-like/triple negative breast cancer, used in this investigation, p53

mutations are known to exist, which raises the question of what else is regulating these

transcripts. Evidence of Ets consensus sequences is exciting, due to the fact that PEA3, a

well known member of the Ets family of transcription factors, is overexpressed in

Page 55: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

34

aggressive breast cancers. Therefore, could the transcription factor PEA3 be responsible

for the regulation of Notch in breast cancer?

Polyomavirus Enhancer Activator 3 (PEA3)

1. Ets Transcription Factors

Ets proteins are a family of transcription factors of which there are over 30 known

members. The members are categorized into 13 groups: Ets, TEL, YAN, SPI, ERG,

PEA3, ELF, DETS4, ELK, GABP, ER71, ERF, and ESE. These transcription factors

share a common DNA Ets domain which binds through its winged helix-turn-helix motif

(Graves and Petersen, 1998; Laudet et al., 1999) to the canonical 11 base pair DNA

sequences with the core being GGAA/T on target genes (Sharrocks, 2001). The affinity

of binding relies on the proximal sequences surrounding the Ets binding site. The

domain is important for protein-protein co-regulatory interactions. Although there is

marked similarities among the Ets domain, differences in a single amino acid within the

C-terminal portion of the Ets domain can lead to altered DNA specificity and interactions

with other transcription factors (Fitzsimmons et al., 1996; Shore et al., 1996). Activation

and/or repression domains are often present in these factors, which aid their

transcriptional control based on context (Oikawa and Yamada, 2003). This further

individualizes and separates the Ets transcription factors into unique groups.

Page 56: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

35

2. Polyomavirus Enhancer Activator 3 (PEA3)

Polyomavirus enhancer activator 3, commonly known as PEA3, E1AF, or ETV4,

is a member of the Ets family of transcription factors and located on chromosome 17q21

(Oikawa and Yamada, 2003). Other family members with PEA3 are ER81 and Erm.

PEA3 is important in embryogenesis and is rarely expressed at elevated levels during

adulthood. Interestingly, PEA3 levels re-emerge during remodeling events and cancer

progression. Approximately 76% of human breast tumors contain elevated levels of

PEA3; the highest found in the triple negative breast cancers conferring overall poor

prognosis (Trimble et al., 1993). Interestingly, mRNA levels of PEA3 demonstrated

marked over-expression, yet endogenous PEA3 protein is rarely detectable (Baert et al.,

1997). This may be impart do to the rapid turnover rate and protein stability (Baert et al.,

1997). Several studies associate PEA3 in the regulation of enzymes involved in tissue

remodeling and invasion such as matrix metalloproteases (MMP-1, MMP-3, MMP-7,

MMP-9) (Benbow and Brinckerhoff, 1997), urokinase-type plasminogen activator (uPA)

(Evans et al., 2001), cyclooxygenase (COX-2) (Subbaramaiah et al., 2002), HER2/ErbB-

2 (Matsui et al., 2006), and IL-8 (Davidson et al., 2003) implicating it as a potential target

for therapeutics in a diseased state. However, little concurring evidence of a mechanistic

role of PEA3 in breast cancer has left the field widely controversial.

A. The Structure of PEA3

The gene is made up of 13 exons spanning approximately 15 kilobases on

chromosome 17q21 (Coutte et al., 1999). The 62 kD protein contains an Ets DNA

Page 57: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

36

binding domain with a tryptophan repeat and a conserved trans-activation domain flanked

by negative regulatory domains (Figure 6) (Bojovic and Hassell, 2001). It also contains

an acidic region and glutamine-rich region, which have activation capabilities (Higashino

et al., 1993). The C-terminus domain of PEA3 has been found to be important to regulate

its stability (Takahashi et al., 2005).

B. Activity and Regulation of PEA3

Activation of PEA3 is through phosphorylation of serine and threonine residues

by the mitogen activation protein kinase pathway (MAPK: Ras, Raf-1, MEK, ERK-1, and

ERK-2) (Wasylyk et al., 1998). The protein is relatively unstable exhibiting a turnover

half-life of of about 30 minutes via the ubiquitin-proteasome pathway (Takahashi et al.,

2005). Recently, PEA3 was suggested to be negatively regulated in part by sumoylation

(Bojovic and Hassell, 2008; Takahashi et al., 2005). SUMO-E3 ligase PIASy has been

implicated directly in sumoylating PEA3 (Nishida et al., 2007). Sumoylation sites have

been identified as lysines 96, 222, and 256 (Bojovic and Hassell, 2008), and have been

demonstrated to either transiently enhance the activity of PEA3 or mark it for degradation

by the ubiquitin-proteasome pathway (Guo and Sharrocks, 2009). Therefore, there is a

direct connection and interplay between both sumoylation and ubiquitinylation pathways.

3. PEA3 and Breast Cancer

PEA3 is overexpressed in 76% of breast cancers. The highest expression is

observed in invasive breast cancers particularly the triple negative subtype (Trimble et al.,

Page 58: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

37

Figure 6: Protein Structure of Polyomavirus Enhancer Activator 3 (PEA3). Abbreviations- phosphorylation motif, P; transactivation domain, TAD; threonine, T; serine, S; lysine, K; sumoylation moiety, SUMO.

Page 59: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

38

1993), which is associated with overall poor prognosis, and is also highly expressed in

HER2 positive tumors (Benz et al., 1997). PEA3 is present during embryogenesis and

expressed within the epithelial buds of the mammary gland (Chotteau-Lelievre et al.,

1997; Chotteau-Lelievre et al., 2003). Later studies confirmed the presence of PEA3 in

undifferentiated epithelial cap cells of the terminal end buds (Shepherd and Hassell,

2001). In a mouse model of MMTV-neu-induced mammary primary adenocarcinomas,

overexpression of PEA3 resulted in enhanced metastatic lesions (Trimble et al., 1993).

Conversely, expression of a dominant negative form of PEA3 resulted in a delay of tumor

onset and progression in a MMTV-neu mouse model (Shepherd et al., 2001). PEA3 also

enhances transcriptional activity of important genes involved in breast cancer metastasis

including, but not limited to cyclooxygenase-2 (Subbaramaiah et al., 2002), MUC4

(Perez et al., 2003), and osteopontin (El-Tanani et al., 2004). Taken together, PEA3 has

been correlated with tumor initiation, progression, and invasiveness and appears to play a

vital role in breast cancer.

Interestingly, PEA3 has been implicated recently in multipotent progenitor

regulation of lineage-specific differentiation (Kurpios et al., 2009) implying a potential

function in mammary stem cells. This is similar to the recent study mentioned previously

regarding Notch-4 in survival of tumor initiating cells/cancer stem cells (Harrison et al.,

2010). This may provide an exciting correlation between PEA3 and Notch-4 expression.

Conflicting studies also have been shown regarding the role of PEA3 in regulation

of HER2 and its clinical correlations. Originally, it was identified that the HER2

promoter contained a conserved Ets consensus site that when mutated dampened HER2

Page 60: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

39

transcription (Scott et al., 1994). Shortly after, PEA3 was shown to be upregulated in

93% of HER2/Neu-overexpressing human breast tumors and correlated with breast

cancer initiation and progression (Benz et al., 1997). Again in 2001, PEA3 was linked to

the positive expression of HER2 helping confirm the potential of PEA3 as a therapeutic

target (Shepherd et al., 2001).

However, other studies demonstrated that PEA3 repressed HER2 expression and

that PEA3 does not positively correlate with HER2 expression in breast cancer (Xia et al.,

2006; Xing et al., 2000). Although the field presents a “gray area” regarding the role of

PEA3 in HER2 regulation, this controversy actually provokes the notion of dynamic

molecular variability. The role of PEA3 on other genes in different systems implies the

importance of cellular context. In fact, further studies demonstrated that PEA3 and c-

JUN independently had slight roles in the regulation of HER2. However, their coordinate

expression along with histone acetyltransferase p300 stimulated transcription of the

HER2 gene by 20-fold in the MCF-7 breast cancer cell line (Matsui et al., 2006). This

stresses the importance of transcription factors such as c-JUN working in tandem with

PEA3 to appropriately regulate PEA3 target genes. Moreover, PEA3 directly interacts

with p300 (Liu et al., 2004) and USF-1 (Firlej et al., 2005), and many other co-regulatory

factors to enhance gene transcription. Therefore could PEA3 be responsible with other

factors such as AP-1 for the regulation of Notch transcription in breast cancer?

Page 61: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

40

Activating Protein 1 (AP-1)

A common player in transcriptional control and implicated in various cellular

reactions is the homo- or heterodimeric transcription factor complex AP-1. The dimeric

complex classically consists of the FOS (c-FOS, FosB, Fra-1, Fra-2) and JUN (c-JUN,

JunB, and JunD) families (Curran and Franza, 1988). Other dimeric complexes can also

include ATF and MAF protein families. c-JUN:c-JUN homodimers can form, but are

less stable than c-JUN:c-FOS heterodimers (Smeal et al., 1989). Homodimers of c-FOS

have yet to be identified (Figure 7). AP-1 contains a leucine zipper motif and binds

through its basic DNA binding domain (DBD) to a canonical DNA binding element:

TGAg/cTCA. This is known as the TPA-responsive element (TRE) named after its

enhanced DNA binding to this consensus site upon treatment with 12-O-

tetradecanoylphorbol-13-acetate (TPA), a tumor promoter. AP-1 is activated through

phosphorylation by the mitogen activating pathway (MAPK) or the stress pathway (JNK)

(Chen et al., 1996). It is negatively regulated by the ubiquitin-proteasome degradation

pathway.

AP-1 cooperates with other proteins including, but not limited to, NFκB,

CBP/p300, Rb, and PEA3 (Hesselbrock et al., 2005; Matthews et al., 2007). AP-1

members generally favor a dynamic “switch of partners” to mediate targeted effects as

opposed to a “static predetermined partner” model. Expression profiles and partnerships

of the different members vary among cell types, even in different breast cancers (Table 2).

The functional role of AP-1 is to promote proliferation, differentiation, inflammation,

Page 62: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

41

Jun-Jun Jun-Fos Jun-ATF

c-Fos Fra-1JunB JunD FosBc-Jun Fra-2

MDA-MB-231

JUN FAMILY FOS FAMILY

Jun-Jun Jun-Fos Jun-ATFJun-Jun Jun-Fos Jun-ATF

c-Fos Fra-1JunB JunD FosBc-Jun Fra-2

MDA-MB-231

JUN FAMILYJUN FAMILY FOS FAMILYFOS FAMILY

Figure 7: AP-1 Members and Dynamic Partnerships.

AP-1 Reference Transformed Invasive Non-invasive Members Normal HBL-100 MDAMB231 T47D

c-FOS ---- High High Low FosB Low ---- ---- High Fra-1 ---- Medium High ---- Fra-2 ---- Low Low ---- c-Jun ---- Low High Low JunB Low ---- Low Low

junD Low High High High

Table 2: Relative Levels of AP-1 Family Members in Breast Cancers. AP-1 expression pattern was determined in separate cell types: HBL-100 cells, transformed mammary epithelial cells derived from a normal lactating breast, MDA-MB-231 cells which are highly invasive, and T47D cells which are weakly or non-invasive as compared to normal breast tissue as a base reference (Bamberger et al. 1999).

Page 63: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

42

and/or apoptosis. The perturbed expression and activity of AP-1 varies among cancers,

and is positively linked to tumor promotion and progression.

1. The c-JUN Family

A. c-JUN

c-JUN was originally identified and characterized as the AP-1 founding member.

The protein is approximately 330 amino acids in length and harbors many domains

important for its function (Hattori et al., 1988). c-JUN contains a N-terminal

transactivation domain (TAD), a domain consisting of a stretch of basic residues

considered the DNA binding domain (DBD), and the C-terminal leucine zipper domain

(ZIP) (Figure 8). The basic DNA binding region allows for appropriate DNA binding by

interaction with the negatively charged DNA backbone. The C-terminal leucine zipper

motif aids in protein-protein interactions and dimerization between AP-1 partners.

Activation of c-JUN occurs through phosphorylation events by MAPK or JNK within the

transactivation domain at serines 63 and 73, and threonines 91 and 93 (Smeal et al., 1991).

Phosphorylation events are important for the stability and interaction with other proteins.

This transactivation domain has been completely removed to create a dominant negative

form of c-JUN and consequently the AP-1 complex itself, known as TAM-67 (Figure 8).

This was originally created by Brown et al. in 1993 (Brown et al., 1993). TAM-67 is a

transdominant inhibitor of AP-1, which can bind DNA but quenches AP-1 complexes by

competing for wildtype c-JUN and c-FOS members (Brown et al., 1994; Brown et al.,

1996) thus dampening AP-1 activity. Like PEA3, ubiquitinylation directs c-JUN

Page 64: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

43

H N3 COOH

TAD DBD ZIP

H N3 COOH

DBD ZIP

c-JUN

TAM-67

TTSS

63 73 91 93H N3 COOH

TAD DBD ZIP

H N3 COOHH N3H N3 COOH

TAD DBD ZIP

H N3 COOH

DBD ZIP

H N3 COOHH N3H N3 COOH

DBD ZIP

c-JUN

TAM-67

TTSS

63 73 91 93

Figure 8: Protein Structure of c-JUN and its Dominant Negative Form, TAM-67. Abbreviations- serine, S; threonine, T; transactivation domain, TAD; DNA binding domain, DBD; basic leucine zipper domain, bZIP.

Page 65: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

44

proteasome-mediated degradation. Interestingly, c-JUN knockout mice are embryonic

lethal showing effects on liver, heart, and hepatoblasts (Eferl and Wagner, 2003).

The critical member of the AP-1 family, c-JUN is associated with cancer acting as

a major transcription factor. It is overexpressed in triple negative breast cancer cells,

MDA-MB-231 cells (Bamberger et al., 1999). c-JUN is associated with proliferation

showing marked proliferation alterations in c-JUN deficient fibroblasts (Wisdom et al.,

1999). Also, c-JUN cooperates with p300 and PEA3 to promote HER2 mediated

transcription in MCF-7 cells, luminal A HER2 expressing breast cancer cells (Matsui et

al., 2006). Correlation studies determined that c-JUN is involved in both proliferation,

hyperphosphorylated Rb, angiogenesis, and vascular endothelial growth factor (VEGF),

showing expression on 38% of invasive breast cancers in 103 breast cancer cases

(Vleugel et al., 2006). Interestingly, high expression of c-JUN has been associated with

insensitivity to chemotherapy and drug resistance in MCF-7 breast cancer cells (Daschner

et al., 1999). Other associations regarding cellular proliferation have shown that c-JUN

expression and activity upregulates cyclin D1, a critical mediator of cell cycle

progression from G1-S phase (Albanese et al., 1995; Bakiri et al., 2000; Herber et al.,

1994) and downregulates p16, an important cell cycle G1/S checkpoint inhibitory protein

(Passegue and Wagner, 2000). In contrast, another member of the JUN family, JunB,

downregulates cyclin D1 (Bakiri et al., 2000) and upregulates p16 (Passegue and Wagner,

2000), which limits cell proliferation and provides an interesting and distinctive role

among each AP-1 member.

Page 66: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

45

B. JunB

JunB has a similar structure to c-JUN, but lacks the important serines 63 and 73

phosphorylation sites on (Kallunki et al., 1996), which may be responsible for the

interesting and opposite role JunB has on cellular transformation. Many studies have

reported an antagonism between c-JUN and JunB (Bakiri et al., 2000; Passegue and

Wagner, 2000); their expression tend to be inversely proportional (Shaulian and Karin,

2002). Its expression in MDA-MB-231 cells is very low (Bamberger et al., 1999). JunB

is less active than c-JUN. c-JUN and c-FOS heterodimers are much more potent

activators of gene transcription than c-JUN and JunB dimers (Deng and Karin, 1993).

This opposite effect is illustrated in overexpression studies in which c-JUN expression

alone can transform immortalized rat fibroblasts, but JunB expression has no

transformation capability (Schutte et al., 1989). This interesting opposition only stresses

the notion that cellular context and dynamic partnership between AP-1 members and their

interacting protein partners determines the appropriate cellular response.

C. JunD

JunD has a similar structure as the other JUN family members, but unlike JunB,

JunD contains the serines 63 and 73 phosphorylation sites as c-JUN. The functional role

of JunD is still being discerned showing both positive and negative effects on cell

proliferation and apoptosis (Pfarr et al., 1994; Weitzman et al., 2000). JunD null mice do

not form spontaneous tumors, yet inhibition of JunD suppresses neoplastic growth

indicative of a tumor promoting effect (Agarwal et al., 1999; Thepot et al., 2000). In a

Page 67: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

46

study investigating 53 mammary carcinomas and three cell lines, relative expression of

JunD varied greatly with no distinct pattern (Bamberger et al., 1999). It is expressed in

MDA-MB-231 cells (Bamberger et al., 1999), but its functional role is relatively

unknown. To further demonstrate the spurious nature of JunD, studies have shown that

when the interaction with JunD and menin, a known tumor suppressor, is abolished, JunD

changes its activity from a tumor suppressive function to one that is tumor promoting

(Agarwal et al., 2003). However, in Ras transformed fibroblasts, JunD protects cells

from oxidative stress and reduces angiogenesis (Gerald et al., 2004). This further

demonstrates the necessity of comprehending appropriate cellular context and not

limiting AP-1 members to a specific function, namely tumor suppressive or tumor

promoting.

2. The c-FOS Family

A. c-FOS

c-FOS was originally identified and characterized when the viral protein bound to

the same consensus as c-JUN (Rauscher et al., 1988). It contains 381 amino acids

consisting of a basic DNA binding domain (DBD), a transactivation domain (TAD), and

a leucine zipper domain (ZIP) (Figure 9). Similarly to c-JUN, c-FOS has been exploited

to create a dominant negative AP-1 known as A-FOS. A-FOS is mutant form of c-FOS

in which the DNA binding domain has been mutated (Figure 9) (Olive et al., 1997). The

dominant negative mutant incorporates an acidic extension made up of many negatively

charged glutamate amino acids (Olive et al., 1997). This charge reversal

Page 68: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

47

H N3 COOH

DBD ZIPTAD

H N3 COOHH N3H N3 COOH

DBD ZIPTAD

H N3 COOH

DBD ZIPTAD

H N3 COOHH N3H N3 COOH

DBD ZIPTAD

A-FOS

c-FOS

TTS

232 325 331 374

T

LEQRAEE LARENEE LEKEAEE LEQELAE LE

ACIDIC EXTENSION

Figure 9: Protein Structure of c-FOS and its Dominant Negative Form, A-FOS. Abbreviations- serine, S; threonine, T; leucine, L; transactivation domain, TAD; DNA binding domain, DBD; basic leucine zipper domain, bZIP.

Page 69: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

48

destroys the intermolecular charge attraction between the negative DNA backbone and

the DNA binding domain of A-FOS, thus dampening AP-1 activity (Ransone et al., 1990).

c-FOS heterodimerizes with c-JUN to form a stable dimeric transcription factor capable

of regulating genes important in cell proliferation, differentiation, and apoptosis

(Shaulian and Karin, 2001). Homodimers of c-FOS do not exist, but additional binding

partners of c-FOS are MAF proteins (Kataoka et al., 1996), CBP (Bannister and

Kouzarides, 1995), and GATA-4 (McBride et al., 2003). Similar to c-JUN, c-FOS is

phosphorylated on threonine 232 by MAP kinases (Deng and Karin, 1994) and by ERKs

on threonines 325, 331, and 374 (Monje et al., 2003) to enhance its activity. c-FOS relies

on dimerization for its activity, since it is highly unstable and rapidly degraded by the

ubiquitin-meditated proteasome degradation (Bossis et al., 2003).

In cancer, c-FOS upregulates genes responsible for angiogenesis such as VEGF

(Marconcini et al., 1999) and MMP-1 and MMP-3 (Hu et al., 1994). It has also been

shown that c-FOS upregulates DNA 5-methylcytosine transferase responsible for

methylating and silencing tumor suppressor genes (Bakin and Curran, 1999).

Alternatively, c-FOS upregulates FAS ligand (FASL) to induce apoptosis through the

extrinsic death-receptor pathway in T-lymphocytes (Kasibhatla et al., 1998). It is highly

expressed in triple negative breast cancer, MDA-MB-231 cells (Bamberger et al., 1999).

Even though the majority of the literature associates active c-FOS with cancer

progression, the specific role of c-FOS depends on cell type and cellular context. With

this in mind, c-FOS becomes a more dynamic partner relying greatly on other cellular

signals.

Page 70: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

49

B. Fra-1

Elevated levels of AP-1, especially Fra-1, are commonly found in the most

aggressive estrogen receptor negative mammary tumors when compared to less

metastatic estrogen receptor positive mammary tumors (Zajchowski et al., 2001).

Exogeneous overexpression of Fra-1 in the non-invasive MCF-7 breast cancer cells

disturbs cell morphology and increases the invasive potential of the cells. Conversely, if

endogenous Fra-1 is inhibited using RNA interference in MDA-MB-231 cells, overall

motility and aggressiveness are diminished (Belguise et al., 2005). In a cDNA array of

breast cancer subtypes, increased expression of both Fra-1 and c-JUN partners was

correlated to overall breast tumor aggressiveness (Zajchowski et al., 2001). Similarly,

Fra-1 expression in epithelioid adenocarcinoma cells demonstrated a positive effect on

motility and invasiveness (Kustikova et al., 1998). In MDA-MB-231 cells, motility was

not affected by Fra-1, but its invasiveness potential was highly enhanced (Milde-

Langosch et al., 2004). Recently, Baan et al. demonstrated that invasive breast cancer

cells including MDA-MD-231 favored the c-JUN:Fra-1 heterodimer, compared to less

invasive breast cancer cells, which favored the c-JUN:c-FOS heterodimer (Baan et al.,

2010). Fra-1 is known to regulate genes involved in proliferation (p19-ARF) (Ameyar-

Zazoua et al., 2005), invasion (MMP-1,9,) (Milde-Langosch et al., 2004), and

angiogenesis (IL-8) (Freund et al., 2004). Taken together, AP-1 complexes involving

Fra-1 most probably are critical for mammary tumorigenesis and invasion.

Page 71: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

50

C. Fra-2

Most focus on AP-1 has encompassed c-JUN, c-FOS, and Fra-1 leaving the other

members like Fra-2 in the shadows. Nevertheless, Fra-2 does play an interesting role in

development and cancer. Fra-2 is involved in cellular differentiation during development.

The expression pattern of Fra-2 is observed in the epithelia and central nervous system

during mouse development (Carrasco and Bravo, 1995). Its expression in triple negative

breast cancer cells MDA-MB-231 is relatively low (Bamberger et al., 1999). Fra-2

overepression in mouse mammary adenocarinoma cells (CSML0) had no affect on

motility, but in a fibroblastoid cell line, its overexpression effected motility (Tkach et al.,

2003). In MDA-MB-231 cells, Fra-2 expression had a mild positive effect on cellular

invasion (Milde-Langosch et al., 2004). Fra-2 tends to be a modular protein partner not

only relying on cellular context, but also the AP-1 core complex.

D. FosB

FosB is the last member of the FOS family and possesses an opposite function

than the other members. An inverse correlation between FosB and Fra-1 has been

observed. In tumor tissues with high Fra-1 expression, FosB is low, but in tissues where

FosB is elevated, low expression of Fra-1 is observed. Also, association of FosB with

well-differentiated and estrogen positive breast cancer has been observed (Bamberger et

al., 1999). The expression of FosB is relatively undetectable in poorly-differentiated

steroid receptor negative clinical breast cancer tissues, but highly expressed in normal

ductal mammary epithelial cells (Milde-Langosch et al., 2004). Opposite of the effects

Page 72: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

51

demonstrated by expression of c-FOS, Fra-1, and Fra-2, FosB has an inhibitory effect on

cell proliferation in correlation studies using breast cancer samples (Milde-Langosch et

al., 2000). Although FosB has an inverse role than its siblings, the AP-1 partner choice

and directed cellular signals could tip the balance and potentially change its function role.

3. AP-1 and PEA3

AP-1 interacts with partners on the promoter of many genes which is dependent

on cellular context to determine function, Similarly, PEA3 exists in a dynamic

equilibrium relying on partners to determine its function (Kurpios et al., 2003).

Interestingly, PEA3 has been shown to partner with AP-1 on several genes such as MMP-

1, MMP-3, MMP-7, MMP-9 (Benbow and Brinckerhoff, 1997), urokinase-type

plasminogen activator (uPA) (Evans et al., 2001), cyclooxygenase (COX-2)

(Subbaramaiah et al., 2002), HER2/ErbB-2 (Matsui et al., 2006), IL-8 (Davidson et al.,

2003), and mammaglobin (Hesselbrock et al., 2005). AP-1 and PEA3 DNA binding

elements are generally within relative close proximity to each other and generally to the

transcriptional start site (Benbow and Brinckerhoff, 1997; Davidson et al., 2003; Evans et

al., 2001; Hesselbrock et al., 2005; Matsui et al., 2006; Subbaramaiah et al., 2002).

Taken together, AP-1 and PEA3 are most probably dynamic transcriptional partners that

tend to share a common gene regulatory unit.

Page 73: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

52

A Systems Approach: Linking Notch, PEA3, & AP-1

Biological systems are made up of interconnecting networks of cellular

communication. These pathways involve a plethora of proteins that when stretched

beyond homeostasis produces a diseased state. Notch signaling, PEA3, and AP-1 are

shown to be atypically regulated in breast carcinomas. Studies have shown that elevated

levels and/or misdirected signaling of these proteins lead to abnormal cell proliferation,

increased aggressive phenotypes, and overall reduced patient survival (Angel and Karin,

1991; Coutte et al., 1999; Kopan and Ilagan, 2009). In our investigations, initial

promoter scanning indicated proximal AP-1 and PEA3 sites on both the Notch-1 and

Notch-4 genes. With vast research independently correlating Notch, PEA3, and AP-1

with breast cancer, we provide, to our knowledge, for the first time a link between these

signaling pathways, with aim at exploiting and manipulating their function to create

innovative strategies for better targeted treatment of triple-negative breast cancer.

Women diagnosed with triple-negative breast cancer have the worst overall prognosis,

frequently present with metastatic tumors, and have few targeted therapy options to date.

And therefore, there is an immediate need to identify and understand the novel targets

that are responsible for the proliferation, survival, and invasive phenotype of breast

cancer.

Page 74: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

CHAPTER III

MATERIALS AND METHODS

Cell Culture

MDA-MB-231, MCF-7, SKBr3 and BT474 breast cancer cells were purchased

from American Type Culture Collection. All cell lines were supplemented with 100 μM

nonessential amino acid and 1% L-glutamine (2 mM). SKBr3 cells [supplemented with

10% fetal bovine serum (FBS)] and MDA-MB-231 cells (5% FBS) were maintained in

Iscove’s Minimal Essential Media (IMEM). MCF-7 cells (10% FBS) were maintained in

Dulbecco's Modified Eagle Medium: Nutrient Mixture F12 (DMEM/F12). BT474 cells

(10% FBS) were maintained in Dulbecco's Modified Eagle Medium (DMEM). All cells

were maintained at 37°C with 95% O2 and 5% CO2.

Drugs and Chemicals

Gamma-secretase inhibitor (MRK-003) was kindly provided by Merck Oncology

International, Inc. (see Diagram 5 for chemical structure). MRK-003 was dissolved in

the solvent dimethylsulfoxide (DMSO) and stored at -80°C. Lactacystin (L6785) was

purchased from Sigma Aldrich (St. Louis, MO, USA), dissolved in water, and stored at -

20°C until use.

53

Page 75: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

54

Expression Vectors

pcDNA3.1 and pcDNA3.1-PEA3 expression vectors were kindly provided by Dr.

Mein-Chie Hung (The University of Texas, MD Anderson Cancer Center, Houston, TX,

USA). Contol CMV-500, CMV-500-AFOS, Control pHMB, and pHMB-TAM67 vectors

were kindly provided by Dr. Richard Schultz (Loyola University Medical Center,

Maywood, IL, USA).

RNA Interference and Reagents

All small interfering RNA (siRNA) reagents were purchased from Santa Cruz

Biotechnologies (Santa Cruz, CA, USA). Transfection reagents were Lipofectamine

2000 and Lipofectamine RNAiMAX purchased from Invitrogen (Carlsbad, CA, USA),

and Fugene 6 was purchased from Roche Diagnostics Corporation (Indianapolis, IN,

USA). Protocols were performed according to the manufacturer’s instructions. Co-

transfections of DNA plasmid and siRNA were performed with Lipofectamine 2000.

The siRNA sequences are shown in Table 3.

Antibodies

Notch-1(C-20)R: (sc-6014-R), Notch-4(H-225): (sc-5594), PEA3 (16): sc-113, c-

FOS(H-125): (sc-7202), c-JUN(G-4): sc-74543, FRA-1(R-20): (sc-605) were purchased

from Santa Cruz Biotechnologies (Santa Cruz, CA, USA). Beta-actin (AC-15): (A5441)

was purchased from Sigma Aldrich (St. Louis, MO, USA) and used as the internal

Page 76: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

55

siRNA Catalog # Sequence Control sc-37007 5’-UUCUCCGAACGUGUCACGU-3’

Notch-1 sc-36095 5’-CACCAGUUUGAAUGGUCAA-3’ 5’-CCCAUGGUACCAAUCAUGA-3’ 5’-CCAUGGUACCAAUCAUGAA-3’

PEA3 sc-36205 5’-CCAGACAAAUCGCCAUCAA-3’ 5’-CGCUCCGAUACUAUUAUGA-3’ 5’-CCAACCACAGAGAACAAGA-3’

c-JUN sc-29223

5’-GUGACGGACUGUUCUAUGA-3’ 5’-CCAGAAAGGAUAUUUAAGA-3’ 5’-GAUGGCCUUUGCUUAUGAA-3’ 5’-GCAUCAUCUGUAGAUACUA-3’

c-FOS sc-29221

5’-CAAGGUGGAACAGUUAUCU-3’ 5’-GGCUUCCCUUGAUCUGACU-3’ 5’-CCGAGCCCUUUGAUGACUU-3’ 5’-GGCAAUAGUGUGUUCUGAU-3’

Fra-1 sc-35405 5’-CCAGCAACUUCUUCUCCAU-3’ 5’-CUGACCAUAUUGUGCUUCA-3’ 5’-CGGAUCUCAGCUUUGAGAA-3’

Table 3: Sequences of siRNA(s).

TARGETS FORWARD PRIMERS REVERSE PRIMERS Notch-1 5'-GTCAACGCCGTAGATGACC-3' 5'-TTGTTAGCCCCGTTCTTCAG-3'

Notch-2 5'-TCCACTTCATACTCACAGTTGA-3' 5'-TGGTTCAGAGAAAACATACA-3'

Notch-3 5'-GGGAAAAAGGCAATAGGC-3' 5'-GGAGGGAGAAGCCAAGTC-3'

Notch-4 5'-AACTCCTCCCCAGGAATCTG-3' 5'-CCTCCATCCAGCAGAGGTT-3'

Hes-5 5'-ATGGCCCCCAGCACTGTGGCC-3' 5'-CTGACAGCCATCTCCAGGATG-3'

Hey-1 5'-TGGATCACCTGAAAATGCTG-3' 5'-TTGTTGAGATGCGAAACCAG-3'

cIAP2 5'-GGACTCAGGTGTTGGGAATCTG-3' 5'-CCTTTAATTCTTATCAAGTACTCACACCTT-3'

HPRT 5'-ATGAACCAGGTTATGACCTTGAT-3' 5'-CCTGTTGACTGGTCATTACAATA-3'

PEA3 5'-AGGAGACGTGGCTCGCTGA-3' 5'-GGGGCTGTGGAAAGCTAGGTT-3'

COX-2 5'-CAGGATACAGCTCCACAGCA-3' 5'-ATCACAGGCTTCCATTGACC-3'

MMP-9 5'-TCGTGGTTCCAACTCGGTTT-3' 5'-GCGGCCCTCGAAGATGA-3'

IL-8 5’-CACCGGAAGGAACCATCTCACT-3’ 5’-TCAGCCCTCTTCAAAAACTTCTCC-3’

Table 4: Sequences of PCR Primers.

Page 77: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

56

control. Secondary antibodies include donkey anti-mouse IgG-HRP (sc-2314) and

donkey anti-rabbit IgG-HRP (sc-2313) (Santa Cruz, CA, USA).

Real-Time RT-PCR

MDA-MB-231 (4x105), MCF-7 (4x105), SKBr3 (5x105), and BT474 (5x105) cells

were plated in a 6-well tissue culture treated dish. Twenty-four hours later, the cells were

transfected with appropriate reagents (7 μL of lipofectamine 2000 for plasmids or 5 μL of

lipofecatime RNAiMAX for siRNA and 5 μL of siRNA reagents (16 nM) and/or 3 μg of

pcDNA3.1-PEA3) and maintained at 37°C 95% O2 and 5% CO2. Forty-eight hours later,

cells were harvested. Total RNA was extracted from the cells using the RNA extraction

kit: RNeasy® Mini Kit (Qiagen, Valencia, CA, USA). Total RNA recovered was then

quantified using the NanoDrop Spectrophotometer (Thermo Scientific). The cDNA was

reverse transcribed (RT) from 1 μg total RNA extracted in a total 100 μL volume

containing 1X RT buffer, 5.5 mM MgCl2, 500 μM dNTPs, 2.5 μM random hexamers, 0.4

U/μL RNase inhibitors, and 1.25 U/μL RT enzyme (MultiScribe™ Reverse Transcriptase

Kit, Applied Biosystems, Foster City, CA, USA). The parameters were as follows: 10

minutes at 25°C, 30 minutes at 48°C, 5 minutes at 95°C, and held at 25°C until use.

Analysis of mRNA relative fold copy number adjusted to hypoxanthine-guanine

phosphoribosyl transferase (HPRT), an endogenous control, was carried out by real-time,

quantitative PCR using iTaq™ SYBR® Green Supermix with ROX (BioRad, Hercules,

CA, USA). In a 96-optical PCR plate, 2.5 μL of cDNA (10 ng/μL) was added to 22.5 μL

of mastermix (2X Syber green Universal Master Mix, and 1 μM forward/reverse primers).

Page 78: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

57

The quantitative PCR parameters were as follows: the initial denature temperature was

95°C for 10minutes; PCR cycling for 40 cycles was carried out at 95°C for 10 seconds,

and 60°C for 45 seconds. A melt curve was added after completion of the 40 cycles set

by the StepOnePlus thermocycler manufacturer (Applied Biosystems). Cycle number at

threshold (Ct) values were subsequently determined relative to the value for the

quantified cDNA that was amplified. Analysis of mRNA copy number adjusted to

hypoxanthine-guanine phosphoribosyl transferase (HPRT), an endogenous control, was

used to calculate relative fold induction or decrease compared to a control sample. The

PCR primers that were used for detection of specific transcripts are shown in Table 4.

Western Blot Analysis

MDA-MB-231 (4x105) cells were plated in a 6-well tissue culture treated dish.

Twenty-four hours later, the cells were transfected with appropriate reagents (7 μL of

lipofectamine 2000 with plasmid addition or 5 μL of lipofecatime RNAiMAX with

siRNA addition and 5 μL of siRNA (16 nM) reagents and/or 3 μg of pcDNA3.1-PEA3)

and maintained at 37°C with 95% O2 and 5% CO2. Forty-eight hours later, cells were

harvested. The cells were lysed in radioimmunoprecipitation assay buffer (RIPA, pH 8.0

containing 50 mM Tris-HCl, 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium

deoxycholate, 0.1% sodium dodecyl sulfate, 25 mM β-glycerophosphate, 1 mM sodium

orthovanadate, 1 mM sodium fluoride, 1 mM phenylmethylsulfonyl fluoride, 1 mg/mL

aprotinin and 1 mg/mL leupeptin). Lysates were sonicated six times for three seconds

each and incubated on ice for 30 minutes. Lysates were then centrifuged at 10,000 g for

Page 79: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

58

5 minutes at 4°C. The supernatant was isolated for further testing. Protein within the

supernatant was measured using the BCA protein concentration assay (Pierce Chemicals,

Rockford, IL), incubated for 30 minutes at 37°C with 95% O2 and 5% CO2, and

measured using a 96 plate UV/Visible spectrophotometer (PolarStar Omega, BMG

Labtech, Durham, NC). Samples were treated with an equal volume of sample buffer

containing NuPAGE 1X LDS Sample buffer, 1X NuPAGE reducing agent or 1 mM DTT.

Samples were heated at 70°C for 10 minutes. Total denatured proteins in lysates were

added to wells of a 4-12% NuPAGE® Bis-Tris Gels in 3-(N-morpholino)

propanesulfonic acid running buffer (MOPS). The gel was run at 175V for 1.5 hours and

separated proteins were transferred to PVDF membranes at 38V for 2 hours in Transfer

Buffer (25 mM Tris, 192 mM glycine, 10% methanol). The membranes were then

blocked in 5% non-fat milk in TBST buffer (10 mM Tris-HCl, pH 7.5, 150 mM NaCl,

0.05% Tween-20, and 0.2%NP-40 at pH 8.0) for one hour at room temperature while

constantly shaking. Primary antibodies were added (1:200) overnight in 5% non-fat milk

in TBST at 4°C. Blots were washed three times in TBST at intervals of 10 minutes at

room temperature while constantly shaking. Secondary antibodies conjugated to

horseradish peroxidase were added (1:2000) to 5% non-fat milk in TBST for one hour at

room temperature while constantly shaking. Blots were then subjected to a second wash

cycle: washed three times in TBST at intervals of 10 minutes at room temperature while

constantly shaking. Substrate for protein detection was SuperSignal® West Dura

(Thermo Scientific, Rockford, IL, USA) added in equal parts, incubated for 5 minutes at

room temperature while rocking, and visualized by the FUGIFILM™ Las-300 imager.

Page 80: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

59

To re-probe western blots, the membranes washed three times in TBST at intervals of 10

minutes at room temperature while vigorously shaking. Restore Plus Western Blot

Stripping buffer (Thermo Scientific) was added to cover the membrane and incubated for

1 hour at 47°C. The blots were then rocked at room temperature for 30 minutes followed

by three quick rinses with deionized water. The membranes were then rocked at room

temperature in deionized water for 5 minutes, and then in TBST for 5 minutes.

Membranes were then stripped and Western blot analysis was repeated as described

above.

Luciferase Constructs and Assay

Notch-4 luciferase constructs were generously provided by Dr. Emery Bresnick

(University of Wisconsin at Madison, Madison, WI, USA) and previously described (Wu

et al., 2005). Notch-1 luciferase constructs were generously provided by Dr. Paolo Dotto

(Massachusetts General Hospital: Cutaneous Biology Research Center, Charlestown, MA,

USA). The AP-1 luciferase construct was generously provided by Dr. Richard Schultz

(Loyola University Medical Center, Maywood, IL, USA). MDA-MB-231 (1x105) cells

were plated in a 6-well tissue culture treated dish. Twenty-four hours later, the cells were

co-transfected in each well with appropriate reagents: 7 μL of lipofectamine 2000 with

plasmid addition 4 μg of Firefly luciferase (AP-1 or Notch-4 wildtype or mutant

reporters) and 0.4 μg pTL-TK (Renilla luciferase and internal control), and/or 5 μL (16

nM) of PEA3 siRNA reagent. The cells were incubated at 37°C with 95% O2 and 5%

CO2. Forty-eight hours later media was removed from the cells, they were washed with

Page 81: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

60

ice cold PBS, and harvested in 1X passive lysis (300 μL) for 30 minutes while rocking at

room temperature. The plates were then frozen at -80°C for 1 hour to over overnight.

The plates were then removed from the freezer and rocked at room temperature for 15

minutes. Ten microliters of each sample was added to a white coated luminescent 96-

well plate and subjected to the manufactured dual-luciferase assay (Promega, Madison,

WI, USA). Luminescence was measured using the Veritas Microplate Luminometer

(Turner Biosystems, Sunnyvale, CA, USA). The range of luminescence was between

350-650 nm; Firefly luciferase read at 560 nm and pTL-TK at 480 nm. The graphs

represent the ratio of Firefly luciferase to Renilla luciferase (560 nm / 480 nm).

Chromatin Immunoprecipitation (ChIP)

MDA-MB-231 cells (3x106) were plated in 150 cm2 petri dishes for 24 hours.

The cells were transfected with 3 μg of pcDNA3.1, 3 μg of pcDNA3.1-PEA3, and 3 μg

of pcDNA3.1-PEA3 and 5 μL of 10 μM solution of PEA3 siRNA for 48 hours at 37°C

with 95% O2 and 5% CO2. The total volume in the wells was 3 mL. Media was

aspirated and the cells washed once with ice-cold PBS. The cells were cross-linked with

1% formaldehyde in fresh media and incubated at 37°C with 95% O2 and 5% CO2 for 10

minutes. A final concentration of 0.125 M glycine in the 150 cm2 petri dishes was added

while rocking at room temperature for 5 minutes to stop the reaction. The media was

aspirated while on ice, and washed twice with ice-cold PBS. Cells were then scraped,

centrifuged at 2000 rpm at 4°C, and lysed in 500 μL SDS lysis buffer (1% SDS, 10 mM

EDTA, 50 mM Tris-HCl, pH 8.1) containing a protease inhibitor cocktail tablet (Roche

Page 82: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

61

Diagnostics, Indianapolis, IN). The samples were allowed to incubate on ice for 10

minutes. Following incubation, the lysates were sonicated using the Branson Sonifier

250 at output 4.5, duty cycle 50, and pulsed 10 times. The lysates were then diluted in IP

dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mM EDTA, 16.7 mM Tris-HCl, 167

mM NaCl, pH 8.1) to a achieve a 1 to 10 dilution ratio and separated into two tubes: 1)

IgG mouse isotype control, and 2) specific PEA3 antibody. Lysates were pre-cleared

with 30 μL of protein G-plus beads (sc-2002, Santa Cruz) for 1 hour at 4°C with agitation.

Supernatants were isolated and 4 μg of PEA3 (sc-113X) antibody or mouse IgG isotype

control were added and incubated at 4°C overnight with agitation. Fifty microliters of

protein G-plus beads (sc-2002, Santa Cruz) were added to the immune complexes for 2

hours while gently rocking. Immune complexes/beads were washed in low salt buffer

(0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl, 150 mM NaCl, pH 8.1),

high salt buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl, 500 mM

NaCl, pH 8.1), LiCl buffer (1% sodium deoxycholate, 1% NP-40, 0.25 M LiCl, 1 mM

EDTA, 10 mM Tris-HCl, pH 8.1), and TE buffer (1 mM EDTA, 10 mM Tris-HCl, pH

8.0). The protein/antibody complexes from beads were eluted in freshly prepared elution

buffer (1% SDS, 0.1 M sodium bicarbonate, pH 8.0). Protein/DNA was reversed

crosslinked by heating at 65°C overnight while gentle rocking. The protein was degraded

using a proteinase solution (0.5 M EDTA, 1 M Tris-HCl, 10 mg/mL proteinase K, pH

8.0) and incubated at 52°C for 1 hour. DNA was isolated using the Qiagen PCR

purification kit (Qiagen, Valencia, CA, USA ). 20 μL of total lysate was set as an input

control. Enrichment at promoter sites was detected quantitative PCR using iTaq™

Page 83: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

62

SYBR® Green Supermix with ROX (BioRad, Hercules, CA, USA). Ct values were

normalized to the IgG control. Quantitative real-time PCR was carried out as described

under the Real-Time PCR method section. The following primers flanking specific

promoter regions used in detection are shown in Table 5. A schematic representation of

the Notch-1 and Notch-4 promoters is shown in Diagram 10.

Co-Immunoprecipitation (Co-IP)

MDA-MB-231 cells (3x106) were plated in 150 cm2 petri dishes for 24 hours.

The cells were transfected with 3 μg of pcDNA3.1, 3 μg of pcDNA3.1-PEA3, and 3 μg

of pcDNA3.1-PEA3 and 5 μL of 10 uM solution of PEA3 siRNA for 48 hours at 37°C

with 95% O2 and 5% CO2. The total volume in the wells was 3 mL. Media was

aspirated and the cells were washed once with ice-cold PBS. The cells were cross-linked

with 1% formaldehyde in fresh media and incubated at 37°C with 95% O2 and 5% CO2

for 10 minutes. A final concentration of 0.125 M glycine in the 150 cm2 petri dishes was

added while rocking at room temperature for 5 minutes to stop reaction. The media was

aspirated while on ice, and washed twice with ice-cold PBS. Cells were then scraped,

centrifuged at 2000 rpm at 4°C, and lysed in 500 μL SDS lysis buffer (1% SDS, 10 mM

EDTA, 50 mM Tris-HCl, pH 8.1) containing a protease inhibitor cocktail tablet. The

samples were allowed to incubate on ice for 10 minutes. Following incubation, the

lysates were sonicated using the Branson Sonifier 250 at output 4.5, duty cycle 50, and

pulsed 10 times. The lysate concentration was ascertained using the protein BCA protein

assay (Pierce Chemicals, Rockford, IL), incubated for 30 minutes at 37°C with 95% O2

Page 84: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

63

ChIP Primers # FORWARD REVERSE

A) Notch-1 Promoter

Negative Control D 5'-GTGCACACGGCTGTCCG-3' 5'-GCGACAACTGGCGACTGAA-3' 2 x PEA3(+Ctrl) A 5'-GCTGCAAGAGCCAAGATGAA-3' 5'-GGTGCCTGTGTTGAAAGCTCT-3' c-ETS E 5'-CTCCTGGCGCTTAACCAGG-3' 5'-CCAGAAAGCACAAACGGGTC-3' AP-1 (A) B 5'-GCCTCCTTAGCTCACCCTGA-3' 5'-TCTTCAGAGGCCCCCTGC-3' AP-1 (B) C 5'-TCCGCAAACCAGGCTCTG-3' 5'-ATTGGGGTGCAGTGCCG-3'

B) Notch-4 Promoter Negative Control D 5'-AGTGGGAGCAGAGGAGGTG-3' 5'-TGGGTCTGACCACTGAGACA-3' CBF-1 (+Ctrl) A 5'-TGGTACCACCCTGGTCAGTAT-3' 5'-GCTCAGGCATTATGAGCTATG-3' AP-1 (A) B 5'-AGCTGCCACTGACACCTTCT-3' 5'-CAGGGAATGCCAGTCAGAAT-3' AP-1 (B) C 5'-ACTTCCCCAGGGGTTGTC-3' 5'-CTTCCTCCTCGGCCTGCT-3'

Table 5: Chromatin Immunoprecipitation Primers.

NOTCH-4 PROMOTER

NOTCH-1 PROMOTER

ETS ETS ETS ETS ETSAP-1 AP-1NOTCH-1

-3.3 kb -2.9 kb -2.7 kb -1.1 kb

A B C D E

ETS ETS ETS ETS ETSAP-1 AP-1NOTCH-1

-3.3 kb -2.9 kb -2.7 kb -1.1 kb

A B C D E

CBF-1 ETS ETSAP-1 AP-1

A B C D

-3.5 kb -700 bp -70 bp

NOTCH-4

CBF-1 ETS ETSAP-1 AP-1

A B C D

-3.5 kb -700 bp -70 bp

NOTCH-4

Figure 10: Schematic Representation of Notch-1 and Notch-4 Promoters Labeled to Designate Areas of Primer Design.

Page 85: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

64

and 5% CO2, and measured using a 96 plate UV/Visible spectrophotometer. The lysates

were equally diluted in IP dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mM

EDTA, 16.7 mM Tris-HCl, 167 mM NaCl, pH 8.1) to a achieve a 1 to 10 dilution ratio

and separated into two tubes: 1) IgG mouse isotype control, and 2) specific PEA3

antibody. Lysates were pre-cleared with 30 μL of protein G-plus beads (sc-2002, Santa

Cruz) for 1 hour at 4°C with agitation. Precleared lysates were incubated with 3 μg of

PEA3 (sc-113X) or mouse IgG overnight. Thirty microliters of protein G-plus beads (sc-

2002, Santa Cruz) were added to the immune complexes for 2 hours while gently rocking.

Immune complexes/beads were washed three times in phosphate buffered saline (PBS).

The pellet was resuspeneded in Laemmli sample buffer supplemented with beta-

mercaptoethanol (BioRad, Hercules, CA, USA) and heated for 5 minutes at 95°C while

vigorously shaking. Western blot technique was used for detection of co-

immunoprecipitation proteins as described in the Western Blot section of these methods.

Antibodies used for detection were: PEA3 (16), c-FOS (H-125), c-JUN (G-4), and FRA-1

(R-20) from Santa Cruz Biotechnologies, CA.

Cell Cycle Analysis

MDA-MB-231 cells (1x105) were seeded in a 6-well plate. After 24 hours, the

cells were treated/transfected with either DMSO + scrambled siRNA, DMSO + PEA3

siRNA, MRK-003 (10 μM) + scrambled siRNA, or MRK-003 (10 μM) + PEA3 siRNA.

After 24 and 48 hours post-treatment/transfection at 37°C with 95% O2 and 5% CO2, the

cells were trypinized, and the media was isolated. Cell and media were centrifuged at

Page 86: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

65

4°C, 500 rpm for 5 minutes. The cell pellet was resuspended in 2 mL ice-cold PBS.

From the 2 mL single cell suspension, 500 μL of solution was placed into FACS tubes.

Cells were pelleted at 1200 rpm (4°C) for 5 minutes. The supernatant was removed by

inverting and decanting and washed by adding 2 mL of ice cold 5% bovine calf serum

(BCS) in PBS. Cells were again pelleted at 1200 rpm (4°C) for 5 minutes and fixed: cells

were resuspend cells in 100 μL ice cold 5% BCS and 600 μL of ice cold 100% EtOH was

slowly added while gently mixing. The tube was incubated on ice for 30 minutes and

then washed by adding 2 mL of ice cold 5% BCS. Cells were then centrifuged at 1200

rpm (4°C) for 5 minutes and resuspended in 500 μL of 10 μg/mL RNase A. They were

incubated at 37°C for 15 minutes and then allowed to incubate at room temperature for 5

minutes. To the 500 μL volume, 500 μL of 100 μg/mL Propidium Iodide (P4170, Sigma

Aldrich) was added, gently mixed, and incubated at room temperature for at least one

hour in a dark area void of light. The samples were measured by flow cytometry (FACS

Canto, BD Biosciences).

Annexin-V Apoptosis Assay

MDA-MB-231 cells (1.5x105) were seeded in a 6-well plate for 24 hours. Cells

were treated and transfected with either DMSO + scrambled siRNA, DMSO + PEA3

siRNA, MRK-003 (10 μM) + scrambled siRNA, and MRK-003 (10 μM) + PEA3 siRNA.

After 24 and 48 hours post-treatment/transfection at 37°C with 95% O2 and 5% CO2,

cells were trypsinized, washed twice with ice-cold PBS, and resuspended in a 1X

Annexin-V binding buffer (0.1 M HEPES/NaOH, 140 mM NaCl, 25 mM CaCl2, pH 7.4).

Page 87: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

66

From that suspension, 100 μL of the solution was placed into a FACS compatible tube.

To the tube, 5 μL of 1 μg/mL FITC-Annexin-V stain (556420, BD Biosciences) and 5 μL

of 50 μg/mL propidium iodide (P4170, Sigma Aldrich) were added, incubated for 15

minutes at room temperature in the dark. Then, 400 μL of 1X Annexin-V binding buffer

was added back and immediately within the hour the samples were measured by flow

cytometry (FACS Canto, BD Biosciences) using an Annexin-V specific antibody.

Control tubes were used for compensation and background normalization: 1) a tube

containing cell suspension in Annexin-V binding buffer alone, 2) a tube containing cell

suspension in Annexin-V binding buffer with FITC-conjugated Annexin-V antibody, 3) a

tube containing cell suspension in Annexin-V binding buffer with Propidium Iodide, and

4) a tube containing cell suspension in Annexin-V binding buffer, FITC-conjugated

Annexin-V antibody, and Propidium Iodide.

Cell Viability Assay

MDA-MB-231 cells (1x105) were seeded in a 6-well plate. After 24 hours, the

cell were treated and transfected with either DMSO + scrambled siRNA, DMSO + PEA3

siRNA, MRK-003 (10 μM) + scrambled siRNA, and MRK-003 (10 μM) + PEA3 siRNA.

After 24 and 48 hours post-treatment and transfection at 37°C with 95% O2 and 5% CO2,

the cells were trypsinized, and the media was collected. Cell and media were centrifuged

at 4°C, 500 rpm for 5 minutes. The cell pellet was resuspended in 2 mL ice-cold PBS.

From the 2 mL single cell suspension, 500 μL of solution was placed into 1.5 mL

Eppendorf tube. 500 μL of Trypan Blue was added, mixed gently, and incubated at room

Page 88: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

67

temperature for 20 minutes. Then, 20 μL of the solution was place on a heamocytometer.

Stained cells were counted at 40X magnification under a standard light microscope

(Leica DMIL, Leica Microsystems), normalized, and graphed as percent viability as

compared to control.

Colony Formation Assay

A 1.75% solution of Nobel agar was autoclaved, allowed slightly to cool, and

added in equal parts [1:1] to IMEM supplemented with 100 μM nonessential amino acid,

1% L-glutamine (2 mM), and 5% FBS (making a final agar solution of 0.8%). To a 6-

well plate, 1 mL of the agar/media solution was added and allowed to completely solidify.

Once cooled, 2 mL of a 1% methylcellulose (M0512, Sigma Aldrich)/IMEM solution

(supplemented with 5% FBS, 1% nonessential amino acids, and 1% L-glutamine) was

added on top of the agar. The methylcellulose/IMEM solution was made as follows: A

2% methylcellulose solution in serum-free media was made by stirring vigorously for 2-3

hours at room temperature and transferred/stirred at 4°C overnight. Then, 5%FBS, 1%

NEAA, and 1% L-glutamine was added back to the solution, stirred at room temperature

for 30 minutes, and transferred/stirred at 4°C for 1-2 hours depending upon incorporation

rate. Once incorporated completely, the solution was centrifuged at 5500 rpm for 30

minutes at 4°C. The solution was then warmed, to accommodate the transfected/treated

cells to 37°C. MDA-MB-231 (1x103 cells/ml) were treated/transfected with either

DMSO + scrambled siRNA, DMSO + PEA3 siRNA, Mrk-003 (5 μM) + scrambled

siRNA, and Mrk-003 (5 μM) + PEA3 siRNA for 24 hours at 37°C and 5% CO2. Forty-

Page 89: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

68

eight hours later, the transfected/treated cells were trypsinized, counted, and added

directly to the methylcellulose solution and 2 mL of 10,000 cells/well were added atop of

the formerly solidified agar coated plates. The assay was left untouched for 14 days at

37°C and 5% CO2. After 14 days, 20 μL of crystal violet stock (0.2% crystal violet and

2% ethanol diluted in deionized water) was added to each well and gently swirled to

make a final stain concentration around 0.002%. The plate was then incubated for 1 hour

in a 37°C, 5% CO2 incubator. Colonies were photographed (4X magnification) and

counted under a standard light microscope. Nine fields were counted per well and

averaged.

Statistical Analysis

The statistics of these experiments are represented by means plus or minus

standard deviations on graphs and numbers in parenthesis that denote standard deviation.

The statistical significance of three or more independent studies in these experiments was

determined by a two-tailed, unpaired Student’s t-test for two comparisons. Statistical

significance of pairwise comparisons of more than two experiments was discerned using

a one-way ANOVA.

Page 90: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

CHAPTER IV

RESULTS

Even with the advent of modern technologies, breast cancer still remains an area

of extensive research with hopes of uncovering novel strategies to help treat patients.

Previous investigations have determined that overexpression of Notch-1 and Notch-4

play a critical role in breast tumorigenesis and PEA3 overexpression is associated with

aggressive breast cancers particularly the triple-negative subtype. Preliminary research

via promoter scanning using NCBI Entrez Gene bank identified on the Notch-1 and

Notch-4 promoters several putative Ets binding sites, some independent with no known

consensus sequences nearby, or interestingly some Ets sites adjacent to AP-1 consensus

sites. Notch-2 and Notch-3 also have putative Ets binding sites, but preliminary real-time

PCR and Western blot data directed the focus on Notch-1 and Notch-4 for these studies.

Knowing the role of both these aberrant pathways in breast cancer, this investigation aims

at determining a link between the Notch pathway and PEA3 signaling. The following

hypothesis was proposed:

PEA3 is a transcriptional activator of Notch-1 and Notch-4 in MDA-MB-231

triple-negative breast cancer cells. Dual inhibition of PEA3 and Notch

signaling will result in a reduction in cell proliferation and survival implicating

better therapeutic targeting. 69

Page 91: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

70

The following aims were formulated to address the hypothesis in MDA-MB-231 cells:

Specific Aim 1. To determine whether PEA3 is an upstream mediator of the

Notch signaling pathway.

Specific Aim 2. To elucidate whether PEA3 acts a direct positive regulator of

Notch-1 and Notch-4 transcription.

Specific Aim 3. To establish a biological significance by determining whether

dual inhibition of Notch and PEA3 leads to reduced cell proliferation and

tumorigenicity in vitro.

This research strategy provides novel evidence of a link between two pathways that are

overexpressed in breast cancer. PEA3 is a transcriptional activator of both Notch-1 and

Notch-4 mRNA in MDA-MB-231 cells. PEA3-mediated Notch-1 transcription is AP-1

independent while Notch-4 transcription requires both PEA3 and AP-1 (Figure 11).

PEA3 and Notch signaling are essential for proliferation and survival in MDA-MB-231

cells. Furthermore, PEA3 is a transcriptional activator of both Notch-1 and Notch-4

mRNA in other breast cancer cells. Thus, the hypothesis remains that dual targeting of

both PEA3 and Notch pathways might provide a new therapeutic strategy for triple-

negative breast cancer as well as possibly therapeutic targeting in other breast cancer

subtypes where PEA3 regulates Notch-1 and Notch-4.

Page 92: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

Our novel findingsPreviously knownTherapeutic strategy

HYPOTHESIS

PEA3

Notch-1

Notch-4

Migration

HESHEY-1

Cyclin-D1

MMP-9COX-2

IL-8

Proliferation

GSI

GSIsiRNA

AP-1

c-JUNFRA-1

AP-1

c-JUNFRA-1

Malignancy

71

Figure 11: Hypothesis of the Investigation. PEA3 is a novel transcriptional activator of Notch-1 and Notch-4 in basal-like breast cancer. Dual inhibition of PEA3 and Notch signaling will result in a reduction in cell proliferation, tumorigenicity, and malignancy implicating better therapeutic targeting and overall survival.

Page 93: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

72

SPECIFIC AIM-1:

To Determine Whether PEA3 is an Upstream Mediator of the

Notch Signaling Pathway.

MDA-MB-231 cells are a representative cell line of triple-negative breast cancer.

MDA-MB-231 cells lack expression of estrogen receptor, progesterone receptor, or

HER2 overexpression. These cells are the model system used for the subsequent studies.

The investigation began by determining the efficiency of PEA3 knockdown by

RNA interference to determine its resulting effects in vitro. Acquiring SMART-POOL of

three specific PEA3 siRNAs from Santa Cruz Biotechnologies (Santa Cruz, CA), siRNA

transfection in a dose dependent manner was performed to evaluate the optimal

concentration of PEA3 siRNA. Using lipofectamine as the transfecting vehicle, 5 μL of

siRNA (16 nM) was transfected into MDA-MB-231 cells. After 48 hours, cells were

harvested for RNA and protein. Real-time PCR of PEA3 transcripts indicated that 5 μL

of siRNA (16 nM) was sufficient to reduce PEA3 transcripts by ~90% (Figure 12).

Endogenous PEA3 is extremely difficult to detect; the protein is relatively unstable

exhibiting a turnover rate of about 30 minutes via the ubiquitin-proteasome pathway on

the C-terminus domain of PEA3 (Takahashi et al., 2005). It is rare to capture the

endogenous PEA3 protein levels (Baert et al., 1997). A Western blot analysis of

endogenous PEA3 protein levels demonstrated the corresponding effect of PEA3 siRNA

Page 94: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

73

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

PEA3 mRNA

* p<0.0001

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

PEA3 mRNA

* p<0.0001

Figure 12: PEA3 Transcripts Upon RNA Interference. MDA-MB-231 cells were transfected with either scrambled siRNA (SCRBi) or 5 μL of PEA3 siRNA (PEA3i) corresponding to overall siRNA concentrations of 16nM. Relative mRNA levels of PEA3 were measured 48 hours later as a knockdown control using quantitative PCR. Transcripts were normalized to HPRT and means were plotted. Error bars represent standard deviation. Statistical significance (p<0.0001) of three experiments was determined by two-tailed, unpaired Student’s t- test.

Page 95: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

74

PEA3

Actin

SCRBi

PEA3i+ –– +

PEA3

Actin

SCRBi

PEA3i+ –– +

1.00 0.55

Figure 13: Endogenous PEA3 protein upon RNA Interference. MDA-MB-231 cells were transfected with either scrambled siRNA (SCRBi) or 5 μL of PEA3 siRNA (PEA3i) corresponding to overall siRNA concentrations of 16nM. Protein levels of PEA3 were detected by Western blot analysis after 48 hours as described in the materials and methods. 50 μg of total protein lysate in each lane was subjected to SDS-PAGE 4-12% gel. Beta-actin was detected as an endogenous control.

Page 96: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

75

on its protein levels (reduced by 50% upon siRNA transfection as measured by

densitometry) (Figure 13).

1. PEA3 Regulates Notch-1 and Notch-4 Transcripts.

To investigate the role of PEA3 on Notch expression, the effect of PEA3 RNA

interference on the expression of all four Notch receptor (1,2,3,4) mRNAs was examined.

Cells were transfected with either scrambled control siRNA or PEA3 siRNA for 48 hours.

As measured by real-time PCR, Notch-1 and Notch-4 transcripts were decreased by 50%

and 70%, respectively upon PEA3 knockdown. Notch-3 transcript levels were

unchanged. Interestingly, Notch-2 levels showed a moderate but significant increase

upon PEA3 knockdown, which upon further investigation may prove to be advantageous

since it has been correlated with pro-apoptotic and breast tumor suppressive function

[(O'Neill et al., 2007) and discussed in Chapter III] (Figure 14).

2. PEA3 Regulates Notch-1 Protein Expression.

To determine if the effect on Notch-1 and Notch-4 transcripts by PEA3 correlated

with protein expression, we measured steady protein levels using Western blotting in

cells transfected with either control (SCRBi) or PEA3 siRNA for 48 hours. The results

showed a reduction of Notch-1 full length (FL) and transmembrane (TM) receptor protein

levels by 40% and 50%, respectively (Figure 15) agreeing with its corresponding

transcripts. In contrast to Notch-1, PEA3 siRNA had no noticeable effects on steady-

state Notch-4 IC protein levels (Figure 5, lanes 1 and 2). However, it is known that the

Page 97: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

NOTCH-1

0.00.20.40.60.81.01.21.41.61.82.0

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

*p<0.001

NOTCH-1

0.00.20.40.60.81.01.21.41.61.82.0

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

*p<0.001

SCRBi PEA3i0.00.20.40.60.81.01.21.41.61.82.0

NOTCH-4R

elat

ive

mR

NA

Lev

els

*p<0.001

SCRBi PEA3i0.00.20.40.60.81.01.21.41.61.82.0

NOTCH-4R

elat

ive

mR

NA

Lev

els

*p<0.001

NOTCH-2

0.00.20.4

0.60.81.01.21.41.61.82.0

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

* p<0.05

NOTCH-2

0.00.20.4

0.60.81.01.21.41.61.82.0

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

NOTCH-2

0.00.20.4

0.60.81.01.21.41.61.82.0

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

* p<0.05

SCRBi PEA3i0.00.2

0.40.60.81.01.21.41.61.8

2.0NOTCH-3

Rel

ativ

e m

RN

A L

evel

s

SCRBi PEA3i0.00.2

0.40.60.81.01.21.41.61.8

2.0NOTCH-3

Rel

ativ

e m

RN

A L

evel

s

SCRBi PEA3i0.00.2

0.40.60.81.01.21.41.61.8

2.0NOTCH-3

Rel

ativ

e m

RN

A L

evel

s

76

Figure 14: Effects of PEA3 siRNA on Notch Transcripts. MDA-MB-231 cells were transfected with either scrambled siRNA (SCRBi) or PEA3 siRNA (PEA3i) for 48 hours. Relative mRNA levels of Notch receptors (NOTCH-1,2,3,4) were measured by quantitative PCR. Transcripts were normalized to HPRT and means were plotted. Error bars represent standard deviation. Statistical significance of three or more experiments was determined by two-tailed, unpaired Student’s t- test.

Page 98: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

77

NOTCH-1

β-ACTIN

FL

TM

SCRBi + –– +PEA3i

NOTCH-1

β-ACTIN

FL

TM

SCRBi + –– +PEA3i

0.0

0.2

0.4

0.6

0.8

1.0

1.2

p<0.01

*

0.0

0.2

0.4

0.6

0.8

1.0

1.2

*

p<0.001

FULL LENGTHNOTCH-1 (FL)

TRANSMEMBRANEPORTION NOTCH-1 (TM)

SCRBi PEA3i SCRBi PEA3i0.0

0.2

0.4

0.6

0.8

1.0

1.2

p<0.01

*

0.0

0.2

0.4

0.6

0.8

1.0

1.2

*

p<0.001

FULL LENGTHNOTCH-1 (FL)

TRANSMEMBRANEPORTION NOTCH-1 (TM)

SCRBi PEA3i SCRBi PEA3i

Den

sito

met

ry (N

orm

aliz

ed to

Act

in)

Den

sito

met

ry (N

orm

aliz

ed to

Act

in)

Figure 15: Protein Levels of Notch-1 Upon PEA3 RNA Interference. MDA-MB-231 cells were transfected with either scrambled siRNA (SCRBi) or 5 μL (16nM) of PEA3 siRNA (PEA3i). Protein levels of Notch-1 were detected by Western blot analysis after 48 hours as described in the materials and methods. 10 μg of total protein lysate in each lane was subjected to SDS-PAGE 10% gel. Both transmembrane portion (TM) and full length portion (FL) of Notch-1 were detected by the antibody. Beta-actin was detected as an endogenous control. Graphs represent quantification of changes in full length and transmembrane length Notch upon transfection with either scrambled or PEA3 siRNA as measured by densitometry. Statistical significance of three or more experiments was determined by two-tailed, unpaired Student’s t-test.

Page 99: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

78

SCRBi + – + –– + – +– – + +

β-ACTIN

NOTCH-4

PEA3i

LACTACYSTIN

SCRBi + – + –– + – +– – + +

β-ACTIN

NOTCH-4

PEA3i

LACTACYSTIN

Figure 16: Protein Levels of Notch-4 Upon PEA3 RNA Interference. MDA-MB-231 cells were transfected with either scrambled siRNA (SCRBi) or 5 μL (16nM) of PEA3 siRNA (PEA3i). Protein levels of Notch-4 were detected by Western blot analysis after 48 hours as described in the materials and methods. Cells were treated with or without 7 μM lactacystin after 24 hours of transfection for an additional 18 hours. 50 μg of total protein lysate in each lane was subjected to SDS-PAGE 4-12% gel. Beta-actin was detected as an endogenous control. Western Blot is representative of more than three experiments.

Page 100: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

79

Notch-4 receptor is sensitive to stress and has a rapid turnover rate with a half life of 1.25

hours which is mediated by the proteosome (Wu et al., 2001). Thus, to determine if

protein stability is critical for regulation of Notch-4 protein by PEA3, a proteasome

inhibitor (lacacystin) was used to treat the cells prior to transfection with control or PEA3

siRNA. The protein levels of Notch-4 IC were increased almost two fold (Figure 16, lane

3) with lacacystin treatment and decreased by almost 90% upon PEA3 knockdown

(Figure 16, lane 4). This result suggests that Notch-4IC protein expression is critically

regulated by protein stability and not necessarily at the transcriptional level. Since PEA3

siRNA demonstrated little effects on Notch-2 and Notch-3 transcripts, their

corresponding protein levels by Western blot analysis were not pursued.

3. Classic Gene Targets of PEA3 and Notch are Decreased by PEA3

Knockdown.

To confirm the effect of PEA3 siRNA on the activity of PEA3, its classical

downstream targets were measured by real-time PCR after transfection with control or

PEA3 siRNA for 48 hours. PEA3 target genes, namely IL-8 and MMP-9, were

significantly reduced by 80% and 60%, respectively (Figure 17). To address whether the

Notch signaling pathway was affected by PEA3 RNA interference, a classic downstream

target of the Notch, HEY-1, was also significantly reduced upon PEA3 knockdown by

40% (Figure 6). Taken together, these results suggest that PEA3 positively regulates

Notch-1 and Notch-4 receptor transcripts, Notch-1 protein and, Notch signaling in MDA-

MB-231 cells.

Page 101: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

80

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

HEY-1

Rel

ativ

e m

RN

A L

evel

s

* p<0.05

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

HEY-1

Rel

ativ

e m

RN

A L

evel

s

*

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

HEY-1

Rel

ativ

e m

RN

A L

evel

s

* p<0.05

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

MMP-9*

Rel

ativ

e m

RN

A L

evel

s

p<0.01

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

MMP-9*

Rel

ativ

e m

RN

A L

evel

s

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

MMP-9*

Rel

ativ

e m

RN

A L

evel

s

p<0.01

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

IL-8*

Rel

ativ

e m

RN

A L

evel

sp<0.001

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

IL-8*

Rel

ativ

e m

RN

A L

evel

s

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

IL-8*

Rel

ativ

e m

RN

A L

evel

sp<0.001

Figure 17: Transcript Levels of Classic PEA3 and Notch Targets Upon PEA3 siRNA. MDA-MB-231 cells were transfected with either scrambled siRNA (SCRBi) or PEA3 siRNA (PEA3i) for 48 hours. Downstream targets of PEA3 (IL-8, MMP-9) and Notch (HEY-1) were measured by quantitative PCR. Transcripts were normalized to HPRT and means were plotted. Error bars represent standard deviation. Statistical significance of three or more experiments was determined by two-tailed, unpaired Student’s t-test.

Page 102: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

81

SPECIFIC AIM-2:

To Elucidate Whether PEA3 Acts as a Direct Positive Regulator of

Notch-1 and Notch-4 Transcription.

To determine the mechanism by which PEA3 regulates Notch-1 and Notch-4

transcription, the promoter regions of Notch-1 and Notch-4 were scanned using NCBI

Entrez Gene bank and identified several putative Ets binding sites (Figure 18).

Interestingly both promoters contained two putative AP-1 sites adjacent to Ets sites. A

classic CBF-1 site was also previously identified in the Notch-4 promoter and used as a

control. Primers were subsequently designed to include these identified regions. Primers

flanking promoter regions containing no known consensus sites were used as negative

controls (Figure 18). Notch-2 and Notch-3 also contain minimal putative Ets sites. The

quantitative PCR results on these promoters indicated mild changes in transcript levels

(Figure 14) and therefore were not pursued. However, future studies on these sites may

prove to be significant and/or may be used as negative controls.

Emphasizing the notion that PEA3 is highly unstable, endogenous protein levels

are difficult to detect (Baert et al., 1997). Consequently, exogenous expression of PEA3

was required to perform subsequent chromatin immunoprecipitations (ChIPs). Empty

vector (pcDNA3.1) and PEA3 expression vector (pcDNA3.1-PEA3) were tested in

MDA-MB-231 cells in a dose dependent manner to determine an optimal concentration.

Upon transfection of pcDNA3.1-PEA3 at concentrations of 0.5 μg, 1.0 μg, 2.0 μg, and

Page 103: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

82 NOTCH-4 PROMOTER

NOTCH-1 PROMOTER

ETS ETS ETS ETS ETSAP-1 AP-1NOTCH-1

-3.3 kb -2.9 kb -2.7 kb -1.1 kb

A B C D E

ETS ETS ETS ETS ETSAP-1 AP-1NOTCH-1

-3.3 kb -2.9 kb -2.7 kb -1.1 kb

A B C D E

CBF-1 ETS ETSAP-1 AP-1

A B C D

-3.5 kb -700 bp -70 bp

NOTCH-4

CBF-1 ETS ETSAP-1 AP-1

A B C D

-3.5 kb -700 bp -70 bp

NOTCH-4

Figure 18: Schematic Representation of Notch-1 and Notch-4 Promoters.

Page 104: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

83

4.0 μg, transcript levels of PEA3 increased in a dose dependent manner (Figure 19).

Moreover, a classic target gene of PEA3, COX-2, was measured and demonstrated a

similar dose dependent increase (Figure 19). Discerning that the maximal effect was at a

concentration of 4 μg, 4 μg concentration of pcDNA3.1-PEA3 and pcDNA3.1 was used

in the subsequent ChIP analyses.

To determine if exogenous PEA3 expression was compatible with

immunoprecipitation, 4 μg of pcDNA3.1 and pcDNA3.1-PEA3 vectors were transfected

in MDA-MB-231 cells for 48 hours prior to being subjected to immunoprecipitation.

Endogenous levels of PEA3 were not detected by immunoprecipitation, however

exogenous expression successfully immunoprecipitated PEA3 protein (Figure 20).

1. PEA3 is Enriched on the Notch-1 and Notch-4 Promoters.

To address whether PEA3 is recruited to the Notch-1 and Notch-4 promoters,

chromatin immunoprecipitation experiments were performed on lysates from MDA-MB-

231 cells that were transfected with PEA3 expression plasmid (pcDNA3.1-PEA3).

Comparable to the immunoprecipitation studies, ChIP analysis on endogenous levels of

PEA3 was not measurable (Figure 21) most likely a result of PEA3 protein instability,

which it is rapidly turned over and undetectable (Baert et al., 1997).

Five Ets sites on the Notch-1 promoter and four on the Notch-4 promoter were

identified and pursued via chromatin immunoprecipitation of PEA3. As a result of ChIP

analysis, PEA3 enrichment was found on both Notch-1 (Figure 22) and Notch-4 (Figure

23) promoter regions within 1.1 kilobase or -70 nuclotides, respectively, upsteam of the

Page 105: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

84

0

10

20

30

40

50

60

70

80

pcDNA3.1 0.5ug 1.0ug 2.0ug 4.0ug

pcDNA3.1-PEA3

Rel

ativ

e m

RN

A L

evel

s

PEA3

0

10

20

30

40

50

60

70

80

pcDNA3.1 0.5ug 1.0ug 2.0ug 4.0ug

pcDNA3.1-PEA3

Rel

ativ

e m

RN

A L

evel

s

PEA3

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

5.0

pcDNA3.1 0.5ug 1.0ug 2.0ug 4.0ug

pcDNA3.1-PEA3

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

5.0

pcDNA3.1 0.5ug 1.0ug 2.0ug 4.0ug

pcDNA3.1-PEA3

COX-2

Rel

ativ

e m

RN

A L

evel

s

Figure 19: Exogenous Expression of PEA3 and Its Effects on Target Genes. MDA-MB-231 cells were transfected with either vector control plasmid (pcDNA3.1) or PEA3 expression vector (pcDNA3.1-PEA3) in a dose dependent manner for 48 hours. Relative mRNA levels of PEA3 and a classic target gene (COX-2) were measured by quantitative PCR. Transcripts were normalized to HPRT and means were plotted.

Page 106: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

85

Heavy IgG

Light IgG

pcDNA3.1 pcDNA3.1-PEA3

IgG PEA3 IgG PEA3

PEA3

Antibodies:

Vectors: I.P.

Conjugated

Heavy IgG

Light IgG

pcDNA3.1 pcDNA3.1-PEA3

IgG PEA3 IgG PEA3

PEA3

Antibodies:

Vectors: I.P.

Heavy IgG

Light IgG

pcDNA3.1 pcDNA3.1-PEA3

IgG PEA3 IgG PEA3

PEA3

Antibodies:

Vectors: I.P.

Conjugated

Figure 20: Immunoprecipiation of Endogenous and Exogenous PEA3. MDA-MB-231 cells were transfected with either pcDNA3.1 or pcDNA3.1-PEA3 vectors. Immunoprecipitation of PEA3 and its isotype IgG control were performed after 48 hours as described in the materials and methods. Total immunoprecipitated protein lysates were subjected to SDS-PAGE 4-12% gel. Heavy and light chain IgG were detected as an endogenous loading control.

Page 107: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

86

0.0

0.5

1.0

1.5

2.0

2.5

3.0

Rel

ativ

e Fo

ld In

duct

ion

* p<0.0001

IgG PEA3

Vector Alone

PEA3 over-expression

NOTCH-4

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

Rel

ativ

e Fo

ld In

duct

ion

p<0.0001*

Vector Alone

PEA3 over-expression

IgG PEA3

NOTCH-1

0.0

0.5

1.0

1.5

2.0

2.5

3.0

0.0

0.5

1.0

1.5

2.0

2.5

3.0

Rel

ativ

e Fo

ld In

duct

ion

* p<0.0001

IgG PEA3

Vector Alone

PEA3 over-expression

NOTCH-4

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

Rel

ativ

e Fo

ld In

duct

ion

p<0.0001*

Vector Alone

PEA3 over-expression

IgG PEA3

NOTCH-1

Figure 21: Chromatin Immunoprecipitation of Endogenous and Exogenous PEA3 on the Notch-1 and Notch-4 Promoter.

MDA-MB-231 cells were transfected with either pcDNA3.1 (Vector alone) or PEA3-pcDNA3.1 overexpression plasmid. Lysates were subjected to chromatin immunoprecipitation using either PEA3 antibody or isotype control IgG on the Notch-1 and Notch-4 promoter. PEA3 enrichment was measured by quantitative PCR and normalized to IgG control. Below graphs is a schematic representation of the Notch-1 and Notch-4 promoters containing the identified enriched Ets sites. Means and standard deviation of more than three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 108: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

IgG PEA3 IgG PEA3

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

Rel

ativ

e Fo

ld In

duct

ion

*

A B C D E

NOTCH-1 PROMOTER

p<0.0001

87

Figure 22: Chromatin Immunoprecipitation of PEA3 on the Notch-1 Promoter. MDA-MB-231 cells were transfected with PEA3 expression plasmid (pcDNA3.1). Lysates were subjected to chromatin immunoprecipitation using either PEA3 antibody or isotype control IgG on the Notch-1. PEA3 enrichment was measured by quantitative PCR and normalized to IgG control. Below graphs is a schematic representation of the Notch-1 promoter containing multiple AP-1 consensus sites and Ets sites. Means and standard deviation of more than three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 109: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

0.0

0.5

1.0

1.5

2.0

2.5

3.0

0.0

0.5

1.0

1.5

2.0

2.5

3.0

Rel

ativ

e Fo

ld In

duct

ion

*IgG PEA3

A B C D

NOTCH-4 PROMOTER

p<0.0001

88

Figure 23: Chromatin Immunoprecipitation of PEA3 on the Notch-4 Promoter. MDA-MB-231 cells were transfected with PEA3 expression plasmid (pcDNA3.1). Lysates were subjected to chromatin immunoprecipitation using either PEA3 antibody or isotype control IgG on the Notch-4 promoter. PEA3 enrichment was measured by quantitative PCR and normalized to IgG control. Below graphs is a schematic representation of the Notch-4 promoters containing multiple AP-1 consensus sites and Ets sites. Means and standard deviation of more than three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 110: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

89

transcriptional start site. The other identified sites via promoter scanning showed no

measurable PEA3 enrichment (Figure 22 and Figure 23).

To address the specificity of the PEA3 antibody to detect recruitment of PEA3 to

these promoter regions, a SMART-POOL of three specific PEA3 siRNAs was used.

Western blot analysis was performed to determine the efficiency of PEA3 siRNA on

exogenous PEA3 expression (Figure 24). As a result from ChIP analysis, PEA3

enrichment on the -1.1 kb site on the Notch-1 promoter and the -70 bp site on the Notch-4

promoter was abrogated upon specific PEA3 knockdown (Figure 25).

2. PEA3 and AP-1 Regulate the Notch-4 Promoter.

Previous studies in vascular endothelial cells performed by Wu et al. at the

University of Wisconsin at Madison indicated the importance of AP-1 on the regulation

of Notch-4 (Wu et al., 2005). To assess if the -70 nucleotide region upstream of the start

site was indeed necessary in MDA-MB-231 cells for the regulation of the Notch-4

promoter by PEA3 as identified by ChIP and potentially AP-1, a wild type AP-1 or

mutant AP-1-containing Notch-4 promoter luciferase reporter constructs were obtained

from the Dr. Emery Bresnick’s laboratory and previously described (Wu et al., 2005).

The wild type construct contained an intact AP-1 and Ets consensus sites at -70

nucleotides. The mutant AP-1 construct contained an ablated AP-1 consensus site using

site-directed mutagenesis; the adjacent Ets binding site was left preserved (Wu et al.,

2005) (Figure 26).

Page 111: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

90

pcDNA3PEA3 vector

Scrambled siRNAPEA3 siRNA

PEA3

+ – –– + + – + –– – +

Actin

pcDNA3PEA3 vector

Scrambled siRNAPEA3 siRNA

PEA3

+ – –– + + – + –– – +

Actin

W.Blot

Figure 24: Western Blot Analysis of PEA3 Expression.

MDA-MB-231 cells were transfected with either pcDNA3.1 and scrambled siRNA (SCRBi), pcDNA3.1-PEA3, PEA3 siRNA, or a combination of pcDNA3.1-PEA3 and PEA3 siRNA (PEA3i). Protein levels of PEA3 were detected by Western blot analysis after 48 hours as described in the materials and methods. Total protein lysate in each lane was subjected to SDS-PAGE 4-12% gel. Beta-actin was detected as an endogenous control.

Page 112: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

91

0.0

0.5

1.0

1.5

2.0

2.5

3.0

NOTCH-4

AP-1

*

PEA3

PEA3 siRNAPEA3

PEA3 siRNA

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

NOTCH-1

c-ETS

*

PEA3-pcDNA3.1PEA3-pcDNA3.1

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

0.0

0.5

1.0

1.5

2.0

2.5

3.0

NOTCH-4

AP-1

*

PEA3

PEA3 siRNAPEA3

PEA3 siRNA

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

NOTCH-1

c-ETS

*

PEA3-pcDNA3.1PEA3-pcDNA3.1

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

p<0.001 p<0.001

Figure 25: PEA3 Enrichment Specificity on the Notch-1 and Notch-4 Promoters as Measured by Chromatin Immunoprecipitation.

MDA-MB-231 cells were co-transfected with PEA3 overexpression plasmid (pcDNA3.1) and either scrambled siRNA (SCRBi) or PEA3 siRNA (PEA3i) for 48 hours. Lysates then were subjected to chromatin immunoprecipitation using either PEA3 antibody or isotype control IgG on the Notch-1 and Notch-4 promoter. PEA3 enrichment was measured by quantitative PCR and normalized to IgG control. Below graphs is a schematic representation of the Notch-1 and Notch-4 promoters containing the identified enriched Ets sites. Means and standard deviation of more than three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 113: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

92

TTGTGTGACTCAGGAAACAGCTCAGACGTG

TTGTGGCTAGACGGAAACAGCTCAGACGTGAP-1 ETS

AP-1 (wt)

AP-1 (mut)

LUCIFERASEETSAP-1~650 bp -70 bp

TTGTGTGACTCAGGAAACAGCTCAGACGTG

TTGTGGCTAGACGGAAACAGCTCAGACGTGAP-1 ETS

AP-1 (wt)

AP-1 (mut)

LUCIFERASEETSAP-1~650 bp -70 bp

Figure 26: Wildtype and Mutant Notch-4 Promoter Luciferase Constructs

Abbreviations: AP-1 consensus site construct, wt; Site-directed AP-1 consensus site mutant construct, mut;

Page 114: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

93

The wildtype or mutant Notch-4 promoter Luciferase constructs were co-

transfected with either scrambled or PEA3 siRNA in our in vitro triple-negative MDA-

MB-231 model system. PEA3 siRNA significantly decreased wild type AP-1-containing

or mutant AP-1-containing reporter activity compared to the scrambled control (Figure

27). These results suggest that either PEA3 or AP-1 is critical for the regulation of the -

70 region within the Notch-4 promoter in MDA-MB-231 cells. The mutant ablated AP-1

Luciferase results suggest that PEA3 may play an independent role from AP-1. However,

the construct does contain an additional, but minimal Ets site upstream of the AP-1:Ets

site, which may account for the further decrease in Luciferase on the mutant AP-1

construct. As a control study, we observed no significant difference in AP-1 luciferase

reporter activity upon PEA3 knockdown indicating that PEA3 is not mediating effects on

the Notch-4 promoter through AP-1 regulation (Figure 28).

To address if PEA3 acts directly with AP-1 on Notch-1 and Notch-4 promoter

regions, since it is known to work in accordance with c-JUN on several other genes

(referenced in Chapter III), transfected lysates with either pHMB empty vector or pHMB-

TAM-67, a dominant negative form of c-JUN missing the transactivation domain (Figure

8) (Liu et al., 2002), were subjected to chromatin immunoprecipitation. PEA3

enrichment on the Notch-1 promoter was not affected by TAM-67 (Figure 29). However,

PEA3 recruitment to an Ets site adjacent to an AP-1 site approximately -70 nucleotides

upstream of the start site of the Notch-4 promoter was abrogated by expression of TAM-

67 (Figure 29) indicating that the transactivation domain of c-JUN is required for PEA3

recruitment. As a control, dual Firefly-Renilla luciferase assays using the AP-1 reporter

Page 115: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

TTGTG

Figure 27: Luciferase Assay of Wildtype and AP-1 Mutant Notch-4 Promoter.

Above is a schematic representation of the Notch-4 luciferase construct indicating mutated AP-1 consensus site and Ets site. MDA-MB-231 cells were co-transfected with either wildtype AP-1 or mutated AP-1 Notch-4 luciferase and either scrambled or PEA3 siRNA. Firefly luciferase (pGL2) was measured normalized to Renilla as an internal transfection control. Means and standard deviation of three or more experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

0.0

0.2

0.4

0.6

0.8

1.0

SCRBi PEA3i SCRBi PEA3i

FIR

EFLY

/ R

ENIL

LA 0.0

0.2

0.4

0.6

0.8

1.0

*

SCRBi PEA3i

FIR

EFLY

/ R

ENIL

LA

Notch-4 (Wt) Luciferase Notch-4 (Mut) Luciferase

*p<0.001

p<0.01

0.0

0.2

0.4

0.6

0.8

1.0

SCRBi PEA3i SCRBi PEA3i

FIR

EFLY

/ R

ENIL

LA 0.0

0.2

0.4

0.6

0.8

1.0

*

SCRBi PEA3i

FIR

EFLY

/ R

ENIL

LA

Notch-4 (Wt) Luciferase Notch-4 (Mut) Luciferase

*p<0.001

p<0.01

TGACTCA

GCTAGAC

GGAAACAGCTCAGACGTG

TTGTG GGAAACAGCTCAGACGTGAP-1 ETS

AP-1 (wt)

AP-1 (mut)

LUCIFERASEETSAP-1~650 bp -70 bp

ETSAP-1~650 bp -70 bp

LUCIFERASE

TTGTG GGAAACAGCTCAGACGTG

TTGTG GGAAACAGCTCAGACGTG

TGACTCA

GCTAGAC

94

AP-1 ETSAP-1 (wt)

AP-1 (mut)

Page 116: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

95

LUCIFERASEAP-1 AP-1 AP-1 AP-1

TGAGTCA TGAGTCA TGAGTCA TGAGTCA

LUCIFERASEAP-1 AP-1 AP-1 AP-1

TGAGTCA TGAGTCA TGAGTCA TGAGTCA

2468

101214

SCRBi PEA3i0

FIR

EFLY

/ R

ENIL

LA

2468

101214

SCRBi PEA3i0

FIR

EFLY

/ R

ENIL

LA

Figure 28: PEA3 Does Not Regulate AP-1 Activity.

Above is a schematic representation of the AP-1 luciferase construct. MDA-MB-231 cells were co-transfected with an AP-1 luciferase and either scrambled or PEA3 siRNA. Firefly luciferase (pGL2) was measured normalized to Renilla as an internal transfection control. Means and standard deviation of three or more experiments were plotted.

Page 117: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

96

c-ETS

NOTCH-1

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

PEA3

PEA3 + TAM-67

0.0

0.5

1.0

1.5

2.0

2.5

AP-1

NOTCH-4

* p<0.01

PEA3

PEA3 +TAM-67

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

c-ETS

NOTCH-1

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

PEA3

PEA3 + TAM-67

0.0

0.5

1.0

1.5

2.0

2.5

0.0

0.5

1.0

1.5

2.0

2.5

AP-1

NOTCH-4

* p<0.01

PEA3

PEA3 +TAM-67

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Figure 29: Dominant Negative c-JUN Ablates PEA3 Recruitment on Notch-4, And has No Effect on PEA3 Recruitment on Notch-1.

MDA-MB-231 cells were co-transfected with PEA3 expression plasmid (pcDNA3.1) and with either pHMB-vector or TAM-67. Lysates were subjected to chromatin immunoprecipitation using either PEA3 antibody or isotype control IgG on the Notch-1 and 4 promoters, respectively. PEA3 enrichment was measured by quantitative PCR and normalized to IgG control. Means and standard deviation of more than three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 118: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

97

construct containing four tandem AP-1 consensus sites (Figure 28) were performed to

determine the efficiency of TAM-67 on AP-1 activity. Upon co-transfection with TAM-

67 and AP-1 luciferase construct, AP-1 activity was diminished by 50% (Figure 30).

To determine if the c-JUN protein specifically is required for PEA3 enrichment

on the identified Notch-4 promoter region, transfected lysates with either scrambled or a

smart-pool of specific c-JUN siRNAs were subjected to chromatin immunoprecipitation.

In agreement, PEA3 enrichment on the Notch-4 promoter region was abrogated upon c-

JUN knockdown (Figure 31, top) suggesting that c-JUN is required for PEA3 enrichment

on the identified Notch-4 promoter region. As a control, c-JUN siRNA was transfected

in MDA-MB-231 cells and subjected to quantitative real-time PCR. As a result, c-JUN

siRNA reduced endogenous c-JUN transcripts by 45% (Figure 31, bottom). Chromatin

immunoprecipitation of c-JUN on Notch-1 and Notch-4 promoters was performed, but

resulted in inconclusive findings. Other investigators have completed successful ChIP(s)

of c-JUN (Zhao et al., 2010), which in our system will require optimization and increased

sensitivity.

To determine whether PEA3 is in a complex with c-JUN since c-JUN is required

for PEA3 enrichment as observed by the previous chromatin immunoprecipitation

experiments, PEA3 was immunoprecipitated and Western blotting was performed to

detect PEA3 and c-JUN proteins. Co-immunoprecipitation demonstrated that PEA3 was

in complex with c-JUN (Figure 32). Taken together, these results strongly indicate that

PEA3 and c-JUN are required to regulate the Notch-4 promoter.

Page 119: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

98

H N3 COOH

TAD DBD ZIP

H N3 COOH

DBD ZIP

c-JUN

TAM-67

TTSS

63 73 91 93H N3 COOH

TAD DBD ZIP

H N3 COOHH N3H N3 COOH

TAD DBD ZIP

H N3 COOH

DBD ZIP

H N3 COOHH N3H N3 COOH

DBD ZIP

c-JUN

TAM-67

TTSS

63 73 91 93

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

1.8

phMB TAM-67

Fire

fly L

ucife

rase

/ R

enilla

AP-1 LuciferaseTAM-67

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

1.8

phMB TAM-67

Fire

fly L

ucife

rase

/ R

enilla

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

1.8

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

1.8

phMB TAM-67

Fire

fly L

ucife

rase

/ R

enilla

AP-1 LuciferaseTAM-67

p<0.05*

Figure 30: Dominant Negative c-JUN (TAM-67) Reduces AP-1 Activity.

Above is a schematic representation of the wildtype and dominant negative c-JUN construct. MDA-MB-231 cells were co-transfected with AP-1 luciferase and either phMB-vector or TAM-67. Firefly luciferase (pGL2) was measured normalized to Renilla as an internal transfection control. Means and standard deviation of three or more experiments were plotted.

Page 120: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

99

PEA3

PEA3 +c-JUNi

AP-1

* p<0.01

NOTCH-4

0.00.5

1.0

1.5

2.0

2.5

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

PEA3

PEA3 +c-JUNi

AP-1

* p<0.01

NOTCH-4

0.00.5

1.0

1.5

2.0

2.5

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

PEA3

PEA3 +c-JUNi

AP-1

* p<0.01

NOTCH-4

0.00.5

1.0

1.5

2.0

2.5

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi c-JUNi

Rel

ativ

e m

RN

A L

evel

s

c-JUN

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi c-JUNi

Rel

ativ

e m

RN

A L

evel

s

c-JUN

p<0.01*

Figure 31: c-JUN is Required for PEA3 Enrichment on the Notch-4 Promoter. MDA-MB-231 cells were co-transfected with PEA3 expression plasmid (pcDNA3.1) and with either scrambled or c-JUN siRNA. Lysates were subjected to chromatin immunoprecipitation using either PEA3 antibody or isotype control IgG on the Notch-4 promoter. PEA3 enrichment was measured by quantitative PCR and normalized to IgG control. Means and standard deviation of more than three experiments were plotted. Below: Quantitative PCR was used to measure the efficiency of c-JUN siRNA knockdown. Transcripts were normalized to HPRT and means were plotted. Error bars represent standard deviation. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 121: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

100

Co-IP: PEA3

c-JUN

IgG (LC)

PEA3

IgG α-PEA3

Figure 32: Co-Immunprecipitation of PEA3 in Complex with c-JUN.

MDA-MB-231 cells were transfected with PEA3 expression plasmid (pcDNA3.1). Lysates were subjected to chromatin immunoprecipitation and subsequent Western blot analysis using either PEA3 antibody or isotype control IgG. Following Western blot analysis, PEA3 and c-JUN proteins were detected by immunoblotting. Western blot are representative of three independent experiments.

Page 122: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

101

Since c-JUN is known to heterodimerize with the FOS family and to understand if

c-FOS is required for PEA3 enrichment on Notch-1 and Notch-4 promoter regions,

transfected lysates with either CMV-500 empty vector or CMV-500-AFOS, a dominant

negative form of c-FOS (Figure 9), were subjected to chromatin immunoprecipitation.

PEA3 enrichment on either the Notch-1 or Notch-4 promoter was not affected by A-FOS

(Figure 33). Although c-JUN and c-FOS are preferred partners, c-JUN homodimers

indeed exist as well as other FOS family members, which may contribute to PEA3

promoter enrichment. The dominant-negative effect of A-FOS on AP-1 activity was

confirmed by co-transfecting A-FOS and AP-1 luciferase construct and performing a dual

Firefly-Renilla luciferase assays (Figure 34). The results demonstrated a 40% reduction

in AP-1 activity upon A-FOS transfection (Figure 34). Taken together, the results

indicate that AP-1, with emphasis on c-JUN, and PEA3 regulate the Notch-4 promoter in

MDA-MB-231 cells.

3. PEA3 and AP-1 Dynamically Regulate Notch-4 Transcription.

AP-1 members c-JUN, Fra-1, and c-FOS are overexpressed in MDA-MB-231

cells (Bamberger et al., 1999). To address which member(s) of the AP-1 complex is

responsible for the regulation of Notch-4 gene transcription with PEA3, MDA-MB-231

cells were transfected with PEA3, Fra-1, c-JUN, or c-FOS siRNA individually for 48

hours.

As measured by quantitative real-time PCR, Notch-4 transcripts were reduced

upon individual knockdown of PEA3, c-JUN, or Fra-1 and that the siRNA combinations

Page 123: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

102

NOTCH-1

0.0

0.51.0

1.52.0

2.53.0

3.54.0

4.5

c-ETS

PEA3

PEA3 +A-FOS

NOTCH-4

AP-1

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

PEA3

PEA3 +A-FOS

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

NOTCH-1

0.0

0.51.0

1.52.0

2.53.0

3.54.0

4.5

c-ETS

PEA3

PEA3 +A-FOS

NOTCH-4

AP-1

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

PEA3

PEA3 +A-FOS

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Rel

ativ

e Fo

ld In

duct

ion

(spe

cific

Ab

/ IgG

)

Figure 33: Dominant Negative c-FOS ( A-FOS) Has No Effect on PEA3 Recruitment on Notch-1 and Notch-4 Promoters.

MDA-MB-231 cells were co-transfected with PEA3 expression plasmid (pcDNA3.1) and with either CMV-500-vector or A-FOS. Lysates were subjected to chromatin immunoprecipitation using either PEA3 antibody or isotype control IgG on the Notch-1 and 4 promoters, respectively. PEA3 enrichment was measured by quantitative PCR and normalized to IgG control. Means and standard deviation of more than three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 124: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

103

H N3 COOH

DBD ZIPTAD

H N3 COOHH N3H N3 COOH

DBD ZIPTAD

H N3 COOH

DBD ZIPTAD

H N3 COOHH N3H N3 COOH

DBD ZIPTAD

A-FOS

c-FOS

TTS

232 325 331 374

T

LEQRAEE LARENEE LEKEAEE LEQELAE LE

ACIDIC EXTENSION

0.00.20.4

0.60.81.01.2

cmv-500 A-FOS

Fire

fly L

ucife

rase

/ R

enill

a

0.00.20.4

0.60.81.01.2

cmv-500 A-FOS

Fire

fly L

ucife

rase

/ R

enill

a

p<0.05*

Figure 34: Dominant Negative c-FOS (A-FOS) Reduces AP-1 Activity.

Above is a schematic representation of the wildtype and dominant negative c-FOS construct. MDA-MB-231 cells were co-transfected with AP-1 luciferase and either CMV-500-vector or A-FOS. Firefly luciferase (pGL2) was measured normalized to Renilla as an internal transfection control. Means and standard deviation of three or more experiments were plotted.

Page 125: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

104

of either c-JUN:PEA3, Fra-1:PEA3, Fra-1:c-JUN, or PEA3:Fra-1:c-JUN had no

additional effect (Figure 35). Interestingly, in the presence of c-FOS siRNA, Notch-4

transcripts were increased suggesting that c-FOS may act as repressor in our MDA-MB-

231 breast cancer in vitro model. The combination of c-JUN and c-FOS siRNAs had no

measurable effect on Notch-4 transcripts compared to control siRNA suggesting the role

of c-FOS as a potential repressor of Notch-4 (Figure 35). The combination of c-FOS and

c-JUN siRNAs had the tendency to restore the transcript levels of Notch-4 to basal levels.

This result would suggest that c-FOS could be a transcriptional repressor of Notch-4

mRNA. A one-way ANOVA resulted in no statistical significance because of the

pairwise multiple comparisons and minimal repeated measurements (n=3). However,

there is a clear trend and a Power Analysis would help to determine how many

independent studies need to be performed to achieve statistical significance.

As a control experiment, Notch-1 transcripts were measured by quantitative real-

time PCR under the same conditions as mentioned above. As a result, Notch-1

transcripts were not affected by AP-1 knockdown using c-JUN, Fra-1, or c-FOS

siRNA(s). However, PEA3 siRNA reduced Notch-1 transcript levels as seen previously

by 50%, indicating that PEA3 acts most probably independently of AP-1 to regulate the

Notch-1 promoter (Figure 36). Interestingly, c-FOS in combination with PEA3 siRNA

did not reduce Notch-1 transcripts. This suggests that there is a trend that c-FOS may

serve as a repressor upstream or downstream on the Notch-1 promoter. Again, statistical

analysis using one-way ANOVA resulted in no significance because of the pairwise

Page 126: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

105

NOTCH-4

0.0

0.5

1.0

1.5

2.0

2.5

SCRBi PEA3i FRA1i cJUNi cJUNiPEA3i

cJUNiFRA1iPEA3i

FRA1iPEA3i

cJUNiFRA1i

Rel

ativ

e m

RN

A L

evel

s

0.0

0.5

1.0

1.5

2.0

2.5

SCRBi PEA3i cFOSi cJUNi cJUNiPEA3i

cJUNicFOSiPEA3i

cFOSiPEA3i

cJUNicFOSi

Rel

ativ

e m

RN

A L

evel

sNOTCH-4

0.0

0.5

1.0

1.5

2.0

2.5

0.0

0.5

1.0

1.5

2.0

2.5

SCRBi PEA3i FRA1i cJUNi cJUNiPEA3i

cJUNiFRA1iPEA3i

FRA1iPEA3i

cJUNiFRA1iSCRBi PEA3i FRA1i cJUNi cJUNi

PEA3i

cJUNiFRA1iPEA3i

FRA1iPEA3i

cJUNiFRA1i

Rel

ativ

e m

RN

A L

evel

s

0.0

0.5

1.0

1.5

2.0

2.5

0.0

0.5

1.0

1.5

2.0

2.5

SCRBi PEA3i cFOSi cJUNi cJUNiPEA3i

cJUNicFOSiPEA3i

cFOSiPEA3i

cJUNicFOSiSCRBi PEA3i cFOSi cJUNi cJUNi

PEA3i

cJUNicFOSiPEA3i

cFOSiPEA3i

cJUNicFOSi

Rel

ativ

e m

RN

A L

evel

s

Figure 35: PEA3 and AP-1 Dynamically Regulate Notch-4 Transcription. MDA-MB-231 cells were transfected individually or with various combinations of PEA3, Fra-1, c-JUN, or c-FOS siRNA. Notch-4 transcripts were normalized to HPRT and measured by quantitative PCR. Means and standard deviation of three or more experiments were plotted. Statistical significance was determined by one-way ANOVA.

Page 127: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

NOTCH-1

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

SCRBi PEA3i FRA1i cJUNi cJUNiPEA3i

cJUNiFRA1iPEA3i

FRA1iPEA3i

cJUNiFRA1i

Rel

ativ

e m

RN

A L

evel

s

SCRBi PEA3i cFOSi cJUNi cJUNiPEA3i

cJUNicFOSiPEA3i

cFOSiPEA3i

cJUNicFOSi

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

Rel

ativ

e m

RN

A L

evel

sNOTCH-1

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

SCRBi PEA3i FRA1i cJUNi cJUNiPEA3i

cJUNiFRA1iPEA3i

FRA1iPEA3i

cJUNiFRA1i

Rel

ativ

e m

RN

A L

evel

s

SCRBi PEA3i cFOSi cJUNi cJUNiPEA3i

cJUNicFOSiPEA3i

cFOSiPEA3i

cJUNicFOSi

0.0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

Rel

ativ

e m

RN

A L

evel

s

106

Figure 36: Notch-1 Transcripts Are Not Affected by AP-1 siRNA Interference.

MDA-MB-231 cells were transfected individually or with various combinations of PEA3, Fra-1, c-JUN, or c-FOS siRNA. Notch-1 transcripts were normalized to HPRT and measured by quantitative PCR. Means and standard deviation of three or more experiments were plotted. Statistical significance was determined by one-way ANOVA.

Page 128: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

107

multiple comparisons and minimal repeated measurements (n=3). More independent

experiments might be needed to achieve statistical significance.

It is also important to mention that none of the siRNA treatments had any

significant effect on each other, confirming that siRNA effects are not interacting with

gene regulation (data not shown). As a final control, quantitative real-time PCR and

Western blot analysis were performed on PEA3, c-JUN, Fra-1, and c-FOS to determined

their individual siRNA knockdown efficiency. After 48 hours, PEA3, c-JUN, Fra-1, and

c-FOS transcripts and protein levels were reduced by approximately 90%, 40%, 80%,

60% after their specific siRNA transfections, respectively (Figure 37).

SPECIFIC AIM-3:

To Establish a Biological Significance by Determining Whether

Dual Inhibition of Notch and PEA3 Leads to Reduced

Cell Proliferation and Tumorigenicity in vitro.

Notch is vital for proliferation and cell fate determination (Miele et al., 2006)

whereas PEA3 is critical for migration and invasion (Hida et al., 1997; Kaya et al., 1996;

Miele et al., 2006). Both aberrant and unregulated activities can lead to malignancy and

overall poor survival (Reedijk et al., 2005; Shepherd et al., 2001). To understand the

biological significance and their effect in our breast cancer model, we explored dual

Page 129: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

108

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi c-JUNi

Rel

ativ

e m

RN

A L

evel

s

c-JUN

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi c-JUNi

Rel

ativ

e m

RN

A L

evel

s

c-JUN

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

PEA3

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi PEA3i

Rel

ativ

e m

RN

A L

evel

s

PEA3

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi FRA-1i

Rel

ativ

e m

RN

A L

evel

s

FRA-1

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi FRA-1i

Rel

ativ

e m

RN

A L

evel

s

FRA-1

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi c-FOSi

Rel

ativ

e m

RN

A L

evel

sc-FOS

0.0

0.2

0.4

0.6

0.8

1.0

1.2

SCRBi c-FOSi

Rel

ativ

e m

RN

A L

evel

sc-FOS

PEA3

ACTIN

c-JUN

ACTIN

FRA-1

ACTIN

c-FOS

ACTIN

p<0.01*

p<0.0001*

p<0.05*

p<0.001*

Figure 37: RNA interference Knockdown of PEA3 and AP-1 Members.

MDA-MB-231 cells were transfected individually PEA3, Fra-1, c-JUN, or c-FOS siRNA. Their transcripts were normalized to HPRT and measured by quantitative PCR to determine siRNA knockdown efficiency. Means and standard deviation of three or more experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 130: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

109

inhibition of PEA3 and Notch using a specific PEA3 siRNA and the pre-clinical γ-

secretase inhibitor (GSI: MRK-003), respectively.

1. Dual inhibition of Notch and PEA3 Inhibits Cell Proliferation.

To determine the effects of dual Notch and PEA3 inhibition on cell proliferation,

MDA-MB-231 cells were transfected with scrambled or PEA3 siRNA alone, or treated

with vehicle or GSI (10 μM), or a combination for 48 hours. Independently, PEA3

knockdown and/or Notch inhibition all had effects on cell cycle (Figure 38). PEA3

knockdown or GSI treatment of MDA-MB-231 cells alone showed an increase in G1

arrest. A significant increase (p<0.05) in G1 arrest was determined in the combination

group of Notch inhibition and PEA3 knockdown as compared to control (Figure 38).

PEA3 knockdown and GSI treatment alone showed a reduction in S-phase, while the

combination of Notch inhibition and PEA3 knockdown significantly reduced S-phase

(p<0.05) as compared to control (Figure 38). PEA3 siRNA and GSI treatment alone

demonstrated a reduction in G2/M cell cycle, but the combination showed a significant

reduction (p<0.05) as compared to control in G2/M (Figure 38). These results indicate

that both PEA3 and Notch, whether working together or independently at the various

stages of the cell cycle, are critical for proliferation of MDA-MB-231 cells.

2. Dual Inhibition of Notch and PEA3 Induces Apoptosis.

To address whether dual inhibition using both PEA3 knockdown and Notch

inhibition affected cell viability potentially through increased apoptosis, MDA-MD-231

Page 131: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

110

0.0010.0020.0030.0040.0050.0060.0070.0080.0090.00

100.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

% of Cells in

Figure 38: Effects of Dual Notch and PEA3 on Cell Proliferation.

MDA-MB-231 cells were transfected with scrambled or PEA3 siRNA alone, or treated with vehicle or a gamma-secretase inhibitor (GSI: MRK-003, 10 μM), or a combination thereof for 48 hours. Cell cycle analysis was performed by flow cytometry. Mean percentage and standard deviation of cells in each experiment were plotted. Statistical significance was determined by one-way ANOVA.

G1-phase

Perc

ent o

f Cel

ls in

G1

* p<0.05

0.00

5.00

10.00

15.00

20.00

25.00

30.00

35.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

Perc

ent o

f Cel

ls in

S

% of Cells in S-phase

* p<0.05

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

0.00

2.00

4.00

6.00

8.00

10.00

12.00

14.00

16.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

0.00

2.00

4.00

6.00

8.00

10.00

12.00

14.00

16.00

Perc

ent o

f Cel

ls in

G2/M

% of Cells in G2/M-phase

* p<0.05

Page 132: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

111

cells were transfected with scrambled or PEA3 siRNA alone, or treated with vehicle or

GSI (10 μM), or a combination, and subjected to Annexin-V staining, and subsequently

measured by flow cytometry 24 hours later as an early marker of apoptosis.

Independently, PEA3 knockdown showed no change in apoptosis (Figure 39). GSI

treatment alone increased apoptosis by 30%. Importantly, the combination of PEA3

knockdown and GSI treatment significantly increased apoptosis by almost 40% as

measured by Annexin-V positive stained cells (Figure 39).

3. Dual Inhibition of Notch and PEA3 Reduces Cell Viability.

To confirm the effects of treatment/transfection on cell viability, a Trypan blue

exclusion assay was performed after MDA-MD-231 cells were transfected with

scrambled or PEA3 siRNA alone, or treated with vehicle or GSI (10 μM), or a

combination for 48 hours. PEA3 siRNA alone showed no change in cell viability, but

GSI alone resulted in decreased viability (Figure 40). In agreement with previous

experiments, the combination of PEA3 knockdown and GSI treatment significantly

diminished percent cell viability by more than 50% (Figure 40). These results taken

together with the cell cycle data indicate that both PEA3 and Notch activities together are

essential for cell proliferation and survival in MDA-MB-231 breast cancer cells.

4. Notch-1 and Notch-4 Overexpression Rescues the Effect of Dual Notch and

PEA3 Inhibition.

Page 133: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

112

% o

f Ann

exin

-V +

Cel

ls

05

10152025303540

24 Hours

p<0.05

VEHICLEPEA3 siRNAGSI (Mrk-003)

+ – – –– + – + – – + +

*%

of A

nnex

in-V

+ C

ells

05

10152025303540

24 Hours

p<0.05

VEHICLEPEA3 siRNAGSI (Mrk-003)

+ – – –– + – + – – + +

*

Figure 39: Dual Inhibition of Notch and PEA3 Increases Apoptosis

MDA-MB-231 cells were transfected with scrambled or PEA3 siRNA alone, or treated with vehicle or a gamma-secretase inhibitor (GSI: MRK-003, 10 μM), or a combination thereof for 24 hours. Annexin-V staining was performed and measured by flow cytometry. Mean percentage and standard deviation of cells in each experiment were plotted. Statistical significance was determined by one-way ANOVA.

Page 134: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

113

0102030405060708090

100

48 Hours

p<0.05%

Cel

l Via

bilit

y

VEHICLEPEA3 siRNAGSI (Mrk-003)

+ – – –– + – + – – + +

*

0102030405060708090

100

48 Hours

p<0.05%

Cel

l Via

bilit

y

VEHICLEPEA3 siRNAGSI (Mrk-003)

+ – – –– + – + – – + +

*

Figure 40: Dual Inhibition of Notch and PEA3 Decrease Cell Viability. MDA-MB-231 cells were transfected with scrambled or PEA3 siRNA alone, or treated with vehicle or a gamma-secretase inhibitor (GSI: MRK-003, 10 μM), or a combination thereof for 48 hours. Percent viable cells were measured by Trypan blue exclusion study using a standard light microscope (10X magnitude). Mean percentage and standard deviation of cells in each experiment were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 135: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

114

To address whether dual inhibition of PEA3 and Notch activities have effects on

anchorage independent growth as a measure of cell tumorigenicity, a colony formation

assay was performed with MDA-MB-231 cells that were transfected with either

scrambled or PEA3 siRNA and treated with vehicle or a gamma-secretase inhibitor (GSI:

MRK-003, 5 μM) independently or in combination. Forty-eight hours after transfection,

cells were suspended in methylcellulose and allowed to grow into colonies over 14 days.

Colonies were photographed (Figure 41) and counted under a 40X magnification using a

standard light microscope, and quantified (Figure 42). As a result, there was a reduction

in the number of colonies to almost 60% after PEA3 siRNA knockdown or by 40% when

Notch was inhibited using a GSI in the presence of vector alone. Furthermore, the percent

number of colonies was reduced to 40% upon the combination of PEA3 knockdown and

GSI treatment (Figure 41, 42).

GSI inhibits cleavage of all four Notch receptors (Kopan and Ilagan, 2009). To

confirm the hypothesis that the effects of PEA3 inhibition on cell growth and survival are

mediated primarily through Notch-1 and/or Notch-4, in MDA-MB-231 cells expression

plasmids for the Notch-1 or Notch-4 intracellular domain (N1IC or N4IC) (10 ng) were

simultaneously co-transfected independently or in combination in all four treatment

groups (i.e. vehicle control, PEA3 siRNA, GSI, or GSI plus PEA3 siRNA) (Figure 41,

column 2, 3, 4, quantified Figure 42). When Notch-1IC, Notch-4IC, or both were

expressed in the absence of PEA3 siRNA, GSI treatment, or their combination, there was

no measurable difference in percent number of colonies compared to control (Figure 41,

row 1: E, I, M compared to control A, quantified Figure 42). The addition of Notch-1IC

Page 136: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

115

NOTCH-1 (IC) NOTCH-4 (IC) NOTCH-1 (IC)NOTCH-4 (IC)

VEHICLE

PEA3i

GSI (5uM)

PEA3iGSI (5uM)

VECTOR

ROW-1

ROW-2

ROW-3

ROW-4

COLUMN-1 COLUMN-2 COLUMN-3 COLUMN-4

A

B

C

D

E

F

G

H

I

J

K

L

M

N

O

P

Figure 41: Notch-1 and Notch-4 Overexpression Rescues the Effect of Dual Notch and PEA3 Inhibition.

MDA-MB-231 cells were transfected with either scrambled or PEA3 siRNA and treated with vehicle or gamma-secretase inhibitor (GSI: MRK-003, 5 μM) independently or in combination for 48 hours. Simultaneously, cells were co-transfected with expression plasmids for Notch-1 and Notch-4 intracellular domain (IC) independently or in combination. Colony formation assay was preformed using methylcellulose and incubated for 14 days. Colonies were photographed using a standard light microscope (40X magnitude) representative of three or more experiments.

Page 137: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

116

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-1 (IC) + NOTCH-4 (IC)

Perc

ent #

of C

olon

ies

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-1 (IC) + NOTCH-4 (IC)

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-1 (IC) + NOTCH-4 (IC)

Perc

ent #

of C

olon

ies

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-1 (IC)

Perc

ent #

of C

olon

ies

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-1 (IC)

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-1 (IC)

Perc

ent #

of C

olon

ies

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-4 (IC)

Perc

ent #

of C

olon

ies

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-4 (IC)

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

0.00

20.00

40.00

60.00

80.00

100.00

120.00

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

GSI +DMSOSCRBi

GSISCRBi PEA3i

DMSOPEA3i

VECTOR NOTCH-4 (IC)

Perc

ent #

of C

olon

ies

**

p<0.05p<0.05

**

p<0.05p<0.05

**

p<0.05p<0.05

Figure 42: Quantification of Notch-1 and Notch-4 Rescue on Colony Formation. MDA-MB-231 cells were transfected with either scrambled or PEA3 siRNA and treated with vehicle or gamma-secretase inhibitor (GSI: MRK-003, 5μM) independently or in combination for 48 hours. Simultaneously, cells were co-transfected with expression plasmids for Notch-1 and Notch-4 intracellular domain (IC) independently or in combination. Colony formation assay was performed using methyl-cellulose and incubated for 14 days. Nine fields were counted and quantified per well and means plotted. Statistical significance was determined by one-way ANOVA.

Page 138: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

117

alone and partially rescued the effects mediated by PEA3 siRNA, GSI, and their

combination on percent colony formation (Figure 42, top). Notch-4IC alone partially

rescued the effects in anchorage independent growth in the presence of GSI treatment

alone and in combination with PEA3 siRNA. Notch-4IC rescue effects on PEA3 siRNA

transfection alone were minimal (Figure 42, middle). In the presence of dual inhibition of

PEA3 and Notch, Notch-1IC expression was able to restore anchorage independent

growth by 80% than Notch-4IC in which only 70% of colony number was restored (Figure

41, row 4: H and L compared to D, respectively, Figure 42). When both Notch-1IC and

Notch-4IC were co-expressed in this combination treatment group, complete restoration of

percent number of colonies was observed by 93% (Figure 41, row 4: P compared to D,

Figure 42, bottom). This result suggests that PEA3 mediates its effect primarily through

expression of Notch-1 and Notch-4. While Notch-1IC had a greater colony restoration

effect than Notch-4IC, both receptors are required to fully restore both the phenotypic

appearance and percent number of colonies. Taken together, the results suggest that

transcription of Notch-1 and Notch-4 genes by PEA3 and the activation of Notch-1 and

Notch-4 receptors (after gamma-secretase cleavage leading to the formation of active

Notch-1IC and Notch-4IC) are both critical biologic events required for proliferation and

survival of triple negative, MDA-MB-231 breast cancer cells in vitro.

5. PEA3 Regulates Notch-1 and Notch-4 mRNA Expression in other

Subtypes of Breast Cancer.

Page 139: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

118

Our investigations are the first to show that PEA3 is an activator of Notch-1 mRNA and

protein expression and Notch-4 mRNA in the triple negative subtype, MDA-MB-231

breast cancer cells. To determine whether this is a common mechanism among the other

subtypes of breast cancer cells, MDA-MB-231 (triple-negative), MCF-7 (Luminal A),

BT474 (Luminal B), and SKBr3 (HER2+) cells were transfected with either scrambled or

PEA3 siRNA. Normalized to MDA-MB-231 cells, MCF-7 cells had slightly less PEA3

transcript levels followed by BT474 cells and then SKBr3 cells (Figure 43). Notch-1 and

Notch-4 transcript levels were compared with either scrambled or PEA3 siRNA among

the cell lines. PEA3 regulation of Notch-1 mRNA was seen in MDA-MB-231, SKBr3,

and MCF-7 cells but not BT474 cells (Figure 44, top). However, PEA3-mediated effects

on Notch-4 transcripts were only observed in MDA-MB-231 (Figure 44, bottom). This

could be in part due to differences in AP-1 activity (Figure 45) or that PEA3 basal

transcript levels were very low in other cell types (Figure 43). Provoked by this notion,

we asked if we could drive the expression of Notch-4 by increasing levels of PEA3 in

breast cancer cells that express low endogenous PEA3 levels. To address this, BT474

and SKBr3 cells were transfected with either pcDNA3.1 empty vector or pcDNA3.1-

PEA3 expression plasmid. Notch-1 transcripts remained unchanged when PEA3 was

overexpressed (Figure 46). Notch-4 transcript levels were significantly increased by nine

fold in BT474 and sixteen fold in SKBr3 breast cancer cells (Figure 46). As a control,

quantitative PCR was performed on PEA3 transcript levels after transfection of

pcDNA3.1 and pcDNA3.1-PEA3 to determine PEA3 overexpression efficiency in both

BT474 and SKBr3 cells (Figure 46, bottom). Taken together, PEA3 is required for

Page 140: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

119

Notch-1 transcription in at least three subtypes of breast cancer cells: MDA-MB-231,

SKBr3, and MCF-7. However, PEA3 is an activator of Notch-4 transcription in MDA-

MB-231 cells where endogenous PEA3 levels are high. Exogenous expression of PEA3

in BT474 and SKBr3 cells provides evidence that it is also a potential activator of Notch-

4 expression in other breast cancer subtypes.

Taken together, results from this study demonstrate for the first time that PEA3 is

a novel activator of Notch-1 and Notch-4 transcription in different subtypes of breast

cancer cells and could prove to be an important therapeutic target (targeted by

nanotechnology, directed siRNAs, or staple interface masking peptides) when combined

with a Notch inhibitor such as a GSI, particularly in triple negative breast cancer.

Page 141: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

120

MDAMB231 SKBr3MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3

i0.00.10.20.30.40.50.60.70.80.91.01.1

PEA3R

ELA

TIVE

mR

NA

LEVE

LSp<0.0001*

p<0.0001*

p<0.001*

p<0.001*

MDAMB231 SKBr3MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3

i0.00.10.20.30.40.50.60.70.80.91.01.1

PEA3R

ELA

TIVE

mR

NA

LEVE

LSp<0.0001*

p<0.0001*

p<0.001*

p<0.001*

Figure 43: Relative PEA3 levels and Knockdown Efficiency

in Breast Cancer Subtypes. MDA-MB-231, MCF-7, BT474, and SKBr3 cells were transfected with either scrambled or PEA3 siRNA. Normalized to MDA-MB-231 cells, the transcript levels of PEA3 were compared and measured by quantitative PCR. Means and standard deviation of three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 142: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

121

0123456789

10111213

REL

ATI

VE m

RN

A LE

VELS

NOTCH-1

MDAMB231 SKBr3 MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

0123456789

10111213

REL

ATI

VE m

RN

A LE

VELS

NOTCH-1

MDAMB231 SKBr3 MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

MDAMB231 SKBr3 MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

0.00.20.40.60.81.01.21.41.61.82.02.22.42.6

REL

ATI

VE m

RN

A L

EVEL

S

NOTCH-4

MDAMB231 SKBr3 MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

0.00.20.40.60.81.01.21.41.61.82.02.22.42.6

REL

ATI

VE m

RN

A L

EVEL

S

NOTCH-4

MDAMB231 SKBr3 MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

MDAMB231 SKBr3 MCF-7 BT474

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

SCRBi

PEA3i

p<0.001*

p<0.001

**

*

p<0.01

p<0.01

Figure 44: PEA3 Regulates Notch in Other Breast Cancer Subtypes. MDA-MB-231, SKBr3, MCF-7, and BT474 cells were transfected with either scrambled or PEA3 siRNA. Normalized to MDA-MB-231 cells, the transcript levels of Notch-1 and Notch-4 were compared and measured by quantitative PCR. Means and standard deviation of three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 143: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

122

Relative AP-1 Activity in Cell Lines

0.0

0.2

0.4

0.6

0.8

1.0

1.2

BT474 MDAMB231 MCF-7 SKBr3

Fire

fly L

ucife

rase

/ R

enill

a

Relative AP-1 Activity in Cell Lines

0.0

0.2

0.4

0.6

0.8

1.0

1.2

BT474 MDAMB231 MCF-7 SKBr3

Fire

fly L

ucife

rase

/ R

enill

a

Figure 45: Relative AP-1 Activity in Different Breast Cancer Subtypes. BT474, MDA-MB-231, MCF-7, and SKBr3 cells were transfected with an AP-1 luciferase reporter construct (pGL2). After 48 hours, 10uL of each sample was added to a white coated luminescent 96-well plate and subjected to the manufactured dual-luciferase assay. Luminescence was measured using the Veritas Microplate Luminometer. The range of luminescence was between 350-650nm; Firefly luciferase read at 560nm and pTL-TK at 480nm. Firefly luciferase was measured and normalized to Renilla as an internal transfection control. Means and standard deviation of three or more experiments were plotted.

Page 144: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

123

0

1

2

3

4

5

6

7

8

9

10

11

12

Vector Alone

Vector Alone

PEA3 over-expression

PEA3 over-expression

NOTCH-1 NOTCH-4

Rel

ativ

e m

RN

A L

evel

s (N

orm

aliz

ed to

Vec

tor C

ontr

ol)

BT474 Cells

* p < 0.05

0

1

2

3

4

5

6

7

8

9

10

11

12

0

1

2

3

4

5

6

7

8

9

10

11

12

Vector Alone

Vector Alone

PEA3 over-expression

PEA3 over-expression

NOTCH-1 NOTCH-4

Rel

ativ

e m

RN

A L

evel

s (N

orm

aliz

ed to

Vec

tor C

ontr

ol)

BT474 Cells

* p < 0.05

0

2

4

6

8

10

12

14

16

18

20

Vector Alone

Vector Alone

PEA3 over-expression

PEA3 over-expression

NOTCH-1 NOTCH-4

Rel

ativ

e m

RN

A L

evel

s(N

orm

aliz

ed to

Vec

tor C

ontr

ol)

SKBr3 Cells

* p < 0.05

0

2

4

6

8

10

12

14

16

18

20

0

2

4

6

8

10

12

14

16

18

20

Vector Alone

Vector Alone

PEA3 over-expression

PEA3 over-expression

NOTCH-1 NOTCH-4

Rel

ativ

e m

RN

A L

evel

s(N

orm

aliz

ed to

Vec

tor C

ontr

ol)

SKBr3 Cells

* p < 0.05

Rel

ativ

e Fo

ld In

duct

ion

0

5

10

15

20

25

30

490

500

510

Std = ±200

BT474 cells

Vector Alone

PEA3 over-expression

Rel

ativ

e Fo

ld In

duct

ion

0

5

10

15

20

25

30

300

310

320 Std = ±75

SKBr3 cells

Vector Alone

PEA3 over-expression

Rel

ativ

e Fo

ld In

duct

ion

0

5

10

15

20

25

30

300

310

320 Std = ±75

SKBr3 cells

Vector Alone

PEA3 over-expression

PEA3 PEA3 Figure 46: PEA3 Regulates Notch-4 in BT474 and SKBr3 Breast Cancer Subtypes.

BT474 and SKBr3 cells were transfected with either empty vector or PEA3 expression plasmid (pcDNA3.1). Notch-1, Notch-4, and PEA3 mRNA levels were normalized to empty vector and measured by quantitative PCR. Means and standard deviation of three experiments were plotted. Statistical significance was determined by two-tailed unpaired Student’s t test.

Page 145: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

CHAPTER V

DISCUSSION

PEA3 was originally identified as a member of the Ets family of transcription

factors (Xin et al., 1992). Since then, it has been observed that PEA3 is expressed during

breast development, is quickly lost upon maturation, and yet re-emerges in metastatic

breast cancers (Benz et al., 1997; Chotteau-Lelievre et al., 2003). Similarly, Notch-1 has

been implicated in breast cancer demonstrating elevated expression and activity in breast

tumors (Reedijk et al., 2005). Little is known about the factors that influence Notch gene

expression, and why its levels are elevated in breast cancer.

MDA-MB-231 cells are an in vitro model of triple negative breast cancer and

therefore lack expression of estrogen receptor α, progesterone receptor, or HER2

overexpression (Sorlie et al., 2001). They also contain a p53 mutation (Weigelt et al.,

2009). Triple negative breast cancers have no known FDA approved, targeted treatment

strategies, and as of yet have limited direct targeted chemotherapeutics. Their standard of

care is a combination of cytotoxic drugs: cyclophosphamide, doxorubicin, and

fluorouracil (Hortobagyi, 1998) or recently shown to be more effective in triple-negative

breast cancer was the combination of cyclophosphamide, methotrexate, and fluorouracil

(Colleoni et al., 2010). Therefore, it is imperative to understand the molecular

infrastructure of triple-negative breast cancers to uncover and provide novel targeted

124

Page 146: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

125

methods to help these patients reach their five year and beyond disease-free survival rate.

The Analysis

The initial results indicating that PEA3 is a transcriptional activator of Notch-1

and Notch-4 in triple-negative MDA-MB-231 cells provides new insight into targeted

therapeutic options for patients afflicted with this illness. Targeting PEA3 in

combination with inhibition of Notch shows promise in vitro to decrease MDA-MB-231

cellular viability (Figure 40), reduce cell proliferation (Figure 38), and increase apoptosis

(Figure 39). What is even more intriguing is the result that PEA3 inhibition slightly, but

significantly increased the transcript levels of Notch-2 (Figure 14). Notch-2 recently has

been implicated as a tumor suppressor (O'Neill et al., 2007; Quillard et al., 2009).

Minimal putative PEA3 Ets sites, although less abundant, were identified on the Notch-2

promoter (data not shown). This could indicate that targeting PEA3 in MDA-MB-231

could be beneficial in two ways: abrogating Notch-1 and Notch-4 transcripts and

stimulating expression of Notch-2, thus increasing the chances of a complete reduction in

proliferation and survival of the cancerous cells. This notion stems from other

investigations of PEA3, which demonstrate it to be both an activator and repressor of

gene transcription (Benz et al., 1997; Scott et al., 1994; Xia et al., 2006; Xing et al.,

2000).

It was demonstrated that PEA3 siRNA alone showed no effect on cell

proliferation (Figure 38), apoptosis (Figure 39), and viability (Figure 40) in vitro

indicating that it is not efficient enough to inhibit PEA3 alone. Firstly, endogenous levels

Page 147: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

126

of Notch-4 protein were not changed after PEA3 siRNA but were altered in the presence

of lactacystin showing that inhibition of proteasome-mediated protein degradation

inhibited pathways that resulted in transcription becoming rate controlling. Secondly,

Notch-2 transcripts, although modest, were significantly increased in the presence of

PEA3 siRNA and Notch-3 transcripts unaffected. A gamma-secretase inhibitor inhibits

the third proteolytic cleavage responsible for the formation of active intracellular Notch-

1,2,3, and 4 (Weijzen et al., 2002). The results shown here suggest that PEA3 only

activates Notch-1 and Notch-4 transcripts. Therefore, one could speculate that the Notch-

3 receptor could compensate for the dampening of Notch-1 and Notch-4 mRNA, and

consequently maintain cell proliferation and viability, which were observed in these

studies.

Notch-4 transcripts were positively regulated by PEA3 (Figure 14), however

steady-state Notch-4 protein levels were not. In the presence of lactacystin, a specific

proteasome inhibitor, the transcriptional effects of PEA3 on Notch-4 were observed

(Figure 14). A speculative explanation could be that the rate of protein degradation could

be decreased upon knockdown of PEA3, which may be a result of PEA3 regulating

components of the proteasome. Another hypothesis could be that post-transcriptional

steps regulating Notch-4 protein levels must be important since the endogenous Notch-4

protein levels do not correlate with Notch-4 mRNA levels. Examples of post-

transcriptional steps include translation regulation, vesicular transport of Notch-4 protein

through the endoplasmic reticulum and Golgi apparatus, ligand activation and gamma-

secretase cleavage regulating plasma membrane Notch-4 protein levels, and proteasome

Page 148: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

127

degradation of ubiquitinated Notch protein or other proteins involved in Notch-4 turnover.

However, several experiments are necessary to answer these speculations. Nevertheless,

the results suggest that Notch-4 is primarily regulated by the proteasome, but when

compromised, the transcriptional effects of PEA3 on Notch-4 protein levels are

observable.

To date, PEA3 has been implicated in regulating various genes in a complex with

additional transcription factors (Firlej et al., 2005; Hesselbrock et al., 2005; Liu et al.,

2004; Matsui et al., 2006). It was observed that AP-1 and PEA3 regulate Notch-4

transcription (Figures 27, 29, and 32). The results of this investigation suggest the

importance of c-JUN, Fra-1, c-FOS, and PEA3 on the regulation of Notch-4 transcription.

Interestingly, Fra-1 and c-FOS also regulate the Notch-4 promoter indicating that they

either activate or dampen the transcription of Notch-4, respectively (Figure 35). This

provides a unique future direction of understanding the partner preference on the Notch-4

promoter and the homeostatic balance needed for appropriate expression of Notch-4 in

response to cellular stimuli. Recently, Baan et al. demonstrated that invasive breast

cancer cells including MDA-MB-231 favored the c-JUN:Fra-1 heterodimer, compared to

less invasive breast cancer cells, which favored the c-JUN:c-FOS heterodimer (Baan et

al., 2010). Preference towards the c-JUN:Fra-1 AP-1 dimer may be responsible for

Notch-4 mRNA modulation and subsequent tumor aggressiveness of MDA-MB-231

triple negative breast cancer cells. The data suggests that PEA3 expression could tip the

balance and aid in this partner choice.

Page 149: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

128

The partner choice of the AP-1 complex can be a heterodimer of JUN and FOS

families or a homodimer of the JUN family (Eferl and Wagner, 2003). It was interesting

to find that c-JUN was required for PEA3 recruitment to the Notch-4 promoter and that c-

FOS was not as shown by the dominant negative ChIP studies (Figure 29 and Figure 33).

Dominant negative c-JUN and c-FOS experiments may answer distinct questions. c-JUN

is highly expressed in triple-negative MDA-MB-231 cells (Bamberger et al., 1999).

TAM-67 only reduced AP-1 function by 50% (Figure 29). However, JUN:FOS is the

preferred partnership, with c-JUN acting as an important member capable of homo- and

hetero-dimerization among family members leading to subsequent AP-1 signaling effects

(Eferl and Wagner, 2003). Thus upon transfection, TAM-67 has the capability to partner

with JUN and FOS families limiting the cellular pool of potent gene regulators (Brown et

al., 1994; Brown et al., 1996). A-FOS on the other hand can only partner the JUN family

and not the FOS family (Olive et al., 1997). Therefore, important FOS family members

are potentially left available to still regulate the Notch-4 promoter, namely Fra-1.

Since dynamic transcription factor partnering is plausible (Beckett, 2004), other

putative regulatory factors must be critical for the regulation of Notch genes, because

PEA3 is not solely responsible for the regulation of Notch-1 and Notch-4 transcription.

This is evident by the observation that PEA3 RNA interference decreased Notch-1 and

Notch-4 transcripts by 50% and 70% (Figure 14). If PEA3 was the sole transcription

factor necessary, the transcripts would have been completely eliminated upon PEA3

knockdown. The results of this investigation demonstrate that PEA3 is one of many

factors involved in the regulation of Notch transcription. A very dynamic interplay by

Page 150: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

129

many additional factors may be responsible for the appropriate regulation of Notch-1 and

Notch-4 transcripts. Moreover, key areas flanking the start site of the Notch-1 and

Notch-4 gene including downstream intronic and extronic regions, unidentified cryptic

sites, or regulatory transcriptional hubs within the gene (areas of three dimensional gene

locus looping as seen in the CFTR gene (Ott et al., 2009)) could be putative sites of

regulation. Endogenous genes are highly coiled, compact, and require chromatin

remodeling events (Zhao et al., 2009). Therefore, PEA3 in addition with other putative

factors potentially carrying histone acetyltransferases (Liu et al., 2004; Matsui et al.,

2006) activity may be required to regulate Notch gene transcription.

These putative factors are yet to be identified especially on the Notch-1 promoter

where the investigations indicate that PEA3 works independently of AP-1 (Figure 36).

However, PEA3 is known in the literature to work in tandem with other transcription

factors at variable genomic positions to regulate its target genes (Firlej et al., 2005; Liu et

al., 2004). Therefore, it is highly unlikely that PEA3 independently regulates Notch-1

without the aid of other factors. Other consensus sites on the Notch-1 promoter near

PEA3 sites are putative NFκB and SP-1 sites (data not shown) as determined by promoter

scanning using NCBI Entrez Gene bank. These factors have not been significantly tested

in vitro to discern if they interact with PEA3, but are emerging as transcription factors of

interest in the regulation of Notch-1. Moreover, if in addition to directly regulating

Notch-1 transcripts, PEA3 carries factors with chromatin remolding capabilities, then the

dynamics of PEA3 regulation becomes more fascinating allowing for remodeling events

(Clapier and Cairns, 2009; Zhao et al., 2009) to occur, exposing silent regulatory sites,

Page 151: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

130

and recruiting other transcription factors possibly the factors mentioned above to regulate

the Notch-1 receptor or even other Notch receptors as well. This requires further studies

to address, but becomes an additional hypothesis. Taken together, PEA3 harbors very

interesting properties in response to cellular stimuli, not only limited to its effects on gene

regulation, but also its effects mediated by pharmacological intervention.

It was demonstrated that dual inhibition of Notch via a gamma-secretase inhibitor

(MRK-003) and PEA3 via siRNA, reduced proliferation (Figure 38), anchorage-

independent growth (Figure 41 and Figure 42), and increased apoptosis in MDA-MB-231

cells (Figure 39). The enhanced sensitivity of MDA-MB-231 cells towards the GSI in

the presence of PEA3 siRNA could be a result of targeting both the transcription and

activity of the Notch pathway (Figure 47). GSI treatment inhibits the third proteolytic

cleavage and attenuates the signaling cascade (Weijzen et al., 2002). However, more

receptors are continually being transcribed at the gene level without control. Thus, it

could be hypothesized that inhibiting at the level of gene transcription and at the level of

receptor cleavage could greatly enhance the inhibition of Notch signaling overall. It was

observed that PEA3 inhibition alone was not sufficient to reduce anchorage dependent

and affect anchorage independent proliferation (Figure 38 and Figure 42) most likely due

the fact that other regulatory factors (Alimirah et al., 2007; Cai et al., 2009; Pajerowski et

al., 2009; Wu and Bresnick, 2007; Wu et al., 2005) including AP-1 (Wu and Bresnick,

2007; Wu et al., 2005) may be important in regulation of Notch transcripts.

PEA3 regulation of Notch-1 and Notch-4 transcripts was observed among other

subtypes of breast cancer (Figure 44) in vitro. Notch-4 transcripts were affected in the

Page 152: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

131

presence of exogenous PEA3 expression (Figure 44). This could be in part due to the

differences in AP-1 activity in the cell types (Figure 45), or that the endogenous levels of

PEA3 differ (Figure 43). BT474 cells have endogenously high activity of AP-1, and

SKBr3 have lower activity (Figure 45). However, in SKBr3 and BT474 cells, in which

PEA3 is endogenously low, exogenous expression of PEA3 increased Notch-4 transcripts

(Figure 46). This suggests together with the Luciferase results seen prior (Figure 27) that

PEA3 is a potent activator of Notch-4 transcription and the levels of AP-1 activity,

although important, may have a secondary role in Notch-4 transcription.

It was observed that PEA3 or GSI alone was able to reduce colony formation by

approximately 40-50% (Figure 42), but the combination was able to reduce colony

formation by more than 60% (Figure 42). When Notch-1IC was transfected into MDA-

MB-231 cells, restoration of colony formation number was quantified (Figure 42),

indicating that Notch-1 may be sufficient to restore the effects of PEA3, GSI, and dual

inhibition. Notch-4IC transfection also restored colony formation but not as sufficiently

as Notch-1IC (Figure 42). Initially, the effects of treatment/transfection appear to be

mediated through Notch-1. However upon closer observation, although Notch-1

exogenous expression produced similar colony numbers as compared to control, the

colonies were smaller in size when visualized under a 40X magnification of a light

microscope (Figure 41). This observation was not quantified, but merely observed.

Notch-4 exogenous expression did not rescue colony number as sufficiently as Notch-1,

but the phenotype of the colonies (not quantified data, mere observation) were similar to

control; the colonies were repeatedly observed to appear more robust and similar in size

Page 153: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

132

to control (Figure 41). Interestingly, dual exogenous expression not only restored the

colony formation number, but also the phenotype of the colonies when compared to

control (not quantified data, mere observation) (Figure 41). This could be explained by

the notion that Notch-1 along with its ligand, Jagged-1, are highly regulated receptors and

have been implicated in cell proliferation and growth (Dickson et al., 2007; Reedijk et al.,

2005). Notch-4 has been implicated in mammary cancer stem cells (Harrison et al.,

2010). Taken together, Notch-1 exogenous expression could be responsible for

proliferation of the cells thus acquiring similar colony numbers (in a agreement to what

was observed using a peptidyl γ-secretase inhibitor, z-Leu-Leu-Nle-CHO reviewed in

Chapter II and (Clementz and Osipo, 2009), in MDA-MB-231 cells (Lee et al., 2008)),

but Notch-4 could be responsible for stimulating the mammary cancer stem cells

(Harrison et al., 2010) creating a more robust colony from mere observation. Together,

they restore the effects of PEA3 and Notch inhibition by possibly stimulating both

proliferating cells and stem cells.

In addition, Harrison et al. has recently implicated Notch-4 in mammary tumor

stem cell survival and self-renewal in which they demonstrated that targeting Notch-4 via

siRNA interference specifically was more effective than a GSI in inhibiting the Notch

pathway (Harrison et al., 2010). In these studies, Notch-4 gene transcription was more

sensitive to PEA3 inhibition. Given this fact, the sensitivity obtained in our MDA-MB-

231 system by the dual inhibition of PEA3 and Notch towards reduced viability (Figure

40) may be explained by the notion that we may be not only targeting proliferation and

Page 154: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

133

survival of bulk cancer cell populations but also the cancer stem cell population (Harrison

et al., 2010; Korkaya and Wicha, 2007; Korkaya and Wicha, 2009).

Notch is a cellular fate determinant, can induce proliferation, and/or

differentiation depending on the cellular environment (Kopan and Ilagan, 2009). PEA3

has been linked to invasion, migration, and aggressiveness of tumor cells (Coutte et al.,

1999). The dual inhibition of Notch by the GSI, and the inhibition of PEA3 by siRNA

could act by preventing two vital arms of cancer progression, namely proliferation and

possibly metastatic potential in vivo. Emerging nanotechnology as a means to direct

siRNA therapies (Del Pino et al., 2010), recent advances in targeted siRNA treatments in

vivo (Ueyama et al., 2010), and the advent of stapled interface peptides (Moellering et al.,

2009) that disrupt transcription factor complexes transforms the notion for specific

targeting of PEA3 into a potential reality.

Here, these studies have provided evidence of a novel combination treatment to

be exploited for the treatment of triple-negative breast cancer, and potentially other breast

cancer subtypes where PEA3 regulates Notch-1 and Notch-4. The findings offer a

potential mechanism that has identified better strategies for future pre-clinical, in vivo

studies which are currently underway. Enhanced sensitivity towards current GSIs in

future clinical trials by inhibition of PEA3 by nanoparticles (Del Pino et al., 2010), small

molecule inhibitors, or future siRNA approaches (Ueyama et al., 2010) may increase

patient response to treatment and could reduce or eliminate recurrence if stem cell

populations are eliminated (Korkaya and Wicha, 2009). Inhibiting PEA3 in combination

with the GSI may also allow for a larger therapeutic window for the use of a GSI,

Page 155: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

134

enabling the reduction of pharmacological doses given to the patient and thus limiting the

resulting undesirable side effects such as gastrointestinal toxicity (Staal and Langerak,

2008; van Es et al., 2005).

Furthermore, the implications of these results lead to many areas of active

research within the clinic. PEA3 expression in breast cancer could become a prognostic

indicator of how a patient may respond to Notch inhibition by a GSI since endogenous

levels of PEA3 (high or low) could determine whether or not a combination treatment is

desirable. From the results observed when PEA3 was endogenously elevated, inhibition

of PEA3 sensitized the cells towards the GSI (Figures 38-42). Therefore, if a patient

presents in the clinic with elevated endogenous levels of PEA3, it may be beneficial to

inhibit both PEA3 and Notch to achieve tumor regression. Inversely, if a patient presents

with endogenously low levels of PEA3, Notch inhibition via a GSI may be only required

as signal targeted therapeutic strategy. Taken together, a patient may acquire a more

unique chemotherapeutic strategy and reduce unnecessary combinatory strategies as

demonstrated with these results. Oncogenic gene profiling acquired by a primary biopsy

is currently measured to decide what appropriate treatment is beneficial to the patient

(Zanetti-Dallenbach et al., 2006). Diagnosis of HER2, estrogen receptor α, and

progesterone receptor expression are already used in the clinic (Carter et al., 1989; Galea

et al., 1992; Mirza et al., 2002). Thus, PEA3 could be another protein to screen to

determine the necessary therapeutic strategy if methods of PEA3 are available. Moreover,

AP-1 activity independently may also prove to be significant in deciding treatment, since

the results indicate that PEA3 and AP-1 are interactive partners in certain breast cancers.

Page 156: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

135

The Model

This investigation has lead to the development of a model of targeting Notch both

at production (transcription of Notch) and activation (formation of the active intracellular

Notch) (Figure 47):

1. Production

It was determined that PEA3 transcriptionally activates Notch-1 independently

where as PEA3 partners with c-JUN on Notch-4 (Figure 22 and Figure 31, 32). The

results also suggest that Fra-1 is an activator and c-FOS a repressor of Notch-4

transcription (Figure 35). This stage involves direct regulation at the level of the Notch

gene. Many events including transcriptional complex formation, active transcriptional

hubs (areas of three dimensional gene locus looping as seen in the CFTR gene (Ott et al.,

2009)), and chromatin remodeling (Zhao et al., 2009) are brought to fruition to

successfully regulate the production of the mature Notch receptor. The dynamic

interplay of these factors identified here, such as AP-1 and PEA3 as well as other

potential players unidentified including histone acetyltransferases (Liu et al., 2004;

Matsui et al., 2006), SP-1 (determined by promoter scanning using NCBI Entrez Gene

bank), and/or basic transcriptional machinery (Firlej et al., 2005; Hesselbrock et al.,

2005) are working to tightly regulate Notch gene transcription. This transcriptional stage

provides the first area for potential targeted therapy, namely inhibition of PEA3, to

abrogate Notch production. Once cells acquire the appropriate cellular stimuli, Notch is

Page 157: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

136

CoR

CoA

NIC

MAML-1

NCoRSMRT

ENDOCYTOSIS

Gamma-SECRETASE

PEN2

NicastrinPresenilin

APH-1

NOTCH-4

TTGTG

Figure 47: Model of the Transcriptional Regulation of Notch-1 and Notch-4 by PEA3. I. Production: PEA3 transcriptionally activates Notch-1 independently where as Notch-4 in a dynamic partnership with AP-1 notably c-JUN, Fra-1 (Activators) and c-FOS (Repressor). Notch is then processed and shuttled to the cellular membrane. II. Activation: Upon engagement of ligand, Notch undergoes two subsequent cleavages. The last cleavage by the gamma-secretase complex is inhibited by GSI, MRK-003, to attenuate Notch signaling and prevent the formation of intracellular Notch (NIC), responsible for displacing co-repressors and recruiting co-activators on CBF-1 driving target genes involved in cell fate, proliferation, and differentiation. Dual targeting of Notch via GSI and PEA3 via siRNA, reduces tumorigenicity by attenuation of Notch signaling at both its production and activation stages resulting in overall growth arrest and increased apoptosis. This combinatory inhibition may provide a more effective targeted therapy for those with triple negative breast cancers, and could provide addition means of targeting in other breast cancer subtypes.

TGACTCAGGAAACAGCTCAGACGTGETSAP-1

~70 bps

Fra-1 c-JUN

c-FOS

NOTCH-1

AATAAATCAGCC AGCACGTGETS

~1.1 kbGGAAGG GGAAGG

NOTCH-1or

NOTCH-4

GROWTH

GSI

siRNA

siRNA

LL ONE

LL TWO

II. ACTIVATION

CE

LIGAND INTERCELLULAR SPACE

TACECE

I. PRODUCTION

PEA3

PEA3

CBF-1

CoA+ + +

NUCLEUS

CYTOSOL

Page 158: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

137

then processed and shuttled to the cellular membrane where it awaits engagement with its

ligand (Kopan and Ilagan, 2009).

2. Activation

Upon engagement of ligand, Notch undergoes two subsequent cleavages. The last

cleavage by the gamma-secretase complex is inhibited by GSI (MRK-003) to attenuate

Notch signaling and prevent the formation of intracellular Notch (NIC), responsible for

displacing co-repressors and recruiting co-activators such as Mastermind on CBF-1

driving target genes involved in cell fate, proliferation, and differentiation (Callahan and

Raafat, 2001). This step is critical for activation of Notch at the post-translational level

and provides a second area for targeted treatment, namely GSI treatment (Weijzen et al.,

2002).

The hypothesis remains that two critical functional areas are open for direct

targeting: transcription and activation. GSI targets the mature receptor (Kopan and

Ilagan, 2004; Weijzen et al., 2002), but production of the receptor is continuing to form.

PEA3 inhibition was only successful at reducing transcripts by 50-80% (Figure 14)

leaving some receptor formation to continue to be produced. If both function areas are

successfully targeted, Notch activation can be attenuated by a GSI and more importantly

the continual production of Notch is dampened by PEA3 siRNA. Therefore, the “global”

abrogation of Notch signaling will reduce the oncogenic effects more successfully than

targeting one or the other independently. Dual targeting of Notch (GSI) and PEA3

(siRNA) reduces tumorigenicity by attenuation of Notch signaling at both production and

Page 159: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

138

activation resulting in overall growth arrest and increased apoptosis. This combinatory

inhibition may provide a more effective targeted therapy for those with triple negative

breast cancers, and could provide additional means of targeting in other breast cancer

subtypes where PEA3 regulates Notch-1 and Notch-4.

Future Investigations

In summary, several areas can be investigated to understand the regulation of

Notch by PEA3. It would be interesting to explore further the notion of PEA3 acting as a

repressor on the Notch-2 promoter. Properly designing primers flanking the Ets sites and

performing subsequent chromatin immunoprecipitations will determine if PEA3 directly

regulates Notch-2. Further confirmation regarding the necessity of certain Ets sites could

be explored by designing promoter luciferase constructs. Some may contain the intact

Ets sites and the other ablated Ets sites via site-directed mutagenesis. Dual luciferase

could determine the importance of each particular site in regulating Notch-2.

PEA3 enrichment on the Notch-1 promoter was independent from AP-1. Other

sites have been identified on the Notch-1 promoter including SP-1 and NFκB. It would

be interesting to understand what additional factors may be regulating Notch-1 in

conjunction with PEA3. Co-immunoprecipitation studies could determine if PEA3 is in

complex with SP-1, NFkB, or p300, for example. Moreover, an in depth analysis of the

Notch-1 promoter may reveal other sites of importance. Continuing this idea, it would be

interesting to determine what other partners work with PEA3 and AP-1, including classic

transcriptional machinery, co-activators, co-repressors, etc.

Page 160: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

139

With the effects observed on cellular proliferation and apoptosis upon dual

inhibition of PEA3 and Notch in vitro, an in vivo study could be formulated. Xenografts

of MDA-MB-231 transfected with PEA3 siRNA could be injected into the mammary fat

pads of nude mice and immediately administered MRK-003 GSI via oral gavage. Tumor

burden and size could be recorded to determine if dual inhibition has the same effects on

tumor proliferation in vivo.

These are just a few areas that could be explored in the future to continue

understanding the role of PEA3 on Notch regulation. What is clear from these studies is

that PEA3 is a transcriptional activator of Notch-1 and Notch-4 in MDA-MB-231 cells,

an example of triple-negative breast cancer. PEA3 inhibition could help MDA-MB-231

cells respond better to a gamma-secretase inhibitor in combination as indicated by these

studies (Figures 38–42). Dual inhibition reduces both anchorage dependent and

independent proliferation (Figure 38, 41, 42) and increases apoptosis (Figure 39). PEA3

may be a great candidate in addition with Notch inhibition for the targeted treatment of

triple-negative breast cancers.

Page 161: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

CHAPTER VI

CLOSING REMARKS

As of yet, there is no known FDA approved, targeted therapy for women

diagnosed with triple-negative breast cancer. They frequently present with metastatic

tumors and have the worst overall prognosis. These studies provide the first evidence of

transcriptional regulation of Notch-1 and Notch-4 by PEA3 in triple-negative and other

breast cancer subtypes. It was demonstrated that Notch-4 gene regulation is in part due to

the presence of AP-1, specifically c-JUN and Fra-1, and PEA3 depending on cellular

context. PEA3 regulates the Notch-1 promoter, but the additional factors that aid in the

regulation are still being investigated. Further evidence reveals that PEA3 inhibition

helps sensitize MDA-MB-231 cells to a gamma-secretase inhibitor, showing promise in

significantly reducing both anchorage dependent and independent growth and increasing

apoptosis. Thus, PEA3 emerges as potential target of inhibition in combination with

inhibiting Notch activity to provide a novel and combined targeted therapy for triple-

negative cancer.

The building blocks of human life, their innate blueprint, rests within the genome.

The genome is a dynamic world of regulation involving opening and closing of DNA

segments, chromatin remolding, histone modifications, and epigenetics. This research

provides insight into gene regulation that can be applied to other genes, systems, and

140

Page 162: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

141

disease states. Complex interplay of transcription factors and cellular stimuli when

altered significantly can lead to the disruption of homeostasis and the development of

human disease. PEA3, AP-1, and NOTCH are just a few proteins among myriads of

human disease. PEA3, AP-1, and NOTCH are just a few proteins among myriads of

others aberrantly expressed during disease. Many thousands of proteins are known to be

active within the cell and their genomic regulation is just as vast. This research aims to

provide another stepping stone to understand gene regulation and breast cancer disease

state. Eradication of the breast cancer disease begins here –at the transcriptome.

Page 163: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

BIBLIOGRAPHY Agarwal SK, Guru SC, Heppner C, Erdos MR, Collins RM, Park SY, Saggar S,

Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ, Burns AL. 1999. Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription. Cell 96(1):143-152.

Agarwal SK, Novotny EA, Crabtree JS, Weitzman JB, Yaniv M, Burns AL,

Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ. 2003. Transcription factor JunD, deprived of menin, switches from growth suppressor to growth promoter. Proc Natl Acad Sci U S A 100(19):10770-10775.

Albanese C, Johnson J, Watanabe G, Eklund N, Vu D, Arnold A, Pestell RG. 1995.

Transforming p21ras mutants and c-Ets-2 activate the cyclin D1 promoter through distinguishable regions. J Biol Chem 270(40):23589-23597.

Alimirah F, Panchanathan R, Davis FJ, Chen J, Choubey D. 2007. Restoration of p53

expression in human cancer cell lines upregulates the expression of Notch1: implications for cancer cell fate determination after genotoxic stress. Neoplasia 9(5):427-434.

Amar S, Moreno-Aspitia A, Perez EA. 2008. Issues and controversies in the treatment of

HER2 positive metastatic breast cancer. Breast Cancer Res Treat 109(1):1-7. Ameyar-Zazoua M, Wisniewska MB, Bakiri L, Wagner EF, Yaniv M, Weitzman JB.

2005. AP-1 dimers regulate transcription of the p14/p19ARF tumor suppressor gene. Oncogene 24(14):2298-2306.

Angel P, Karin M. 1991. The role of Jun, Fos and the AP-1 complex in cell-proliferation

and transformation. Biochim Biophys Acta 1072(2-3):129-157. Apelqvist A, Li H, Sommer L, Beatus P, Anderson DJ, Honjo T, Hrabe de Angelis M,

Lendahl U, Edlund H. 1999. Notch signalling controls pancreatic cell differentiation. Nature 400(6747):877-881.

Baan B, Pardali E, Ten Dijke P, van Dam H. 2010. In situ proximity ligation detection of

cJun/AP-1 dimers reveals increased levels of cJun/Fra1 complexes in aggressive breast cancer cell lines in vitro and in vivo. Mol Cell Proteomics.

142

Page 164: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

143 Baert JL, Monte D, Musgrove EA, Albagli O, Sutherland RL, de Launoit Y. 1997.

Expression of the PEA3 group of ETS-related transcription factors in human breast-cancer cells. Int J Cancer 70(5):590-597.

Bakin AV, Curran T. 1999. Role of DNA 5-methylcytosine transferase in cell

transformation by fos. Science 283(5400):387-390. Bakiri L, Lallemand D, Bossy-Wetzel E, Yaniv M. 2000. Cell cycle-dependent variations

in c-Jun and JunB phosphorylation: a role in the control of cyclin D1 expression. Embo J 19(9):2056-2068.

Bamberger AM, Methner C, Lisboa BW, Stadtler C, Schulte HM, Loning T, Milde-

Langosch K. 1999. Expression pattern of the AP-1 family in breast cancer: association of fosB expression with a well-differentiated, receptor-positive tumor phenotype. Int J Cancer 84(5):533-538.

Bannister AJ, Kouzarides T. 1995. CBP-induced stimulation of c-Fos activity is

abrogated by E1A. Embo J 14(19):4758-4762. Bartsch R, Ziebermayr R, Zielinski CC, Steger GG. 2010. Triple-negative breast cancer.

Wien Med Wochenschr 160(7-8):174-181. Beckett D. 2004. Functional switches in transcription regulation; molecular mimicry and

plasticity in protein-protein interactions. Biochemistry 43(25):7983-7991. Bedard PL, Cardoso F, Piccart-Gebhart MJ. 2009. Stemming resistance to HER-2

targeted therapy. J Mammary Gland Biol Neoplasia 14(1):55-66. Belguise K, Kersual N, Galtier F, Chalbos D. 2005. FRA-1 expression level regulates

proliferation and invasiveness of breast cancer cells. Oncogene 24(8):1434-1444. Benbow U, Brinckerhoff CE. 1997. The AP-1 site and MMP gene regulation: what is all

the fuss about? Matrix Biol 15(8-9):519-526. Benz CC, O'Hagan RC, Richter B, Scott GK, Chang CH, Xiong X, Chew K, Ljung BM,

Edgerton S, Thor A, Hassell JA. 1997. HER2/Neu and the Ets transcription activator PEA3 are coordinately upregulated in human breast cancer. Oncogene 15(13):1513-1525.

Berdnik D, Torok T, Gonzalez-Gaitan M, Knoblich JA. 2002. The endocytic protein

alpha-Adaptin is required for numb-mediated asymmetric cell division in Drosophila. Dev Cell 3(2):221-231.

Page 165: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

144 Berry NB, Fan M, Nephew KP. 2008. Estrogen receptor-alpha hinge-region lysines 302

and 303 regulate receptor degradation by the proteasome. Mol Endocrinol 22(7):1535-1551.

Blaumueller CM, Artavanis-Tsakonas S. 1997. Comparative aspects of Notch signaling

in lower and higher eukaryotes. Perspect Dev Neurobiol 4(4):325-343. Bojovic BB, Hassell JA. 2001. The PEA3 Ets transcription factor comprises multiple

domains that regulate transactivation and DNA binding. J Biol Chem 276(6):4509-4521.

Bojovic BB, Hassell JA. 2008. The transactivation function of the Pea3 subfamily Ets

transcription factors is regulated by sumoylation. DNA Cell Biol 27(6):289-305. Bossis G, Ferrara P, Acquaviva C, Jariel-Encontre I, Piechaczyk M. 2003. c-Fos proto-

oncoprotein is degraded by the proteasome independently of its own ubiquitinylation in vivo. Mol Cell Biol 23(20):7425-7436.

Brou C, Logeat F, Gupta N, Bessia C, LeBail O, Doedens JR, Cumano A, Roux P, Black

RA, Israel A. 2000. A novel proteolytic cleavage involved in Notch signaling: the role of the disintegrin-metalloprotease TACE. Mol Cell 5(2):207-216.

Brown PH, Alani R, Preis LH, Szabo E, Birrer MJ. 1993. Suppression of oncogene-

induced transformation by a deletion mutant of c-jun. Oncogene 8(4):877-886. Brown PH, Chen TK, Birrer MJ. 1994. Mechanism of action of a dominant-negative

mutant of c-Jun. Oncogene 9(3):791-799. Brown PH, Kim SH, Wise SC, Sabichi AL, Birrer MJ. 1996. Dominant-negative mutants

of cJun inhibit AP-1 activity through multiple mechanisms and with different potencies. Cell Growth Differ 7(8):1013-1021.

Buzdar AU. 2008. Fulvestrant--a novel estrogen receptor antagonist for the treatment of

advanced breast cancer. Drugs Today (Barc) 44(9):679-692. Buzdar AU, Baum M, Cuzick J. 2006. Letrozole or tamoxifen in early breast cancer. N

Engl J Med 354(14):1528-1530; author reply 1528-1530. Cai J, Lee J, Kopan R, Ma L. 2009. Genetic interplays between Msx2 and Foxn1 are

required for Notch1 expression and hair shaft differentiation. Dev Biol 326(2):420-430.

Callahan R, Raafat A. 2001. Notch signaling in mammary gland tumorigenesis. J

Mammary Gland Biol Neoplasia 6(1):23-36.

Page 166: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

145 Carrasco D, Bravo R. 1995. Tissue-specific expression of the fos-related transcription

factor fra-2 during mouse development. Oncogene 10(6):1069-1079. Carter CL, Allen C, Henson DE. 1989. Relation of tumor size, lymph node status, and

survival in 24,740 breast cancer cases. Cancer 63(1):181-187. Carter P, Presta L, Gorman CM, Ridgway JB, Henner D, Wong WL, Rowland AM, Kotts

C, Carver ME, Shepard HM. 1992. Humanization of an anti-p185HER2 antibody for human cancer therapy. Proc Natl Acad Sci U S A 89(10):4285-4289.

Chan DA, Giaccia AJ. 2008. Targeting cancer cells by synthetic lethality: autophagy and

VHL in cancer therapeutics. Cell Cycle 7(19):2987-2990. Cheang MC, van de Rijn M, Nielsen TO. 2008. Gene expression profiling of breast

cancer. Annu Rev Pathol 3:67-97. Chen RH, Juo PC, Curran T, Blenis J. 1996. Phosphorylation of c-Fos at the C-terminus

enhances its transforming activity. Oncogene 12(7):1493-1502. Cheng P, Zlobin A, Volgina V, Gottipati S, Osborne B, Simel EJ, Miele L, Gabrilovich

DI. 2001. Notch-1 regulates NF-kappaB activity in hemopoietic progenitor cells. J Immunol 167(8):4458-4467.

Chotteau-Lelievre A, Desbiens X, Pelczar H, Defossez PA, de Launoit Y. 1997.

Differential expression patterns of the PEA3 group transcription factors through murine embryonic development. Oncogene 15(8):937-952.

Chotteau-Lelievre A, Montesano R, Soriano J, Soulie P, Desbiens X, de Launoit Y. 2003.

PEA3 transcription factors are expressed in tissues undergoing branching morphogenesis and promote formation of duct-like structures by mammary epithelial cells in vitro. Dev Biol 259(2):241-257.

Chou J, Provot S, Werb Z. 2010. GATA3 in development and cancer differentiation: cells

GATA have it! J Cell Physiol 222(1):42-49. Clapier CR, Cairns BR. 2009. The biology of chromatin remodeling complexes. Annu

Rev Biochem 78:273-304. Clementz AG, Osipo C. 2009. Notch versus the proteasome: what is the target of gamma-

secretase inhibitor-I? Breast Cancer Res 11(5):110. Coates AS, Keshaviah A, Thurlimann B, Mouridsen H, Mauriac L, Forbes JF, Paridaens

R, Castiglione-Gertsch M, Gelber RD, Colleoni M, Lang I, Del Mastro L, Smith I,

Page 167: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

146 Chirgwin J, Nogaret JM, Pienkowski T, Wardley A, Jakobsen EH, Price KN, Goldhirsch A. 2007. Five years of letrozole compared with tamoxifen as initial adjuvant therapy for postmenopausal women with endocrine-responsive early breast cancer: update of study BIG 1-98. J Clin Oncol 25(5):486-492.

Colleoni M, Cole BF, Viale G, Regan MM, Price KN, Maiorano E, Mastropasqua MG,

Crivellari D, Gelber RD, Goldhirsch A, Coates AS, Gusterson BA. 2010. Classical cyclophosphamide, methotrexate, and fluorouracil chemotherapy is more effective in triple-negative, node-negative breast cancer: results from two randomized trials of adjuvant chemoendocrine therapy for node-negative breast cancer. J Clin Oncol 28(18):2966-2973.

Coutte L, Monte D, Baert J, de Launoit Y. 1999. Genomic organization of the human

e1af gene,a member of Ets transcription factors. Gene 240(1):201-207. Curran T, Franza BR, Jr. 1988. Fos and Jun: the AP-1 connection. Cell 55(3):395-397. Curry CL, Reed LL, Golde TE, Miele L, Nickoloff BJ, Foreman KE. 2005. Gamma

secretase inhibitor blocks Notch activation and induces apoptosis in Kaposi's sarcoma tumor cells. Oncogene 24(42):6333-6344.

Daschner PJ, Ciolino HP, Plouzek CA, Yeh GC. 1999. Increased AP-1 activity in drug

resistant human breast cancer MCF-7 cells. Breast Cancer Res Treat 53(3):229-240.

Davidson B, Goldberg I, Gotlieb WH, Kopolovic J, Risberg B, Ben-Baruch G, Reich R.

2003. Coordinated expression of integrin subunits, matrix metalloproteinases (MMP), angiogenic genes and Ets transcription factors in advanced-stage ovarian carcinoma: a possible activation pathway? Cancer Metastasis Rev 22(1):103-115.

Dean-Colomb W, Esteva FJ. 2008. Her2-positive breast cancer: herceptin and beyond.

Eur J Cancer 44(18):2806-2812. Del Pino P, Munoz-Javier A, Vlaskou D, Rivera Gil P, Plank C, Parak WJ. 2010. Gene

Silencing Mediated by Magnetic Lipospheres Tagged with Small Interfering RNA. Nano Lett.

Deng T, Karin M. 1993. JunB differs from c-Jun in its DNA-binding and dimerization

domains, and represses c-Jun by formation of inactive heterodimers. Genes Dev 7(3):479-490.

Deng T, Karin M. 1994. c-Fos transcriptional activity stimulated by H-Ras-activated

protein kinase distinct from JNK and ERK. Nature 371(6493):171-175.

Page 168: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

147 Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA,

Rawlinson E, Sun P, Narod SA. 2007. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 13(15 Pt 1):4429-4434.

Dickson BC, Mulligan AM, Zhang H, Lockwood G, O'Malley FP, Egan SE, Reedijk M.

2007. High-level JAG1 mRNA and protein predict poor outcome in breast cancer. Mod Pathol 20(6):685-693.

Dievart A, Beaulieu N, Jolicoeur P. 1999. Involvement of Notch1 in the development of

mouse mammary tumors. Oncogene 18(44):5973-5981. Dirbas FM. 2009. Accelerated partial breast irradiation: where do we stand? J Natl

Compr Canc Netw 7(2):215-225. Dunwoodie SL, Henrique D, Harrison SM, Beddington RS. 1997. Mouse Dll3: a novel

divergent Delta gene which may complement the function of other Delta homologues during early pattern formation in the mouse embryo. Development 124(16):3065-3076.

Eferl R, Wagner EF. 2003. AP-1: a double-edged sword in tumorigenesis. Nat Rev

Cancer 3(11):859-868. El-Tanani M, Platt-Higgins A, Rudland PS, Campbell FC. 2004. Ets gene PEA3

cooperates with beta-catenin-Lef-1 and c-Jun in regulation of osteopontin transcription. J Biol Chem 279(20):20794-20806.

Eliasz S, Liang S, Chen Y, De Marco MA, Machek O, Skucha S, Miele L, Bocchetta M.

2010. Notch-1 stimulates survival of lung adenocarcinoma cells during hypoxia by activating the IGF-1R pathway. Oncogene 29(17):2488-2498.

Evans CP, Stapp EC, Dall'Era MA, Juarez J, Yang JC. 2001. Regulation of u-PA gene

expression in human prostate cancer. Int J Cancer 94(3):390-395. Farmer P, Bonnefoi H, Becette V, Tubiana-Hulin M, Fumoleau P, Larsimont D,

Macgrogan G, Bergh J, Cameron D, Goldstein D, Duss S, Nicoulaz AL, Brisken C, Fiche M, Delorenzi M, Iggo R. 2005. Identification of molecular apocrine breast tumours by microarray analysis. Oncogene 24(29):4660-4671.

Firlej V, Bocquet B, Desbiens X, de Launoit Y, Chotteau-Lelievre A. 2005. Pea3

transcription factor cooperates with USF-1 in regulation of the murine bax transcription without binding to an Ets-binding site. J Biol Chem 280(2):887-898.

Page 169: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

148 Fischer A, Schumacher N, Maier M, Sendtner M, Gessler M. 2004. The Notch target

genes Hey1 and Hey2 are required for embryonic vascular development. Genes Dev 18(8):901-911.

Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, Fisher ER, Jeong JH,

Wolmark N. 2002a. Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347(16):1233-1241.

Fisher B, Jeong JH, Anderson S, Bryant J, Fisher ER, Wolmark N. 2002b. Twenty-five-

year follow-up of a randomized trial comparing radical mastectomy, total mastectomy, and total mastectomy followed by irradiation. N Engl J Med 347(8):567-575.

Fitzsimmons D, Hodsdon W, Wheat W, Maira SM, Wasylyk B, Hagman J. 1996. Pax-5

(BSAP) recruits Ets proto-oncogene family proteins to form functional ternary complexes on a B-cell-specific promoter. Genes Dev 10(17):2198-2211.

Foltz DR, Santiago MC, Berechid BE, Nye JS. 2002. Glycogen synthase kinase-3beta

modulates notch signaling and stability. Curr Biol 12(12):1006-1011. Freund A, Jolivel V, Durand S, Kersual N, Chalbos D, Chavey C, Vignon F, Lazennec G.

2004. Mechanisms underlying differential expression of interleukin-8 in breast cancer cells. Oncogene 23(36):6105-6114.

Fulford LG, Easton DF, Reis-Filho JS, Sofronis A, Gillett CE, Lakhani SR, Hanby A.

2006. Specific morphological features predictive for the basal phenotype in grade 3 invasive ductal carcinoma of breast. Histopathology 49(1):22-34.

Fung TT, Hu FB, McCullough ML, Newby PK, Willett WC, Holmes MD. 2006. Diet

quality is associated with the risk of estrogen receptor-negative breast cancer in postmenopausal women. J Nutr 136(2):466-472.

Galea MH, Blamey RW, Elston CE, Ellis IO. 1992. The Nottingham Prognostic Index in

primary breast cancer. Breast Cancer Res Treat 22(3):207-219. Gerald D, Berra E, Frapart YM, Chan DA, Giaccia AJ, Mansuy D, Pouyssegur J, Yaniv

M, Mechta-Grigoriou F. 2004. JunD reduces tumor angiogenesis by protecting cells from oxidative stress. Cell 118(6):781-794.

Graves BJ, Petersen JM. 1998. Specificity within the ets family of transcription factors.

Adv Cancer Res 75:1-55.

Page 170: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

149 Guo B, Sharrocks AD. 2009. Extracellular signal-regulated kinase mitogen-activated

protein kinase signaling initiates a dynamic interplay between sumoylation and ubiquitination to regulate the activity of the transcriptional activator PEA3. Mol Cell Biol 29(11):3204-3218.

Haines N, Irvine KD. 2003. Glycosylation regulates Notch signalling. Nat Rev Mol Cell

Biol 4(10):786-797. Harrison H, Farnie G, Howell SJ, Rock RE, Stylianou S, Brennan KR, Bundred NJ,

Clarke RB. 2010. Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Cancer Res 70(2):709-718.

Hattori K, Angel P, Le Beau MM, Karin M. 1988. Structure and chromosomal

localization of the functional intronless human JUN protooncogene. Proc Natl Acad Sci U S A 85(23):9148-9152.

Hennessy BT, Gonzalez-Angulo AM, Stemke-Hale K, Gilcrease MZ, Krishnamurthy S,

Lee JS, Fridlyand J, Sahin A, Agarwal R, Joy C, Liu W, Stivers D, Baggerly K, Carey M, Lluch A, Monteagudo C, He X, Weigman V, Fan C, Palazzo J, Hortobagyi GN, Nolden LK, Wang NJ, Valero V, Gray JW, Perou CM, Mills GB. 2009. Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res 69(10):4116-4124.

Herber B, Truss M, Beato M, Muller R. 1994. Inducible regulatory elements in the

human cyclin D1 promoter. Oncogene 9(7):2105-2107. Hesselbrock DR, Kurpios N, Hassell JA, Watson MA, Fleming TP. 2005. PEA3, AP-1,

and a unique repetitive sequence all are involved in transcriptional regulation of the breast cancer-associated gene, mammaglobin. Breast Cancer Res Treat 89(3):289-296.

Hida K, Shindoh M, Yoshida K, Kudoh A, Furaoka K, Kohgo T, Fujinaga K, Totsuka Y.

1997. Expression of E1AF, an ets-family transcription factor, is correlated with the invasive phenotype of oral squamous cell carcinoma. Oral Oncol 33(6):426-430.

Higashino F, Yoshida K, Fujinaga Y, Kamio K, Fujinaga K. 1993. Isolation of a cDNA

encoding the adenovirus E1A enhancer binding protein: a new human member of the ets oncogene family. Nucleic Acids Res 21(3):547-553.

Hind D, Ward S, De Nigris E, Simpson E, Carroll C, Wyld L. 2007. Hormonal therapies

for early breast cancer: systematic review and economic evaluation. Health Technol Assess 11(26):iii-iv, ix-xi, 1-134.

Page 171: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

150 Hortobagyi GN. 1998. Treatment of breast cancer. N Engl J Med 339(14):974-984. Howell A. 2006. Pure oestrogen antagonists for the treatment of advanced breast cancer.

Endocr Relat Cancer 13(3):689-706. Hsieh JJ, Zhou S, Chen L, Young DB, Hayward SD. 1999. CIR, a corepressor linking the

DNA binding factor CBF1 to the histone deacetylase complex. Proc Natl Acad Sci U S A 96(1):23-28.

Hu C, Dievart A, Lupien M, Calvo E, Tremblay G, Jolicoeur P. 2006. Overexpression of activated murine Notch1 and Notch3 in transgenic mice blocks mammary gland development and induces mammary tumors. Am J Pathol 168(3):973-990.

Hu E, Mueller E, Oliviero S, Papaioannou VE, Johnson R, Spiegelman BM. 1994.

Targeted disruption of the c-fos gene demonstrates c-fos-dependent and -independent pathways for gene expression stimulated by growth factors or oncogenes. Embo J 13(13):3094-3103.

Iso T, Kedes L, Hamamori Y. 2003. HES and HERP families: multiple effectors of the

Notch signaling pathway. J Cell Physiol 194(3):237-255. Jones C, Mackay A, Grigoriadis A, Cossu A, Reis-Filho JS, Fulford L, Dexter T, Davies

S, Bulmer K, Ford E, Parry S, Budroni M, Palmieri G, Neville AM, O'Hare MJ, Lakhani SR. 2004. Expression profiling of purified normal human luminal and myoepithelial breast cells: identification of novel prognostic markers for breast cancer. Cancer Res 64(9):3037-3045.

Jordan VC, Haldemann B, Allen KE. 1981. Geometric isomers of substituted

triphenylethylenes and antiestrogen action. Endocrinology 108(4):1353-1361. Kallunki T, Deng T, Hibi M, Karin M. 1996. c-Jun can recruit JNK to phosphorylate

dimerization partners via specific docking interactions. Cell 87(5):929-939. Kao J, Salari K, Bocanegra M, Choi YL, Girard L, Gandhi J, Kwei KA, Hernandez-

Boussard T, Wang P, Gazdar AF, Minna JD, Pollack JR. 2009. Molecular profiling of breast cancer cell lines defines relevant tumor models and provides a resource for cancer gene discovery. PLoS One 4(7):e6146.

Kasibhatla S, Brunner T, Genestier L, Echeverri F, Mahboubi A, Green DR. 1998. DNA

damaging agents induce expression of Fas ligand and subsequent apoptosis in T lymphocytes via the activation of NF-kappa B and AP-1. Mol Cell 1(4):543-551.

Kataoka K, Noda M, Nishizawa M. 1996. Transactivation activity of Maf nuclear

oncoprotein is modulated by Jun, Fos and small Maf proteins. Oncogene 12(1):53-62.

Page 172: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

151 Kaya M, Yoshida K, Higashino F, Mitaka T, Ishii S, Fujinaga K. 1996. A single ets-

related transcription factor, E1AF, confers invasive phenotype on human cancer cells. Oncogene 12(2):221-227.

Kennecke H, Yerushalmi R, Woods R, Cheang MC, Voduc D, Speers CH, Nielsen TO,

Gelmon K. 2010. Metastatic behavior of breast cancer subtypes. J Clin Oncol 28(20):3271-3277.

Klueg KM, Parody TR, Muskavitch MA. 1998. Complex proteolytic processing acts on Delta, a transmembrane ligand for Notch, during Drosophila development. Mol Biol Cell 9(7):1709-1723.

Kopan R, Ilagan MX. 2004. Gamma-secretase: proteasome of the membrane? Nat Rev

Mol Cell Biol 5(6):499-504. Kopan R, Ilagan MX. 2009. The canonical Notch signaling pathway: unfolding the

activation mechanism. Cell 137(2):216-233. Korkaya H, Wicha MS. 2007. Selective targeting of cancer stem cells: a new concept in

cancer therapeutics. BioDrugs 21(5):299-310. Korkaya H, Wicha MS. 2009. HER-2, notch, and breast cancer stem cells: targeting an

axis of evil. Clin Cancer Res 15(6):1845-1847. Kurpios NA, MacNeil L, Shepherd TG, Gludish DW, Giacomelli AO, Hassell JA. 2009.

The Pea3 Ets transcription factor regulates differentiation of multipotent progenitor cells during mammary gland development. Dev Biol 325(1):106-121.

Kurpios NA, Sabolic NA, Shepherd TG, Fidalgo GM, Hassell JA. 2003. Function of

PEA3 Ets transcription factors in mammary gland development and oncogenesis. J Mammary Gland Biol Neoplasia 8(2):177-190.

Kustikova O, Kramerov D, Grigorian M, Berezin V, Bock E, Lukanidin E, Tulchinsky E.

1998. Fra-1 induces morphological transformation and increases in vitro invasiveness and motility of epithelioid adenocarcinoma cells. Mol Cell Biol 18(12):7095-7105.

Lanford PJ, Lan Y, Jiang R, Lindsell C, Weinmaster G, Gridley T, Kelley MW. 1999.

Notch signalling pathway mediates hair cell development in mammalian cochlea. Nat Genet 21(3):289-292.

Langerod A, Zhao H, Borgan O, Nesland JM, Bukholm IR, Ikdahl T, Karesen R,

Borresen-Dale AL, Jeffrey SS. 2007. TP53 mutation status and gene expression profiles are powerful prognostic markers of breast cancer. Breast Cancer Res 9(3):R30.

Page 173: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

152 Laudet V, Hanni C, Stehelin D, Duterque-Coquillaud M. 1999. Molecular phylogeny of

the ETS gene family. Oncogene 18(6):1351-1359. Le Borgne R, Bardin A, Schweisguth F. 2005. The roles of receptor and ligand

endocytosis in regulating Notch signaling. Development 132(8):1751-1762. Lee CW, Raskett CM, Prudovsky I, Altieri DC. 2008. Molecular dependence of estrogen

receptor-negative breast cancer on a notch-survivin signaling axis. Cancer Res 68(13):5273-5281.

Lewis HD, Leveridge M, Strack PR, Haldon CD, O'Neil J, Kim H, Madin A, Hannam JC,

Look AT, Kohl N, Draetta G, Harrison T, Kerby JA, Shearman MS, Beher D. 2007. Apoptosis in T cell acute lymphoblastic leukemia cells after cell cycle arrest induced by pharmacological inhibition of notch signaling. Chem Biol 14(2):209-219.

Lindsell CE, Shawber CJ, Boulter J, Weinmaster G. 1995. Jagged: a mammalian ligand

that activates Notch1. Cell 80(6):909-917. Liu Y, Borchert GL, Phang JM. 2004. Polyoma enhancer activator 3, an ets transcription

factor, mediates the induction of cyclooxygenase-2 by nitric oxide in colorectal cancer cells. J Biol Chem 279(18):18694-18700.

Liu Y, Ludes-Meyers J, Zhang Y, Munoz-Medellin D, Kim HT, Lu C, Ge G, Schiff R,

Hilsenbeck SG, Osborne CK, Brown PH. 2002. Inhibition of AP-1 transcription factor causes blockade of multiple signal transduction pathways and inhibits breast cancer growth. Oncogene 21(50):7680-7689.

Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, Moore DT, Perou CM.

2006. Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol 19(2):264-271.

Maier MM, Gessler M. 2000. Comparative analysis of the human and mouse Hey1

promoter: Hey genes are new Notch target genes. Biochem Biophys Res Commun 275(2):652-660.

Marconcini L, Marchio S, Morbidelli L, Cartocci E, Albini A, Ziche M, Bussolino F,

Oliviero S. 1999. c-fos-induced growth factor/vascular endothelial growth factor D induces angiogenesis in vivo and in vitro. Proc Natl Acad Sci U S A 96(17):9671-9676.

Marshall SF, Bernstein L, Anton-Culver H, Deapen D, Horn-Ross PL, Mohrenweiser H,

Peel D, Pinder R, Purdie DM, Reynolds P, Stram D, West D, Wright WE, Ziogas

Page 174: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

153 A, Ross RK. 2005. Nonsteroidal anti-inflammatory drug use and breast cancer risk by stage and hormone receptor status. J Natl Cancer Inst 97(11):805-812.

Masuya M, Katayama N, Hoshino N, Nishikawa H, Sakano S, Araki H, Mitani H, Suzuki

H, Miyashita H, Kobayashi K, Nishii K, Minami N, Shiku H. 2002. The soluble Notch ligand, Jagged-1, inhibits proliferation of CD34+ macrophage progenitors. Int J Hematol 75(3):269-276.

Matsui K, Sugimori K, Motomura H, Ejiri N, Tsukada K, Kitajima I. 2006. PEA3 cooperates with c-Jun in regulation of HER2/neu transcription. Oncol Rep 16(1):153-158.

Matthews CP, Colburn NH, Young MR. 2007. AP-1 a target for cancer prevention. Curr

Cancer Drug Targets 7(4):317-324. Maughan KL, Lutterbie MA, Ham PS. 2010. Treatment of breast cancer. Am Fam

Physician 81(11):1339-1346. Mauri D, Polyzos NP, Salanti G, Pavlidis N, Ioannidis JP. 2008. Multiple-treatments

meta-analysis of chemotherapy and targeted therapies in advanced breast cancer. J Natl Cancer Inst 100(24):1780-1791.

McArthur H. 2009. An overview of HER-targeted therapy with lapatinib in breast cancer.

Adv Ther 26(3):263-271. McBride K, Charron F, Lefebvre C, Nemer M. 2003. Interaction with GATA

transcription factors provides a mechanism for cell-specific effects of c-Fos. Oncogene 22(52):8403-8412.

McGill MA, Dho SE, Weinmaster G, McGlade CJ. 2009. Numb regulates post-endocytic

trafficking and degradation of Notch1. J Biol Chem 284(39):26427-26438. McGill MA, McGlade CJ. 2003. Mammalian numb proteins promote Notch1 receptor

ubiquitination and degradation of the Notch1 intracellular domain. J Biol Chem 278(25):23196-23203.

Micalizzi DS, Farabaugh SM, Ford HL. 2010. Epithelial-mesenchymal transition in

cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia 15(2):117-134.

Miele L, Golde T, Osborne B. 2006. Notch signaling in cancer. Curr Mol Med 6(8):905-

918. Milde-Langosch K, Bamberger AM, Methner C, Rieck G, Loning T. 2000. Expression of

cell cycle-regulatory proteins rb, p16/MTS1, p27/KIP1, p21/WAF1, cyclin D1

Page 175: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

154

and cyclin E in breast cancer: correlations with expression of activating protein-1 family members. Int J Cancer 87(4):468-472.

Milde-Langosch K, Roder H, Andritzky B, Aslan B, Hemminger G, Brinkmann A,

Bamberger CM, Loning T, Bamberger AM. 2004. The role of the AP-1 transcription factors c-Fos, FosB, Fra-1 and Fra-2 in the invasion process of mammary carcinomas. Breast Cancer Res Treat 86(2):139-152.

Mirza AN, Mirza NQ, Vlastos G, Singletary SE. 2002. Prognostic factors in node-

negative breast cancer: a review of studies with sample size more than 200 and follow-up more than 5 years. Ann Surg 235(1):10-26.

Moellering RE, Cornejo M, Davis TN, Del Bianco C, Aster JC, Blacklow SC, Kung AL,

Gilliland DG, Verdine GL, Bradner JE. 2009. Direct inhibition of the NOTCH transcription factor complex. Nature 462(7270):182-188.

Monje P, Marinissen MJ, Gutkind JS. 2003. Phosphorylation of the carboxyl-terminal

transactivation domain of c-Fos by extracellular signal-regulated kinase mediates the transcriptional activation of AP-1 and cellular transformation induced by platelet-derived growth factor. Mol Cell Biol 23(19):7030-7043.

Morgan TH. 1917. The theory of the gene. American Naturalist 51(609):513-544. Morris GJ, Naidu S, Topham AK, Guiles F, Xu Y, McCue P, Schwartz GF, Park PK,

Rosenberg AL, Brill K, Mitchell EP. 2007. Differences in breast carcinoma characteristics in newly diagnosed African-American and Caucasian patients: a single-institution compilation compared with the National Cancer Institute's Surveillance, Epidemiology, and End Results database. Cancer 110(4):876-884.

Mouridsen HT. 1990. New cytotoxic drugs in treatment of breast cancer. Acta Oncol

29(3):343-347. Nickoloff BJ, Qin JZ, Chaturvedi V, Denning MF, Bonish B, Miele L. 2002. Jagged-1

mediated activation of notch signaling induces complete maturation of human keratinocytes through NF-kappaB and PPARgamma. Cell Death Differ 9(8):842-855.

Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T,

Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM. 2004. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10(16):5367-5374.

Page 176: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

155 Nishida T, Terashima M, Fukami K, Yamada Y. 2007. PIASy controls ubiquitination-

dependent proteasomal degradation of Ets-1. Biochem J 405(3):481-488. O'Mahony D, Bishop MR. 2006. Monoclonal antibody therapy. Front Biosci 11:1620-

1635. O'Neill CF, Urs S, Cinelli C, Lincoln A, Nadeau RJ, Leon R, Toher J, Mouta-Bellum C,

Friesel RE, Liaw L. 2007. Notch2 signaling induces apoptosis and inhibits human MDA-MB-231 xenograft growth. Am J Pathol 171(3):1023-1036.

Oikawa T, Yamada T. 2003. Molecular biology of the Ets family of transcription factors. Gene 303:11-34.

Okajima T, Irvine KD. 2002. Regulation of notch signaling by o-linked fucose. Cell

111(6):893-904. Olive M, Krylov D, Echlin DR, Gardner K, Taparowsky E, Vinson C. 1997. A dominant

negative to activation protein-1 (AP1) that abolishes DNA binding and inhibits oncogenesis. J Biol Chem 272(30):18586-18594.

Ott CJ, Blackledge NP, Kerschner JL, Leir SH, Crawford GE, Cotton CU, Harris A. 2009.

Intronic enhancers coordinate epithelial-specific looping of the active CFTR locus. Proc Natl Acad Sci U S A 106(47):19934-19939.

Pajerowski AG, Nguyen C, Aghajanian H, Shapiro MJ, Shapiro VS. 2009. NKAP is a

transcriptional repressor of notch signaling and is required for T cell development. Immunity 30(5):696-707.

Passegue E, Wagner EF. 2000. JunB suppresses cell proliferation by transcriptional

activation of p16(INK4a) expression. Embo J 19(12):2969-2979. Peppercorn J, Perou CM, Carey LA. 2008. Molecular subtypes in breast cancer

evaluation and management: divide and conquer. Cancer Invest 26(1):1-10. Perez A, Barco R, Fernandez I, Price-Schiavi SA, Carraway KL. 2003. PEA3

transactivates the Muc4/sialomucin complex promoter in mammary epithelial and tumor cells. J Biol Chem 278(38):36942-36952.

Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross

DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D. 2000. Molecular portraits of human breast tumours. Nature 406(6797):747-752.

Page 177: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

156 Pfarr CM, Mechta F, Spyrou G, Lallemand D, Carillo S, Yaniv M. 1994. Mouse JunD

negatively regulates fibroblast growth and antagonizes transformation by ras. Cell 76(4):747-760.

Quillard T, Devalliere J, Chatelais M, Coulon F, Seveno C, Romagnoli M, Barille Nion S,

Charreau B. 2009. Notch2 signaling sensitizes endothelial cells to apoptosis by negatively regulating the key protective molecule survivin. PLoS One 4(12):e8244.

Ransone LJ, Visvader J, Wamsley P, Verma IM. 1990. Trans-dominant negative mutants

of Fos and Jun. Proc Natl Acad Sci U S A 87(10):3806-3810. Rauscher FJ, 3rd, Sambucetti LC, Curran T, Distel RJ, Spiegelman BM. 1988. Common

DNA binding site for Fos protein complexes and transcription factor AP-1. Cell 52(3):471-480.

Reedijk M, Odorcic S, Chang L, Zhang H, Miller N, McCready DR, Lockwood G, Egan

SE. 2005. High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res 65(18):8530-8537.

Reis-Filho JS, Pinheiro C, Lambros MB, Milanezi F, Carvalho S, Savage K, Simpson PT,

Jones C, Swift S, Mackay A, Reis RM, Hornick JL, Pereira EM, Baltazar F, Fletcher CD, Ashworth A, Lakhani SR, Schmitt FC. 2006. EGFR amplification and lack of activating mutations in metaplastic breast carcinomas. J Pathol 209(4):445-453.

Reis-Filho JS, Tutt AN. 2008. Triple negative tumours: a critical review. Histopathology

52(1):108-118. Rizzo P, Miao H, D'Souza G, Osipo C, Song LL, Yun J, Zhao H, Mascarenhas J, Wyatt

D, Antico G, Hao L, Yao K, Rajan P, Hicks C, Siziopikou K, Selvaggi S, Bashir A, Bhandari D, Marchese A, Lendahl U, Qin JZ, Tonetti DA, Albain K, Nickoloff BJ, Miele L. 2008. Cross-talk between notch and the estrogen receptor in breast cancer suggests novel therapeutic approaches. Cancer Res 68(13):5226-5235.

Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE, Jr., Davidson NE, Tan-Chiu E,

Martino S, Paik S, Kaufman PA, Swain SM, Pisansky TM, Fehrenbacher L, Kutteh LA, Vogel VG, Visscher DW, Yothers G, Jenkins RB, Brown AM, Dakhil SR, Mamounas EP, Lingle WL, Klein PM, Ingle JN, Wolmark N. 2005. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 353(16):1673-1684.

Page 178: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

157 Ronchini C, Capobianco AJ. 2001. Induction of cyclin D1 transcription and CDK2

activity by Notch(ic): implication for cell cycle disruption in transformation by Notch(ic). Mol Cell Biol 21(17):5925-5934.

Rouzier R, Wagner P, Morandi P, Pusztai L. 2005. Gene expression profiling of primary

breast cancer. Curr Oncol Rep 7(1):38-44. Rowinsky EK, Onetto N, Canetta RM, Arbuck SG. 1992. Taxol: the first of the taxanes,

an important new class of antitumor agents. Semin Oncol 19(6):646-662. Sachdev JC, Jahanzeb M. 2008. Evolution of bevacizumab-based therapy in the

management of breast cancer. Clin Breast Cancer 8(5):402-410. Sansone P, Storci G, Giovannini C, Pandolfi S, Pianetti S, Taffurelli M, Santini D,

Ceccarelli C, Chieco P, Bonafe M. 2007. p66Shc/Notch-3 interplay controls self-renewal and hypoxia survival in human stem/progenitor cells of the mammary gland expanded in vitro as mammospheres. Stem Cells 25(3):807-815.

Sasamura T, Sasaki N, Miyashita F, Nakao S, Ishikawa HO, Ito M, Kitagawa M,

Harigaya K, Spana E, Bilder D, Perrimon N, Matsuno K. 2003. neurotic, a novel maternal neurogenic gene, encodes an O-fucosyltransferase that is essential for Notch-Delta interactions. Development 130(20):4785-4795.

Saxena MT, Schroeter EH, Mumm JS, Kopan R. 2001. Murine notch homologs (N1-4)

undergo presenilin-dependent proteolysis. J Biol Chem 276(43):40268-40273. Schutte J, Minna JD, Birrer MJ. 1989. Deregulated expression of human c-jun transforms

primary rat embryo cells in cooperation with an activated c-Ha-ras gene and transforms rat-1a cells as a single gene. Proc Natl Acad Sci U S A 86(7):2257-2261.

Scott GK, Daniel JC, Xiong X, Maki RA, Kabat D, Benz CC. 1994. Binding of an ETS-

related protein within the DNase I hypersensitive site of the HER2/neu promoter in human breast cancer cells. J Biol Chem 269(31):19848-19858.

Sharrocks AD. 2001. The ETS-domain transcription factor family. Nat Rev Mol Cell Biol

2(11):827-837. Shaulian E, Karin M. 2001. AP-1 in cell proliferation and survival. Oncogene

20(19):2390-2400. Shaulian E, Karin M. 2002. AP-1 as a regulator of cell life and death. Nat Cell Biol

4(5):E131-136.

Page 179: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

158 Shawber C, Boulter J, Lindsell CE, Weinmaster G. 1996. Jagged2: a serrate-like gene

expressed during rat embryogenesis. Dev Biol 180(1):370-376. Shepherd T, Hassell JA. 2001. Role of Ets transcription factors in mammary gland

development and oncogenesis. J Mammary Gland Biol Neoplasia 6(1):129-140. Shepherd TG, Kockeritz L, Szrajber MR, Muller WJ, Hassell JA. 2001. The pea3

subfamily ets genes are required for HER2/Neu-mediated mammary oncogenesis. Curr Biol 11(22):1739-1748.

Shi S, Stanley P. 2003. Protein O-fucosyltransferase 1 is an essential component of Notch

signaling pathways. Proc Natl Acad Sci U S A 100(9):5234-5239. Shore P, Whitmarsh AJ, Bhaskaran R, Davis RJ, Waltho JP, Sharrocks AD. 1996.

Determinants of DNA-binding specificity of ETS-domain transcription factors. Mol Cell Biol 16(7):3338-3349.

Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG,

Udove J, Ullrich A, et al. 1989. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244(4905):707-712.

Sledge GW, Jr. 1992. Cisplatin and platinum analogues in breast cancer. Semin Oncol

19(1 Suppl 2):78-82. Smeal T, Angel P, Meek J, Karin M. 1989. Different requirements for formation of Jun:

Jun and Jun: Fos complexes. Genes Dev 3(12B):2091-2100. Smeal T, Binetruy B, Mercola DA, Birrer M, Karin M. 1991. Oncogenic and

transcriptional cooperation with Ha-Ras requires phosphorylation of c-Jun on serines 63 and 73. Nature 354(6353):494-496.

Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van

de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Eystein Lonning P, Borresen-Dale AL. 2001. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98(19):10869-10874.

Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H,

Pesich R, Geisler S, Demeter J, Perou CM, Lonning PE, Brown PO, Borresen-Dale AL, Botstein D. 2003. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100(14):8418-8423.

Spalding BJ. 2008. Thumbs up for Avastin. Nat Biotechnol 26(4):365.

Page 180: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

159 Staal FJ, Langerak AW. 2008. Signaling pathways involved in the development of T-cell

acute lymphoblastic leukemia. Haematologica 93(4):493-497. Stylianou S, Clarke RB, Brennan K. 2006. Aberrant activation of notch signaling in

human breast cancer. Cancer Res 66(3):1517-1525. Subbaramaiah K, Norton L, Gerald W, Dannenberg AJ. 2002. Cyclooxygenase-2 is

overexpressed in HER-2/neu-positive breast cancer: evidence for involvement of AP-1 and PEA3. J Biol Chem 277(21):18649-18657.

Subramaniam D, Ramalingam S, Houchen CW, Anant S. 2010. Cancer stem cells: a

novel paradigm for cancer prevention and treatment. Mini Rev Med Chem 10(5):359-371.

Sun X, Artavanis-Tsakonas S. 1997. Secreted forms of DELTA and SERRATE define

antagonists of Notch signaling in Drosophila. Development 124(17):3439-3448. Suzuki R, Ye W, Rylander-Rudqvist T, Saji S, Colditz GA, Wolk A. 2005. Alcohol and

postmenopausal breast cancer risk defined by estrogen and progesterone receptor status: a prospective cohort study. J Natl Cancer Inst 97(21):1601-1608.

Swaby RF, Jordan VC. 2008. Low-dose estrogen therapy to reverse acquired

antihormonal resistance in the treatment of breast cancer. Clin Breast Cancer 8(2):124-133.

Takahashi A, Higashino F, Aoyagi M, Yoshida K, Itoh M, Kobayashi M, Totsuka Y,

Kohgo T, Shindoh M. 2005. E1AF degradation by a ubiquitin-proteasome pathway. Biochem Biophys Res Commun 327(2):575-580.

Thepot D, Weitzman JB, Barra J, Segretain D, Stinnakre MG, Babinet C, Yaniv M. 2000.

Targeted disruption of the murine junD gene results in multiple defects in male reproductive function. Development 127(1):143-153.

Tkach V, Tulchinsky E, Lukanidin E, Vinson C, Bock E, Berezin V. 2003. Role of the

Fos family members, c-Fos, Fra-1 and Fra-2, in the regulation of cell motility. Oncogene 22(32):5045-5054.

Trimble MS, Xin JH, Guy CT, Muller WJ, Hassell JA. 1993. PEA3 is overexpressed in

mouse metastatic mammary adenocarcinomas. Oncogene 8(11):3037-3042. Ueyama K, Ikeda K, Sato W, Nakasato N, Horie-Inoue K, Takeda S, Inoue S. 2010.

Knockdown of Efp by DNA-modified small interfering RNA inhibits breast cancer cell proliferation and in vivo tumor growth. Cancer Gene Ther 17(9):624-632.

Page 181: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

160 Usary J, Llaca V, Karaca G, Presswala S, Karaca M, He X, Langerod A, Karesen R, Oh

DS, Dressler LG, Lonning PE, Strausberg RL, Chanock S, Borresen-Dale AL, Perou CM. 2004. Mutation of GATA3 in human breast tumors. Oncogene 23(46):7669-7678.

Uyttendaele H, Marazzi G, Wu G, Yan Q, Sassoon D, Kitajewski J. 1996. Notch4/int-3, a

mammary proto-oncogene, is an endothelial cell-specific mammalian Notch gene. Development 122(7):2251-2259.

van de Rijn M, Perou CM, Tibshirani R, Haas P, Kallioniemi O, Kononen J, Torhorst J,

Sauter G, Zuber M, Kochli OR, Mross F, Dieterich H, Seitz R, Ross D, Botstein D, Brown P. 2002. Expression of cytokeratins 17 and 5 identifies a group of breast carcinomas with poor clinical outcome. Am J Pathol 161(6):1991-1996.

van Es JH, van Gijn ME, Riccio O, van den Born M, Vooijs M, Begthel H, Cozijnsen M,

Robine S, Winton DJ, Radtke F, Clevers H. 2005. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 435(7044):959-963.

Veronesi U, Cascinelli N, Mariani L, Greco M, Saccozzi R, Luini A, Aguilar M,

Marubini E. 2002. Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347(16):1227-1232.

Vleugel MM, Greijer AE, Bos R, van der Wall E, van Diest PJ. 2006. c-Jun activation is

associated with proliferation and angiogenesis in invasive breast cancer. Hum Pathol 37(6):668-674.

Wang W, Struhl G. 2005. Distinct roles for Mind bomb, Neuralized and Epsin in

mediating DSL endocytosis and signaling in Drosophila. Development 132(12):2883-2894.

Wasylyk B, Hagman J, Gutierrez-Hartmann A. 1998. Ets transcription factors: nuclear

effectors of the Ras-MAP-kinase signaling pathway. Trends Biochem Sci 23(6):213-216.

Weigelt B, Kreike B, Reis-Filho JS. 2009. Metaplastic breast carcinomas are basal-like

breast cancers: a genomic profiling analysis. Breast Cancer Res Treat 117(2):273-280.

Weijzen S, Rizzo P, Braid M, Vaishnav R, Jonkheer SM, Zlobin A, Osborne BA,

Gottipati S, Aster JC, Hahn WC, Rudolf M, Siziopikou K, Kast WM, Miele L. 2002. Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells. Nat Med 8(9):979-986.

Page 182: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

161 Weitzman JB, Fiette L, Matsuo K, Yaniv M. 2000. JunD protects cells from p53-

dependent senescence and apoptosis. Mol Cell 6(5):1109-1119. Weng AP, Millholland JM, Yashiro-Ohtani Y, Arcangeli ML, Lau A, Wai C, Del Bianco

C, Rodriguez CG, Sai H, Tobias J, Li Y, Wolfe MS, Shachaf C, Felsher D, Blacklow SC, Pear WS, Aster JC. 2006. c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma. Genes Dev 20(15):2096-2109.

Wisdom R, Johnson RS, Moore C. 1999. c-Jun regulates cell cycle progression and

apoptosis by distinct mechanisms. Embo J 18(1):188-197. Wu G, Lyapina S, Das I, Li J, Gurney M, Pauley A, Chui I, Deshaies RJ, Kitajewski J.

2001. SEL-10 is an inhibitor of notch signaling that targets notch for ubiquitin-mediated protein degradation. Mol Cell Biol 21(21):7403-7415.

Wu J, Bresnick EH. 2007. Glucocorticoid and growth factor synergism requirement for

Notch4 chromatin domain activation. Mol Cell Biol 27(6):2411-2422. Wu J, Iwata F, Grass JA, Osborne CS, Elnitski L, Fraser P, Ohneda O, Yamamoto M,

Bresnick EH. 2005. Molecular determinants of NOTCH4 transcription in vascular endothelium. Mol Cell Biol 25(4):1458-1474.

Wu L, Aster JC, Blacklow SC, Lake R, Artavanis-Tsakonas S, Griffin JD. 2000.

MAML1, a human homologue of Drosophila mastermind, is a transcriptional co-activator for NOTCH receptors. Nat Genet 26(4):484-489.

Xia WY, Lien HC, Wang SC, Pan Y, Sahin A, Kuo YH, Chang KJ, Zhou X, Wang H, Yu

Z, Hortobagyi G, Shi DR, Hung MC. 2006. Expression of PEA3 and lack of correlation between PEA3 and HER-2/neu expression in breast cancer. Breast Cancer Res Treat 98(3):295-301.

Xin JH, Cowie A, Lachance P, Hassell JA. 1992. Molecular cloning and characterization

of PEA3, a new member of the Ets oncogene family that is differentially expressed in mouse embryonic cells. Genes Dev 6(3):481-496.

Xing X, Wang SC, Xia W, Zou Y, Shao R, Kwong KY, Yu Z, Zhang S, Miller S, Huang

L, Hung MC. 2000. The ets protein PEA3 suppresses HER-2/neu overexpression and inhibits tumorigenesis. Nat Med 6(2):189-195.

Yamaguchi N, Oyama T, Ito E, Satoh H, Azuma S, Hayashi M, Shimizu K, Honma R,

Yanagisawa Y, Nishikawa A, Kawamura M, Imai J, Ohwada S, Tatsuta K, Inoue J, Semba K, Watanabe S. 2008. NOTCH3 signaling pathway plays crucial roles in

Page 183: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

162 the proliferation of ErbB2-negative human breast cancer cells. Cancer Res 68(6):1881-1888.

Zajchowski DA, Bartholdi MF, Gong Y, Webster L, Liu HL, Munishkin A, Beauheim C,

Harvey S, Ethier SP, Johnson PH. 2001. Identification of gene expression profiles that predict the aggressive behavior of breast cancer cells. Cancer Res 61(13):5168-5178.

Zanetti-Dallenbach R, Vuaroqueaux V, Wight E, Labuhn M, Singer G, Urban P,

Eppenberger U, Holzgreve W, Eppenberger-Castori S. 2006. Comparison of gene expression profiles in core biopsies and corresponding surgical breast cancer samples. Breast Cancer Res 8(4):R51.

Zavadil J, Cermak L, Soto-Nieves N, Bottinger EP. 2004. Integration of TGF-beta/Smad

and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. Embo J 23(5):1155-1165.

Zhang SM, Hankinson SE, Hunter DJ, Giovannucci EL, Colditz GA, Willett WC. 2005.

Folate intake and risk of breast cancer characterized by hormone receptor status. Cancer Epidemiol Biomarkers Prev 14(8):2004-2008.

Zhao C, Gao H, Liu Y, Papoutsi Z, Jaffrey S, Gustafsson JA, Dahlman-Wright K. 2010.

Genome-wide mapping of estrogen receptor-beta-binding regions reveals extensive cross-talk with transcription factor activator protein-1. Cancer Res 70(12):5174-5183.

Zhao R, Bodnar MS, Spector DL. 2009. Nuclear neighborhoods and gene expression.

Curr Opin Genet Dev 19(2):172-179.

Page 184: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

VITA

The author, Anthony G. Clementz, was born in Aurora, IL on July 14th, 1983. He

is the last child of five children of Peter and Toni Clementz. He has four amazing

siblings all sharing interest in the sciences: Gina, a doctor of dermatology; Peter, an

electrician; Mia, a science teacher; and Andria, a registered nurse.

Anthony graduated as a Cadet First Lieutenant at Marmion Military Academy in

2001. He graduated from Northern Illinois University with high recognition with his

bachelors in Chemistry and Biochemistry in 2005. Anthony received numerous awards

while in school including the American Chemical Society Award in Analytical Chemistry,

Scholastic Excellence Award, Golden Key International Honor Society, National Society

of Collegiate Scholars, CRC Press Frosh Chemistry Achievement Award, and the

National Deans List.

After graduation, he worked for Flavors of North America (FONA) as a flavor scientist

and technician. Anthony entered the doctoral program in the Department of Cell Biology,

Neurobiology, and Anatomy in the Division of Molecular and Cellular Biochemistry at

Loyola University Medical Center in August 2006. He joined Dr. Clodia Osipo’s

laboratory in June 2007 where he began his work on novel crosstalk between Notch and

ErBB2 signaling and transcriptional regulation of Notch receptors. While at Loyola

163

Page 185: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

164

University he has been a graduate school council representative for the Division of

Molecular and Cellular Biochemistry, and has participated in assisting students with

disabilities. Anthony has taught lectures on Hormone Signaling in both graduate student

courses: Molecular Oncogenesis and Signal Transduction. He also was an active

facilitator for the first year medical students in their course: Cell Molecular Biology and

Genetics. Anthony is an active member as an Associate Member of American

Association for Cancer Research and an American Chemical Society affiliate. He has

also been an active judge for the Chicago Public Schools Science Fairs. Anthony’s

research presented here has been submitted and is under review for a novel patent with

Merck Pharmaceuticals.

Anthony G. Clementz also has a musical artistic side for he is an accomplished

pianist and singer having played and sung various musicals and events throughout the

Chicagoland area. He is also a graduate of iO!, formly Improv Olympic!, and Chicago’s

Second City and actively performs improv and sketch comedy.

Anthony G. Clementz has recently accepted a position as adjunct professor in the

Department of Chemistry at DePaul University. Also, following completion of his

dissertation, he will continue his career in transcription regulation as a post-doctoral

researcher at Northwestern University Children’s Memorial Center in the laboratory of

Dr. Ann Harris, professor and director of the Human Molecular Genetics Program. He

plans to investigate the “transcription hubs” of the CFTR gene known to be responsible

for the development of Cystic Fibrosis. He also plans to use his knowledge of

Page 186: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

165

oncogenesis to elucidate the role of Collagen XV as a tumor suppressor in pancreatic

cancer both in vitro and in vivo.

Page 187: Polyomavirus Enhancer Activator 3 (PEA3), a Member of the

DISSERTATION APPROVAL SHEET

The dissertation submitted by Anthony G. Clementz has been read and approved by the

following committee:

Clodia Osipo, Ph.D., Assistant Professor of Pathology Loyola University Chicago Mary Druse-Manteuffel, Ph.D., Professor of Cell Biology, Neurobiology, and Anatomy Division of Molecular and Cellular Biochemistry Loyola University Chicago Richard Schultz, Ph.D., Professor of Cell Biology, Neurobiology, and Anatomy Division of Molecular and Cellular Biochemistry Loyola University Chicago Maurizio Bocchetta, Ph.D., Associate Professor of Pathology Loyola University Chicago Paola Rizzo, Ph.D., Assistant Professor of Pathology Loyola University Chicago

The final copies have been examined by the director of the dissertation and the signature

which appears below verifies the fact that any necessary changes have been incorporated

and that the dissertation is now given final approval by the committee with reference to

content and form.

The dissertation is therefore accepted in partial fulfillment of the requirements for the

degree of Doctor of Philosophy.

_______________________ ____________________________________

Date Director’s Signature