© 2012 can zhang - university of...

154
1 EPIGENETIC REGULATION OF APOPTOSIS DURING DEVELOPMENT AND TUMORIGENESIS IN DROSOPHILA By CAN ZHANG A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2012

Upload: others

Post on 30-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

1

EPIGENETIC REGULATION OF APOPTOSIS DURING DEVELOPMENT AND TUMORIGENESIS IN DROSOPHILA

By

CAN ZHANG

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2012

Page 2: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

2

© 2012 Can Zhang

Page 3: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

3

In loving memory of my father

Page 4: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

4

ACKNOWLEDGMENTS

Completing my Ph.D. degree has probably been the most challenging task I have

undertaken in the first 29 years of my life. I have experienced my best and worst

memories on this remarkable journey. It would not have been possible for me to

complete this doctoral dissertation without the help of the following individuals.

Firstly, I give my utmost gratitude to my father Chuanxin Zhang and my mother

Yongqin Shen, for bringing me into the world and educating me with love and courage. I

could never have reached this far without their parenthood. No such word exists that

can express my deepest love and enormous appreciation to them.

I would like to thank my advisor, Dr. Lei Zhou, for his continuous support of my

Ph.D. study. His patience, inspiration and motivation made my achievements possible.

He has shown me the curiosity, enthusiasm and persistence a real scientist should

carry, and he always encourages me to overcome all obstacles and guides me to

become independent in every aspect of my life.

I also want to express a special thanks to my dissertation committee: Dr. Jorg

Bungert, Dr. Brian Harfe, Dr. James Resnick, and Dr. Naohiro Terada, for their valuable

suggestions, inspiration and continuous support.

My sincere thanks also go to the formal and current members of Zhou laboratory,

especially Dr. Yanping Zhang, Dr. Nianwei Lin, Bo Liu, Guangyao Li, Hannah Wang,

John Pang, Michelle Chung and Denis Titov for their kind and patient help. I also would

like to thank my IDP classmates and best friends, including Weiyi Ni, Shuibin Lin, Chen

Ling, Huiming Xia, Yue Liu and Robert Ng, as well as their families. I am incredibly

fortunate to have met them in Gainesville and become lifetime friends. They have filled

my life with so much happiness and made my research career much less painful.

Page 5: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

5

Last and most importantly, I want to thank my husband, Bing Yao, whose love and

encouragement allowed me to finish this journey. He always has an unwavering faith in

me when I experience tough moments. His company makes my life more delightful and

meaningful. Bing, I hope this work makes you proud.

Page 6: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

6

TABLE OF CONTENTS page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES ............................................................................................................ 9

LIST OF FIGURES ........................................................................................................ 10

LIST OF ABBREVIATIONS ........................................................................................... 12

ABSTRACT ................................................................................................................... 13

CHAPTER

1 BACKGROUND AND INTRODUCTION ................................................................. 15

Regulation of Apoptosis in Drosophila .................................................................... 15 The Apoptosis Regulatory Pathway in Drosophila ............................................ 15

Developmental Cell Death in Drosophila .......................................................... 16 DNA Damage-induced Cell Death in Drosophila .............................................. 17

Developmental Control of Tissue Growth in Drosophila ......................................... 19

Organ Size Control ........................................................................................... 20

Hippo pathway ........................................................................................... 20 EGFR pathway ........................................................................................... 21

Cell Competition ............................................................................................... 22 Extra Sex Combs, Chromatin, and Cellular Identity ................................................ 24

Identification of PcG and TrxG Proteins as Chromatin Regulators ................... 24

Chromatin Structures and Epigenetic Regulations ........................................... 26 PcG/TrxG Mediated Chromatin Modifications .................................................. 27

Epigenetic Regulation of Apoptosis ........................................................................ 29 Dysregulation of PcG Targeting ....................................................................... 29

Epigenetic Silencing of Pro-apoptotic Genes ................................................... 30

2 MONITORING DNA ACCESSIBILITY AND EPIGENETIC STATUS IN INDIVIDUAL CELLS AND LIVE ANIMALS WITH A FLUORESCENT REPORTER ............................................................................................................ 37

Introduction ............................................................................................................. 37

Materials and Methods............................................................................................ 39 Generation of IRER{ubi-DsRed} ....................................................................... 39 Fly Handling ..................................................................................................... 40 DNase I Sensitivity Assay and Chromatin Immunoprecipitation (ChIP) ............ 40

Immunofluorescence ........................................................................................ 41

Flourometric Measurement of DsRed Expression ............................................ 41 Results .................................................................................................................... 42

Page 7: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

7

Inserting the ubi-DsRed Reporter into IRER via Homologous Recombination .............................................................................................. 42

Verifying the Responsiveness of the Reporter to Changes in DNA Accessibility ................................................................................................... 44

Monitoring Cell and Tissue Specific Epigenetic Changes Using IRER{ubi-DsRed} .......................................................................................................... 46

Monitoring Epigenetic Status of IRER at Organismal Level ............................. 49 Discussions ............................................................................................................. 51

Epigenetic Regulation with Cellular Resolution ................................................ 52 Limitations of the Strategy ................................................................................ 53

3 AN EPIGENETICALLY REGULATED INTERGENIC REGION CONTROLS ORGAN SIZE AND MEDIATES DMYC -INDUCED APOPTOSIS IN DROSOPHILA ........................................................................................................ 64

Introduction ............................................................................................................. 64

Materials and Methods............................................................................................ 66 Drosophila Strains and Culture ......................................................................... 66

Clone Induction ................................................................................................ 66 Immunohistochemistry and Microscopy............................................................ 66 Wing Size Measurement .................................................................................. 67

Gene Expression Analysis ................................................................................ 67

Statistics ........................................................................................................... 68

Bioinformatics ................................................................................................... 68 Results .................................................................................................................... 69

IRER Mediates DNA-damage-induced Pro-apoptotic Gene Expression in Post-embryonic Tissues ................................................................................ 69

The cis Regulatory Function of IRER is Required for Tissue Homeostasis and Organ Size Control ................................................................................. 70

IRER is Required for the Induction of Apoptosis following dMyc-induced Overproliferation ............................................................................................ 75

Cells with Relatively Open IRER are More Sensitive to dMyc-induced Cell Death ............................................................................................................ 77

Cells Lacking the cis Regulatory Function of IRER have the Propensity to Overgrow and are Resistant to Stress-induced Cell Death. .......................... 79

The RHG Genomic Regulatory Block Contains Consensus Myc Binding Sites .............................................................................................................. 80

Discussions ............................................................................................................. 81 IRER Serves as the Gatekeeper for Overproliferation-Induced Apoptosis ....... 82 Functional Significance of Epigenetic Regulation of IRER in Development

and in Tumorigenesis .................................................................................... 85

4 A STRESS- RESPONSIVE REGULATORY REGION IS REQUIRED FOR COMPETITION- INDUCED APOPTOSIS ............................................................. 108

Introduction ........................................................................................................... 108 Materials and Methods.......................................................................................... 110

Page 8: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

8

Drosophila Strains and Culture ....................................................................... 110

Clone Induction .............................................................................................. 110 Microscopy and Immunohistochemistry.......................................................... 110

Gene Expression Analysis .............................................................................. 110 Results .................................................................................................................. 111

IRER is Required for Super Competition-induced Elimination of Loser Cells . 111 IRER is Required for Super Competition-induced hid Expression in Loser

Cells ............................................................................................................ 111 IRER is Required for the Elimination of scrib Knockdown Cells ..................... 112

Disruption of Cell Polarity led to de-Repression of IRER ................................ 114 Discussions ........................................................................................................... 115

Myc-induced Cell Autonomous Apoptosis vs. Cell Non-autonomous Apoptosis .................................................................................................... 115

Epigenetic Regulation and Cell Competition .................................................. 116

5 DISCUSSION AND PERSPECTIVE ..................................................................... 127

Coordinated Transcriptional Regulation of the RHG Genes by Locus Control Regions ............................................................................................................. 127

Epigenetic Regulation and Cancer ....................................................................... 128 Is Cell Competition Relevant to Cancer? .............................................................. 130 Unraveling Epigenetic Regulation and Tumorigenesis in Drosophila .................... 132

Perspectives ......................................................................................................... 134

What Mechanism(S) Controls the Epigenetic Regulation of IRER? ............... 135 Which Signaling Pathways Regulate the Function IRER? .............................. 135

What is the Relevance of our Study to Human Research? ............................ 136

LIST OF REFERENCES ............................................................................................. 138

BIOGRAPHICAL SKETCH .......................................................................................... 154

Page 9: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

9

LIST OF TABLES

Table page 1-1 Many histone modifiers are evolutionary conserved and implicated in

tumorigenesis. .................................................................................................... 35

2-1 List of RNAi lines used for the screen of epigenetic modifiers of IRER. ............. 63

3-1 Drosophila strains used in this study .................................................................. 89

Page 10: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

10

LIST OF FIGURES

Figure page 1-1 Apoptotic pathways in Drosophila, mammals and C.elegans. ............................ 32

1-2 PcG and TrxG proteins regulate gene expression by modulating chromatin structure. ............................................................................................................ 33

1-3 Schematic diagram summarizing previous findings regarding the IRER.. .......... 34

2-1 Generation of IRER{ubi-DsRed}. ........................................................................ 55

2-2 The expression of IRER{ubi-DsRed} from embryos and larvae. ......................... 56

2-3 Verification of the IRER{ubi-DsRed} reporter for DNA accessibility and epigenetic status of IRER.. ................................................................................. 57

2-4 Monitoring the change of epigenetic status of IRER in wing discs.. .................... 58

2-5 The expression pattern of the ubi-DsRed reporter is largely controlled by the local chromosome environment. ......................................................................... 59

2-6 Su(var)3-9 knockdown-induced DsRed expression is not due to abnormal activation of the ubiquitin promoter. .................................................................... 60

2-7 The expression of IRER{ubi-DsRed} is responsible to HDAC.. .......................... 61

2-8 Monitoring the epigenetic status of IRER in different genetic backgrounds and in response to dietary restriction (DR).. ....................................................... 62

3-1 IRER mediates DNA damage –induced reaper and hid expression in the developing wing discs. ........................................................................................ 90

3-2 IRER is required for the control of imaginal disc size.. ....................................... 91

3-3 The cis regulatory function of IRER is required for organ size control. ............... 92

3-4 Differentially increased compartment size in Df(IRER) wings. ............................ 94

3-5 Homozygous Df(IRER) embryos have enlarged central nervous systems. ........ 95

3-6 Decreased expression levels of reaper in homozygous Df(IRER) embryos.. ..... 96

3-7 IRER is required for dMyc –induced cell death. .................................................. 97

3-8 Cell-autonomous role of IRER regulates dMyc-induced overgrowth.. ................ 99

3-9 Cells with an open IRER are more sensitive to dMyc -induced cell death. ....... 100

Page 11: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

11

3-10 Su(var)3-9 is required for the epigenetic silencing of IRER in wing discs. ........ 101

3-11 Epigenetic silencing of IRER is responsive to stress. ....................................... 102

3-12 Cells lacking IRER have the propensity to overgrown and are resistant to stress –induced cell death. ............................................................................... 103

3-13 Df(IRER) clones overgrow their twin spots in the eye discs.............................. 105

3-14 High order chromatin organization brings IRER into the proximity of multiple RHG gene promoters. ...................................................................................... 106

3-15 Diagram summarizing the mechanism of IRER. ............................................... 107

4-1 IRER is required for super competition -induced elimination of loser cells.. ..... 120

4-2 IRER is required for super competition -induced hid expression in loser cells.. 121

4-3 IRER is required for the elimination of scrib knockdown cells........................... 122

4-4 Scrib knockdown leads to differentiation defects of photoreceptor cells.. ......... 123

4-5 Scrib knockdown phenotype in eyes.. .............................................................. 124

4-6 IRER is required for scrib RNAi-induced reaper and hid expression from eye discs. ................................................................................................................ 125

4-7 Disruption of cell polarity leads to de-repression of IRER.. ............................... 126

Page 12: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

12

LIST OF ABBREVIATIONS

AEL After egg laying

ChIP Chromatin Immuno-precipitation

CNS Central nervous system

Cyc Cyclin

DIAP Drosophila inhibitor of apoptosis protein

H3K9me3 Trimethylated histone 3 lysine 9

H3K27me3 Trimethylated histone 3 lysine 27

Hdac1 Histone deacetylase 1

HMTase Histone methyltransferase

IBM IAP-binding motif

IRER Irradiation responsive enhancer region

JNK c-Jun amino-terminal kinase

NDR Nuclear Dbf2-Related

NTS Neoplastic tumor suppressor

PcG Polycomb group

PRE Polycomb response elements

PUMA p53 upregulated modulator of apoptosis

QPCR Quantitative polymerase chain reaction

Su(Hw) Suppressor of hairy wing

Su(var)3-9 Suppressor of variegation 3-9

TNF Tumor necrosis factor

trxG Trithorax group

TUNEL Terminal deoxynucleotidyl transferase dUTP nick end labeling

UAS Upstream activation sequence

Page 13: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

13

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

EPIGENETIC REGULATION OF APOPTOSIS DURING DEVELOPMENT AND

TUMORIGENESIS IN DROSOPHILA

By Can Zhang

August 2012

Chair: Lei Zhou Major: Medical Sciences -- Genetics

The balance between proliferation and apoptosis is essential for tissue

homeostasis during animal development. Induction of apoptosis, especially in the

context of elevated oncogene expression and overproliferation, has been considered as

a major cell-autonomous tumor suppression mechanism. At the same time, emerging

evidence suggests that cellular competition–induced apoptosis of cells with lower fitness

reflects a surveillance mechanism to optimize the organismal fitness, and is likely also

implicated in the process of tumor development.

Our previous study has characterized an intergenic region, the IRER (Irradiation

Responsive Enhancer Region), required for mediating DNA damage–induced

expression of multiple pro-apoptotic genes and cell death. Interestingly, the DNA

accessibility of this region, and consequently the responsiveness of the pro-apoptotic

genes to DNA damage, is controlled by histone modification. In this study, we provided

evidence indicating that the IRER functions as a locus control region for determining cell

numbers during development and for the induction of pro-apoptotic genes in response

to oncogenic stress. Animals lacking the IRER are viable but have superfluous cells in

multiple tissues. The regulatory function of the IRER is also required for the induction of

Page 14: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

14

reaper and the compensatory apoptosis following dMyc–induced overproliferation to

prevent hyperplasia. Therefore, similar to mammalian systems, overproliferation -

induced apoptosis overlaps with the DNA damage response in Drosophila.

Cell competition is a “social” behavior which eliminates viable but suboptimal cells

via apoptosis. Some growth factors, such as dMyc, can transform cells into super

competitors and induce apoptosis in neighboring cells with lower levels of dMyc. We

show that the IRER is required for dMyc-induced non-cell autonomous apoptosis by

controlling the expression of pro-apoptotic gene hid. In addition, cells mutant for

neoplastic tumor suppressor scribble are outcompeted by surrounding wild type cells

through apoptosis. We found that the IRER is required for pro-apoptotic gene

expression and cell death in scribRNAi cells. In the absence of the IRER, the scribRNAi

cells generated in larval epithelium are able to escape the competitive stress and grow

into neoplastic tumors in adult flies. Moreover, the chromatin conformation of IRER is

responsible to the cellular levels of scrib, and is de-repressed in scribRNAi cells. The

increased accessibility of the IRER may render cells more sensitive to competitive

stress and favor apoptosis in loser cells. Taken together, our studies provide the

mechanistic link between epigenetic regulation of apoptosis and tumorigenesis.

Page 15: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

15

CHAPTER 1 BACKGROUND AND INTRODUCTION

Regulation of Apoptosis in Drosophila

Apoptosis is a tightly regulated and conserved process of cell suicide in

multicellular organisms. The apoptotic cells are characterized by a series of

morphological changes including membrane ‘blebbing’, cell shrinkage, chromatin

condensation and nuclear fragmentation (Kerr et al., 1972). During normal

development, apoptosis helps to remove cells that are no longer needed to sculpt

organs. In adult tissues, cell death balances perfectly with cell division to adjust cell

number and maintain organ size. In addition, the damaged and potentially dangerous

cells need to be removed via apoptosis. Genetic or epigenetic deregulation of this

physiological process leads to severe developmental defects and diseases such as

tumors and neurodegenerative disease (Baehrecke, 2002). Drosophila has made a

great contribution to our understanding of the regulation of cell death due to the fruit fly’s

practicability in large-scaled genetic screens, low gene redundancy, and the availability

of powerful genetic tools.

The Apoptosis Regulatory Pathway in Drosophila

The components and mechanisms of apoptosis are highly conserved from flies to

humans (Figure 1-1). In general, the final execution of apoptosis requires activation of

caspases such as the initiator caspase Dronc and Dredd (Chen et al., 1998; Quinn et

al., 2000; Xu et al., 2005), and the effector caspases Drice and Dcp-1 (Xu et al., 2006).

However, the fact that caspases are ubiquitously expressed in all living cells suggests

caspases are simultaneously subject to negative regulation (Kornbluth and White,

2005). Indeed, the inhibitor of apoptosis proteins (IAPs) are identified as the “brakes on

Page 16: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

16

death”: they inhibit caspase activation and therefore suppress apoptosis (Salvesen and

Duckett, 2002; Vaux and Silke, 2005). In fruit flies, Drosophila IAP-1 (Diap1) plays an

essential role in protecting cells from inappropriate caspase activation and constitutive

apoptosis. All cells die synchronously in embryos lacking functional Diap1

(Chandraratna et al., 2007; Goyal et al., 2000; Wang et al., 1999).

On the other hand, in response to apoptotic signals or developmental cues, a

group of pro-apoptotic proteins are transcriptionally upregulated in doomed cells to

inhibit IAP and induce apoptosis (Song and Steller, 1999). Four pro-apoptotic genes,

including reaper (White et al., 1994), head involution defective (hid) (Grether et al.,

1995), grim (Chen et al., 1996), and sickle (Christich et al., 2002; Srinivasula et al.,

2002; Wing et al., 2002), have been identified in the Drosophila genome through genetic

screens and microarray studies. Coincidently, all four pro-apoptotic genes reside in a

highly conserved ~350 kb genetic regulatory block, share the same transcription

orientation, and are often coordinately regulated during apoptosis (Lin et al., 2009). The

proper transcriptional and translational regulation of pro-apoptotic genes is essential for

triggering apoptosis in cells destined to die.

Developmental Cell Death in Drosophila

Apoptosis plays crucial roles throughout the entire Drosophila development. The

earliest apoptotic events start at embryonic stage 11 (about 7 hours after egg laying

(AEL)) in the precephalic region and quickly spreads to other segments by stage 12-14

(8-10 hours AEL) (Abrams et al., 1993). It has been well appreciated that the segmental

repeated pattern of apoptosis helps to establish the precise border of segmentation in

the late-stage embryo (Pazdera et al., 1998). Indeed, the temporal and spatial pattern of

embryonic cell death associates with and contributes to certain developmental events

Page 17: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

17

such as head involution and germ band retraction. In post-embryonic tissues such as

the eye imaginal discs, cell death occuring between 24 and 30 hours after pupal

formation (APF) is required to remove surplus interommatidial cells; disruption of this

process leads to a rough eye phenotype (Wolff and Ready, 1991). During

metamorphosis, larval tissues are reorganized and develop into adult structures. Many

larval structures, such as salivary glands, are no longer needed and are removed by

ecdysone-mediated programmed cell death (PCD) (Jiang et al., 1997). In newly

emerged adult flies, PCD is initiated to remove wing epithelial cells at the time of wing

spreading (Kimura et al., 2004). In addition to these normal developmental events,

apoptosis also plays a role in removing cells incorrectly specified during development

(Werz et al., 2005).

DNA Damage-induced Cell Death in Drosophila

Although cells have evolved with a series of mechanisms to preserve genome

stability, the integrity of DNA is still frequently challenged by a wide variety of insults.

Cells are faced with the decision to either undergo DNA repair to correct the damage, or

initiate apoptosis to eliminate themselves from the organism (Friedberg, 1995). Defects

in the appropriate apoptotic response may lead to an accumulation of cells carrying

mutations and genomic instability, which can potentially have devastating

consequences for the organism such as tumorigenesis.

Ionizing radiation is able to induce single- and double- strand breaks in DNA, and

such DNA lesions need to be repaired by appropriate repair mechanisms or be

eliminated through apoptosis. A high dosage of irradiation (e.g. 40Gy) reliably induces

apoptosis within 20 minutes in early stage Drosophila embryos, which involves the

induction of pro-apoptotic genes reaper, hid, and sickle (Brodsky et al., 2004; White et

Page 18: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

18

al., 1994; Zhang et al., 2008). Although the imaginal discs from third in star larvae show

very little developmental apoptosis (Milan et al., 1997), they respond with elevated

levels of apoptosis as soon as 4 hours after irradiation, as recognized by TUNEL and

cleaved Caspase 3 immunostaining (Perez-Garijo et al., 2004). Because of its ability to

elicit a rapid apoptotic response and its broad application to cancer therapy, ionizing

radiation has been widely used as a tool to study the molecular mechanisms and signal

transductions of DNA damage-induced cell death.

Abundant evidence suggests that transcriptional mechanisms play crucial roles in

regulating DNA damage-induced apoptosis. The genetic requirement of transcription

factors such as P53 in irradiation-induced cell death underscores the importance of the

transcriptional machinery. Like its mammalian ortholog, the Drosophila P53 (DmP53) is

also required for the DNA damage response. Following irradiation and many other types

of stress, cellular levels of DmP53 increase dramatically. In addition, irradiation-induced

immediate apoptosis is totally abolished in flies DmP53 null mutants (Brodsky et al.,

2004; Sogame et al., 2003). Genetic and microarray analysis performed on Drosophila

embryos indicate that all irradiation responsive genes are direct or indirect

transcriptional targets of DmP53, including pro-apoptotic genes reaper, hid and sickle

(Brodsky et al., 2004). While P53 is required for a rapid induction of cell death following

DNA damage, P53-independent mechanisms do exist in both mammals and fruit flies,

albeit with a reduced and delayed response (Urist et al., 2004; Wichmann et al., 2006).

In flies, DmP53-independent apoptosis proves to be hid-dependent. It works in parallel

with the DmP53-dependent pathway to limit DNA damage-induced aneuploidy during

development (McNamee and Brodsky, 2009).

Page 19: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

19

In addition to the P53-pathway, the evolutionarily conserved c-Jun N-terminal

kinase (JNK) signal cascade also plays a crucial role in stress response. JNK activity,

when monitored in vivo with an enhancer trap line of puc-lacZ, is strongly induced in the

wing discs by γ-irradiation (puc, puckered, is a downstream target of the JNK pathway).

Moreover, radiation-induced apoptosis is greatly attenuated when the JNK pathway is

blocked, suggesting the vital role of JNK in irradiation-induced apoptosis (McEwen and

Peifer, 2005). In addition to irradiation stress, JNK also regulates apoptosis in response

to many other stresses, such as UV exposure (Jassim et al., 2003), ectopic

morphogens expression (Adachi-Yamada et al., 1999), and growth factor withdrawal

(Luo et al., 2007).

Developmental Control of Tissue Growth in Drosophila

How to achieve proper organ size is a fundamental question in developmental

biology. Cell growth, cell division, and cell death have to be coordinately regulated by

physiological cues to maintain homeostasis in multicellular organisms (Conlon and Raff,

1999). Genetic studies from invertebrates and vertebrates strongly suggest the

existence of an intrinsic signal that stops growth at the right time during development.

One piece of evidence comes from a famous liver regeneration test performed in 1930s:

when two thirds of a rat liver was removed, the remaining part of the liver regenerated

and eventually made up the lost mass (Michalopoulos and DeFrances, 1997). Since

then, the molecular mechanisms of organ size control have been intensively studied

during the past decade and conspicuous progress has been made. This has expanded

our knowledge of how cell growth and cell death are balanced in normal development,

and how a disturbed balance contributes to disease development.

Page 20: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

20

Organ Size Control

Hippo pathway

Genetic screens in Drosophila led to the identification of an evolutionarily

conserved Hippo signaling pathway which is essential for organ size control (Pan,

2010). The core components of this pathway include the Ste20-like kinases Hippo (Hpo)

and its co-activator Salvador (Sav), the NDR family protein kinase Warts (Wts) and its

adaptor protein Mob as a tumor suppressor (Mats), and the transcriptional co-activator

Yorkie (Yki). These proteins function in a kinase cascade to control organ size. Hpo and

Sav physically interact with each other to phosphorylate and activate Wts, which in turn

phosphorylates Yki with the help of Mats. The non-phosphorylated Yki is able to enter

the nucleus to mediate transcription of its target genes, such as the growth regulator

dMyc, the cell cycle promoting factor cycE, the apoptosis inhibitor Diap1, and the anti-

apoptotic microRNA bantam and others. Upon phosphorylation, Yki stalls in the

cytoplasm and thus fails to activate target gene transcription. Therefore, Yki is a growth-

promoter whereas Sav/Hpo/Wts function as tumor suppressors to simultaneously

restrict proliferation and promote apoptosis. Indeed, most components of the Hippo

pathway were originally identified as hyperplastic tumor suppressors. Clones of Hpo,

Sav or Wts mutant cells display increased proliferation and are relatively resistant to

apoptosis. Consequently, animals carrying these mutant clones develop enlarged

tissues without differentiation defects (Halder and Johnson, 2011; Hariharan and Bilder,

2006; Pan, 2010; Zhao et al., 2011).

Studies over the past few years have uncovered several upstream regulators that

positively regulate the Hippo pathway, including the cytoskeleton proteins Merlin (Mer)

and Expanded (Ex) (Hamaratoglu et al., 2006), their binding partner Kibra (Baumgartner

Page 21: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

21

et al., 2010; Genevet et al., 2010; Yu et al., 2010), the trans-membrane proteins

Crumbs (Crb) (Chen et al., 2010; Robinson et al., 2010), Fat (Ft) and Dachsous (Ds)

(Bennett and Harvey, 2006; Cho et al., 2006; Silva et al., 2006; Willecke et al., 2006). It

is interesting to notice that unlike the core components in the Hippo pathway (i.e.

Sav/Hpo/Wts), mutations in each of these upstream regulators fail to induce a strong

overgrown phenotype, suggesting that they may cooperate with each other to regulate

Hippo signal. The list of upstream regulators was further expanded by including a group

of cell-polarity proteins, such as the atypical protein kinase C (aPKC) and the Scribble

(Scrib)-Lethal giant larvae (Lgl)-Disc large (Dlg) protein complex (Chen et al., 2010;

Grzeschik et al., 2010; Ling et al., 2010; Robinson et al., 2010). These players could

regulate the Hippo pathway by affecting the cellular localization of Hpo and the

transcriptional activity of Yki. These studies implicate the cooperation of cell polarity, cell

contact and cell growth in organ size control. Further studies are required to uncover the

molecular mechanisms and biological significance of how polarity factors regulate cell

growth via the Hippo signaling pathway.

EGFR pathway

Many organs develop to appropriate and reproducible size in spite of a

considerable plasticity in cell growth rates. This suggests the organ size regulation

might employ certain secreted extracellular signals such as morphogens from

neighboring tissues or compartments. Such a mechanism has been nicely characterized

and shown to control compartment size in Drosophila embryos. Spi/EGFR is secreted

from neighboring anterior compartments in limited amounts and its up-taken by cells in

posterior compartments stimulates their growth and proliferation. However, the

increased cell proliferation eventually causes a drop of cellular levels of Spi, which in

Page 22: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

22

turn leads to reduced cell size and compensatory apoptosis, leaving the entire

compartment size unchanged (Parker, 2006). The EGFR signal promotes cell survival

by activating a downstream MAP-kinase signal, which can phosphorylate and inactivate

Hid protein to block Hid-induced apoptosis (Bergmann et al., 1998). Therefore,

insufficiency of Spi/EGFR signal may trigger apoptosis by releasing Hid activity. Thus, in

organs where cell survival is growth factor-dependent, apoptosis regulation presumably

plays a key role in controlling organ size.

Cell Competition

Recent studies of flies have shed light on how proliferation signals and death

signals are coupled to determine cell fates in a growing tissue. In one study, de la Cova

and colleagues reported that local expression of proto-oncogene dMyc is sufficient to

induce non-cell autonomous apoptosis in neighboring wild type cells, through a process

called cell competition. Cell competition happens when cells with different fitness meet

each other in the same compartment, and results in the expanding of stronger cells at

the expense of the suboptimal population (de Beco et al., 2012). The removal of viable

but less-fitted cells through cell competition may reflect a process of positive selection,

which ensures accumulation of cells with higher fitness in the organism.

The first evidence of cell competition was obtained from a series of Minute (M)

mutations targeting ribosome protein-coding genes. Cells heterozygous for Minute (M/+)

have defects in ribosomal function and are vigorously out-competed by wild type cells

(Morata and Ripoll, 1975). When clones of M/+ cells are induced in otherwise wild type

wing discs, the M/+ cells fail to be recovered from the adult wing due to disproportional

elimination by apoptosis (Moreno et al., 2002a). Cell competition also happens under

many other circumstances. For example, mutations of Hpo/Sav/Wats or over-

Page 23: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

23

expression of Yki can transform cells into super competitors who out compete

surrounding wild type cells. In addition, mutations of several Hippo pathway

components, including hpo, sav, mats, warts, ex and fat, allow further survival of M/-

cells, suggesting them as important regulators for cell competition (Tyler et al., 2007). It

has been postulated that Hippo-induced competition is attributable to the differential

expression of its target gene dMyc, a hypothesis which needs to be further verified.

Nonetheless, the discovery that imbalances in the Hippo pathway affects cell

competition shed lights on the involvement of cell competition in growth regulation and

tumor progression.

One consequence of cell competition is the elimination of loser cells via apoptosis,

which requires the activation of JNK signal. However, the contribution of JNK to loser

cell death varies depending on situation. In Minute- or dMyc-induced cell competition,

JNK seems to play a minor role in triggering cell death, as evidenced by continued loser

cell death even when the JNK signal is totally blocked (de la Cova et al., 2004; Moreno

and Basler, 2004; Moreno et al., 2002a). Conversely, JNK is absolutely required and

responsible for eliminating cells mutant for neoplastic tumor suppressors (NTSs). The

Drosophila TNF homologue Eiger (Egr) is up-regulated from neighboring wild type cells

and/or hemocytes, which is further taken up by the NTS mutant cells to activate JNK

signal (Igaki et al., 2009). In the absence of Eiger or JNK, these NTS mutant cells are

able to escape from competitive stress, and grow into neoplastic tumor. Recent studies

have revealed additional roles of JNK in inducing cell death following competition. First,

JNK is able to enhance Hippo activity in NTS mutant clones, which leads to decreased

expression of Hippo target genes including dMyc. The reduced dMyc signal could in

Page 24: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

24

turn boost their elimination (Chen et al., 2012). In addition, JNK may also play a role in

promoting the compensatory proliferation from winner cells (Fan and Bergmann, 2008;

Ryoo et al., 2004).

Although many advances have been made in our understanding of the molecular

mechanisms of cell competition, there are still a lot of unanswered questions. For

example, how the cells are identified as winner and loser status when they confront

each other, what is the physiological significance of adapting cell competition, whether

there is a common cause of competition in different situations, how the activated JNK

signal is transformed to death consequence, etc. Understanding these questions will

help us to draw a more comprehensive picture of the competition phenomenon, and

more importantly, to transform our knowledge to clinical research and disease

treatment.

Extra Sex Combs, Chromatin, and Cellular Identity

Identification of PcG and TrxG Proteins as Chromatin Regulators

The accurate placement of segmental structures along the anterior-posterior axis

of the animal body is defined by the highly conserved homeotic (Hox) gene family.

Dysregulation of Hox genes lead to homeotic transformation, that is transformation of

one body segment into another (Pearson et al., 2005). Therefore, in order to maintain

cellular identity, the Hox gene expression pattern has to be properly established and

tightly maintained. In Drosophila, the Hox gene expression is initiated by transient

transcriptional factors encoded by gap genes and pair-rule genes in early

embryogenesis. However, the transient transcription factors disappear soon after

turning on the Hox genes. Development of an organism requires a cellular system to

“remember” the appropriate Hox gene expression status ascribed to individual cell.

Page 25: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

25

Genetic analysis unraveled two groups of genes affecting cellular identity:

Polycomb group (PcG) and Trithorax group (TrxG). The first two PcG genes - Extra sex

combs (Esc) and Polycomb (Pc), were described in Drosophila by Lewis and colleagues

in the 1940s. They were named after the mutation phenotype: male flies grew extra sex

combs on the second and third legs, which were usually restricted to the first

legs(Lewis, 1978; Lewis, 1947). Further mechanistic studies revealed that the PcG

mutant cells inappropriately reactivate specific Hox genes which should have been

repressed, transforming one body segment into another (Jurgens, 1985; Struhl, 1983).

This failure in the cellular memory system leads to the idea that PcG functions to

maintain the repressed state of Hox gene (Pirrotta, 1997). Although originally identified

in Drosophila, PcG function has been shown to be fairly well conserved throughout

evolution: several Polycomb mutants in mice exhibit anterior-posterior transformations

and other abnormalities of the axial skeleton (Akasaka et al., 1996; Core et al., 1997;

del Mar Lorente et al., 2000).

The TrxG proteins have been characterized as an antagonistic system of PcG

proteins, which set up an active state for the Hox gene. In the absence of Trithorax

(TRX), the best characterized member in TrxG, multiple homeotic genes become

repressed in a PcG-dependent fashion from cells where they are expressed in early

stage embryos. Consequently, flies show segmental transformations, similar to the

phenotypes of Hox gene mutants (Breen and Harte, 1991; Orlando and Paro, 1995).

Therefore, PcG and TrxG work together, through either repressive or activating

mechanisms, to achieve the appropriate temporal and spatial pattern of Hox gene

expression. Although PcG and TrxG proteins were firstly discovered to regulate Hox

Page 26: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

26

genes, further studies have identified a variety of target genes involved in stem cell

maintenance, cell cycle control, apoptosis, etc. Genetic lesions inducing inappropriate

PcG and TrxG activity may perturb these fundamental biological processes, leading to

pathogenesis such as tumor (Mills, 2010).

Chromatin Structures and Epigenetic Regulations

The term “epigenetics” is used to define the heritable changes of gene expression

pattern that are not caused by changes of primary DNA sequence. Epigenetic

inheritance can be produced by distinct mechanisms, such as DNA methylation,

chromatin modifications, and non-coding RNA. Here we focus on the epigenetic

regulation through the modification of chromatin structures.

In eukaryotic cells, DNA wraps around core histone octamers to form

nucleosomes, which are further folded into higher order chromatin. Different chromatin

conformations are usually associated with varying DNA accessibility and transcriptional

potential. In general, an open chromatin or ‘euchromatin’ facilitates transcription

whereas a compact chromatin or ‘heterochromatin’ makes the underlying genes highly

resistant to transcriptional activity. The impact of heterochromatin configuration on gene

silencing has been noticed decades ago through the study of position effect variegation

(PEV), which reveals that gene activity is dependent on its position relative to a

heterochromatin region on chromosome. When a Drosophila gene required for red eye

pigmentation was placed in proximity to the pericentric heterochromatin, it became

silenced in a subset of cells and resulted in a mosaic eye color (Muller, 1932).

The chromatin structures can be changed by histone modifications. The N-terminal

tails of core histones that protrude from nucleosomes are subject to a variety of post-

translational covalent modifications. The histone modifications provide a scaffold for the

Page 27: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

27

recruitment of regulatory proteins or chromatin remodeling factors, which in turn define

distinct chromatin states(Jenuwein and Allis, 2001). For example, trimethylation at

lysine 9 or lysine 27 on histone H3 serves as repressive histone mark for the

transcriptionally silent heterochromatin, whereas trimethylation at lysine 4 on histone H3

as well as acetylation on histone H3 has been closely linked transcriptionally active

chromatin.

PcG/TrxG Mediated Chromatin Modifications

In flies, there are at least two types of multi-protein complexes working together to

conduct PcG-mediated silencing. They are referred to as the Polycomb Repressive

Complex 1 and 2 (PRC1 and PRC2), which function to maintain and establish the

silenced chromatin states, respectively. The more recently characterized PhoRC mainly

contains PHO/PHOL and SFMBT (Scm-related gene containing four MBT domains),

which may process DNA-binding properties (Schwartz and Pirrotta, 2007).

The biochemically purified Drosophila PRC2 core complex consists of Enhancer of

zeste (E(Z)), Suppressor of zeste 12 (SU(Z)12) and Extra sex comb (ESC). E(Z) carries

an enzymatically active SET domain, which has histone methyltransferase activity and

is able to catalyze trimethylation at lysine27 of histone H3 (H3K27Me3), a repressive

histone mark associated with gene silencing (Cao et al., 2002; Czermin et al., 2002;

Müller et al., 2002). The Drosophila PRC1 complex is more diverse and mainly contains

Polycomb (PC), Posterior sex combs (PSC), Polyhomeotic (PH) and RING (Saurin et

al., 2001; Shao et al., 1999). The Chomodomain in PC specifically recognizes the

H3K27Me3 established by PRC2, and functions to stabilize the repressed status of

chromatin. The core components of mammalian PRC1 and PRC2 are very similar to

Page 28: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

28

those in Drosophila but involve more paralogs (Table 1-1), which may function as

alternatives to target different genes or different tissues (Levine et al., 2002).

How PRCs modulate transcriptional silencing remains to be fully understood. The

potential mechanisms include facilitating chromatin compaction, impeding RNA Pol II

initiation and elongation, recruiting DNMTs (DNA methyltransferases) to target genes,

as well as blocking the modulation of SWI/SNF chromatin remodeling complex

(Sparmann and van Lohuizen, 2006). In addition, the RING finger domain of RING

protein possesses E3 ubiquitin ligase activity and induces mono-ubiquitination of

H2AK119, which is also associated with gene silencing (Cao et al., 2005; de Napoles et

al., 2004; Wang et al., 2004).

Similar to PcGs, TrxG proteins also form multi-component complexes. They

maintain an active chromatin state through either direct histone modification or ATP-

dependent nucleosome remodeling (Strahl and Allis, 2000; Vignali et al., 2000). The

founding member of the Drosophila TrxG family, TRX, is a histone methyltransferase

which catalyzes H3K4 trimethylation to favor transcriptional activation (Santos-Rosa et

al., 2002). TrxG proteins also regulate chromatin dynamics through nucleosome

remodeling: The SWI-SNF complex contains ATP-dependent chromatin remodeling

proteins, which are able to alter the nucleosome structures to facilitate basal

transcription machinery (Smith and Peterson, 2005).

In addition to direct methylation, both PcG and TrxG complexes can recruit other

histone modifiers to ensure transcriptional repression or activation. For example, they

both can recruit histone demethylases (HDM). The TrxG proteins can recruit HDMs (e.g.

UTX) that specifically remove methyl groups from repressive histone mark of

Page 29: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

29

H3K27me3. A human HDM, JARID1, has been found to demethylate H3K4me3 and

associate with PcG proteins (Agger et al., 2007; Hong et al., 2007; Lee et al., 2007; Shi,

2007). Other histone modifiers such as histone acetyltransferase (HAT) and histone

deacetylase (HDAC) are also recruited either directly or indirectly to modulate

transcription (Mills, 2010; Sparmann and van Lohuizen, 2006) (Fig 1-2). It is believed

that the coordinated removal of repressive marks and deposition of positive marks (and

vice versa) is important for chromatin dynamics and transcription.

Epigenetic Regulation of Apoptosis

Dysregulation of PcG Targeting

Epigenetic regulation could be disturbed to promote tumorigenesis without

significant changes of the global level of suppressive histone modification. For instance,

the long non-coding RNA HOTAIR can promote tumorigenesis through genome-wide

re-targeting of PRC-2 to a pattern that resembles undifferentiated/lowly differentiated

cells (Gupta et al., 2010). Such aberrant targeting of PcG proteins is known to promote

the silencing of tumor-suppressor genes, which are in a transcription-ready (bivalent)

state in normal stem cells (Ohm et al., 2007).

Altered PcG targeting can be induced by oncogenic proteins. For instance,

oncogenic Ras can lead to repression of Fas expression. Although at the end stage the

silencing of Fas (and several other tumor suppressor genes) is manifested as both DNA

hypermethylation and increased suppressive histone modification, a mechanistic

analysis indicated that PcG proteins such as Bmi1 and EzH2 are required for Ras-

induced silencing of Fas (Gazin et al., 2007a). Interestingly, a small adenoviral protein,

E4-ORF6, can activate an unknown mechanism that leads to the formation of facultative

heterochromatins in P53-targeted stress-responsive genes (Soria et al., 2010a). Since

Page 30: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

30

E4-ORF6 lacks any enzymatic domain that can modify histone tails, it must act through

a cellular pathway that can specifically silence P53-targeted stress responsive genes.

Insight into this mechanism would certainly advance our understanding of how PcG-

mediated epigenetic silencing can be targeted to specific groups of genes.

Epigenetic Silencing of Pro-apoptotic Genes

Epigenetic regulation of P53-targeted stress-responsive genes is also observed

during Drosophila development. A 33kb intergenic region located in the pro-apoptotic

gene cluster is responsible for mediating P53-dependent induction of reaper, hid, and

sickle. This region, Irradiation Responsive Enhancer Region (IRER), is open in early

embryonic stages when most cells are proliferating, conveying high sensitivity to DNA

damage and other stresses. However, at embryonic stage 12, when most cells enter

post-mitotic differentiation, this region forms a heterochromatin-like structure enriched

for both H3K9Me3 and H3K27Me3. Consequently, the three pro-apoptotic genes can no

longer be induced following irradiation (Zhang et al., 2008). This open-to-closed

transition of IRER requires the function of PcG proteins such as Pc and Su(z)12, as well

as HDAC and Su(var)3-9 (Figure 1-3). Interestingly, epigenetic suppression is strictly

limited to the irradiation-responsive enhancer region without affecting the transcribed

regions of the pro-apoptotic genes. This limitation is important since those pro-apoptotic

genes are expressed in a cell lineage –specific pattern in late stage embryos, and are

required for lineage specific cell death (Tan et al., 2011). The demarcation of epigenetic

blocking is achieved by a chromatin barrier separating IRER from the promoter and

transcribed regions of reaper. This barrier, located within a 294bp DNA fragment,

appears to be highly conserved as it can block heterochromatin propagation when

tested in a vertebrate system (Lin et al., 2011).

Page 31: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

31

The epigenetic blocking of IRER provided a nice system for elucidating the cellular

mechanisms that regulate targeted suppressive histone modification at tumor-

suppressor/stress-responsive genes. However, it left several fascinating questions

unanswered. First, genome-wide alignment revealed that IRER is highly conserved

throughout evolution: the underlying DNA sequence is even better conserved than the

neighboring coding sequence for reaper, suggesting the crucial regulatory function may

exist in this region (Zhang et al., 2008). Second, a more detailed study needs to be

performed to gain an insight into specific mechanisms controlling the epigenetic

regulation of IRER. My dissertation research defined the functional significance of IRER

in development and tumorigenesis, by providing evidence that an open IRER not only

conveys sensitivity to irradiation and DNA damage, but also increases the cellular

sensitivity to overproliferation- and competition-induced cell death. In addition, I

developed a novel method to monitor the epigenetic status of IRER at cellular resolution

in this study, which revealed a dynamic pattern of IRER accessibility, which likely

reflects the stochastic nature of epigenetic regulation of stress-responsive genes in a

given cell type.

Page 32: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

32

Figure 1-1. Apoptotic pathways in Drosophila, mammals and C.elegans. The key cell

death regulators are conserved across different species and the functional orthologues are highlighted in the same color. In Drosophila, in response to apoptotic stimuli, expression of the pro-apoptotic genes hid, reaper, grim, and sickle releases the initiator caspases (i.e. Dronk and Dreed) and effecter caspases (i.e. Drice and Dcp-1) from inhibition by Diap1, and induces apoptosis. Stimulatory and inhibitory interactions are indicated by arrows and T-bars, respectively. The extrinsic apoptotic pathway (or: death receptor pathway) is not included in this figure. IAP, inhibitor of apoptosis; Dark (also named as Dapaf-1 or HAC-1), Drosophila Apaf-1-related killer; SMAC, second mitochondria-derived activator of caspases; DIABLO, direct IAP-binding protein with low pI; Cyto c, cytochrome c; APAF1, apoptotic-protease-activating factor-1; CED, cell-death abnormality.

Page 33: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

33

Figure 1-2. PcG and TrxG proteins regulate gene expression by modulating chromatin

structure. PcG and TrxG proteins directly methylate specific histone residues to establish repressive (H3K27me3) and active (H3K4me3) histone marks, respectively. In addition, they are able to recruit enzymes that modulate other histone modifications such as acetylation and demethylation as well as DNA methylation. PcG complexes can associate with HDACs, H3K4me3-specific HDMs and DNMTs to suppress gene expression, whereas TrxG complexes recruit HATs and H3K27me3-specific HDMs to activate gene expression.

Page 34: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

34

Figure 1-3. Schematic diagram summarizing previous findings regarding the IRER.

Using insulator-containing P-element insertions, previous work from our lab has mapped the irradiation responsive enhancer region (IRER) to the 33kb intergenic sequence on the 3rd chromosome. It is located between two pro-apoptotic genes reaper and sickle, including the putative p53-response element (P53 RE). This enhancer region is subject to PcG-mediated epigenetic regulation and undergoes an open-to-closed transition during embryonic stage 11-12. The open chromatin structure in early stage embryos facilitates irradiation-induced transcription of reaper and hid and leads to apoptosis; whereas the condensed chromatin in late staged embryos precludes transcription and blocks apoptosis. The facultative heterochromatin formation is restricted to IRER by the IRER left barrier (ILB), which allows the reaper promoter to remain open throughout development and accessible to regulation. The barrier activity requires binding of the Cut protein, which may recruit chromatin modifying enzymes such as CBP; mechanistically, much remains to be elucidated.

Page 35: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

35

Table 1-1. Many histone modifiers are evolutionary conserved and implicated in tumorigenesis

Complex Drosophila protein Human homologues Functional domains Biochemical activity

PcG proteins

PRC1 Polycomb (PC) CBX2, CBX4, CBX6, CBX7, CBX8

Chromodomain Binding to H3K27me3

Posterior sex comb (PSC)

PCGF1(NSPc1), PCGF2(MEL18), PCGF4(BMI1)

Zinc finger Cofactor for Ring

Polyhomeotic (PH) PHC1, PHC2, PHC3 Zinc finger and SAM Required for silencing

Sex combs extra (SCE or RING)

RING1A, RING1B, RNF2

RING zinc finger H2AK119 ubiquitin ligase

Sex comb on midleg (SCM)

SCMH1, SCML2 SAM, MBT, Zinc finger

recruitment of the PcG protein

PRC2 Enhancer of zeste (E(Z))

EZH1 and EZH2 SET Histone methyltransferase, establish H3K27me3

Extra sex combs (ESC)

EED WD40 repeats Co-factor for E(Z)

Extra sex combs-like (ESCL)

EED WD40 repeats Co-factor for E(Z)

Suppressor of zeste 12 (SU(Z)12)

SUZ12 Zinc finger Co-factor for E(Z)

Polycomb-like (PCL) PCL1(PHF1), PCL2(MTF2), PCL3(PHF19)

PHD

PhoRC Pleiohomeotic (PHO) YY1, YY2 Zinc finger DNA binding

Pleiohomeotic-like (PHOL)

YY1, YY2 Zinc finger DNA binding

SFMBT L3MBTL2, MBTD1 MBT, SAM Binding to mono- and dimethyl H3K9, H4K20

TrxG proteins

TAC1 Trithorax (TRX) MLL, MLL2, MLL3, MLL5

SET Histone methyltransferase, establishes H3K4me3

dCBP CBP KIX, IBiD, zinc finger Histone acetyltransferase

dUTX UTX JmjC di- and trimethylated H3K27 demethylase

ASH1 ASH1L SET H3K4 and H3K36 methylase

ASH2 ASH2L, WDR5 WD40 repeats Essential for H3K4me3

Page 36: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

36

Table 1-1. Continued

Complex Drosophila protein Human homologues Functional domains Biochemical activity

TrxG proteins (continued)

TrxG proteins (continued)SWI-SNF nucleosome remodeling complex

OSA ARID1A, ARID1B

BRM BRM, BRG1 SWI-SNF-like helicase, Bromodomain

ATPase activity, binds acetylated histones

SNR1 SNF5, ARID4A, ARID4B

Non-catalytic core subunit

Other related histone modifiers

SIR2 SIRT1 Zinc finger NAD+-dependent

class III histone deacetylase

LID JARID1 JmjC di- and trimethylated H3K4 demethylase

SU(VAR)3-3 LSD1 SWIRM, amine oxidase domain

mono- and dimethylated H3K4 demethylase

PcG, polycomb group; TrxG, trithorax group; TAC, trithorax acetylation complex; SFMBT, Scm-related gene containing four MBT domains; CBP, CREB-binding protein; ASH, absent, small, or homeotic discs; SIR, silent information regulator; UTX, ubiquitously transcribed tetratricopeptide repeat, X chromosome; LID, little imaginal discs; CBX, chromobox homologue; PHC, polyhomeotic homologue; EZH, enhancer of zeste homologue; EED, embryonic ectoderm development; YY, Yin-Yang transcription factor; MLL, mixed lineage leukemia; LSD, lysine specific demethylase; PHD, plant homeodomain; WDR5, WD repeat domain 5.

Page 37: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

37

CHAPTER 2 MONITORING DNA ACCESSIBILITY AND EPIGENETIC STATUS IN INDIVIDUAL

CELLS AND LIVE ANIMALS WITH A FLUORESCENT REPORTER

Introduction

Even though all cells share the same genome in one organism, which genes are

expressed and when/how they are expressed are largely determined by epigenetic

regulation. This is achieved in large part by controlling the accessibility of the DNA to

the basic transcription machinery and DNA-binding transcription factors (Grewal and

Moazed, 2003; Schwartz and Pirrotta). The major epigenetic mechanisms that affect

DNA accessibility are DNA methylation and histone modification. However, DNA

methylation in Drosophila is restricted to early stage of embryogenesis and lacks a

systematic study (Lyko et al., 2000; Nanty et al., 2011) The main form of epigenetic

regulation in Drosophila is post-translational histone modification. While “active” histone

marks lead to open chromatin structure and increased transcription, “suppressive”

histone marks are usually associated with condensed conformations and reduced levels

of gene expression (Jenuwein and Allis, 2001).

DNA accessibility and epigenetic modification can be monitored by biochemical

assays such as DNase I sensitivity assay, chromatin immunoprecipitation (ChIP), etc.

These methods generally require homogenizing 106 ~ 107 cells to produce a readout.

The obtained result actually reflects an averaged level of the tested population. The

methods are applicable to the cultured cell lines but may not be suitable for measuring

epigenetic status in vivo, given our knowledge that epigenetic regulation could be, and

in many cases is, cell specific. Cells in the same tissue may have totally different

epigenetic profiles according to their differentiation status and many other factors. Even

cells of the same tissue type and differentiation status may display significant individual

Page 38: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

38

variation in epigenetic regulation. For example, when the white gene is inserted in close

proximity of a pericentromeric region, its expression is influenced by the centromere

heterochromatin formation and variegates among eye cells in the transgenic fly

(Karpen, 1994).

The change of DNA accessibility can directly influence the responsiveness of

genes to environmental stimuli. It is well known that cellular sensitivity to irradiation-

induced cell death can vary dramatically depending on cell type and differentiation

status. Using the developing Drosophila embryo as a model, we found that the

sensitive-to-resistant transition of the radiation responsiveness of pro-apoptotic genes

was mediated by an epigenetic mechanism (Zhang et al., 2008). The irradiation

responsive enhancer region (IRER) is required for mediating the irradiation-

responsiveness of three pro-apoptotic genes reaper, hid, and sickle. IRER is “open” or

accessible in proliferating and undifferentiated cells during early embryogenesis (3-7hr

after egg laying (AEL)). Irradiation treatment of embryos at this stage induces a 5-10

fold increase of the pro-apoptotic genes within 15~30 minutes, followed by wide-spread

apoptosis. However, this region forms a closed, DNase I –resistant structure in most

cells during post-mitotic differentiation (7-12-hr AEL) and renders the three pro-

apoptotic genes irresponsive to irradiation (Zhang et al., 2008). The open-to-closed

transition of IRER is accompanied by a dramatic increase of both trimethylated H3K27

(H3K27Me3) and trimethylated H3K9 (H3K9Me3) in the region. The epigenetic blocking

of IRER requires the function of HDAC-1, Su(var)3-9 (H3K9 methytransferase), and

several key Polycomb group (PcG) proteins (Zhang et al., 2008).

Page 39: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

39

In Drosophila embryogenesis, cell nuclei undergo 13 rounds of proliferation during

the blastoderm stage prior to onset of differentiation. This unique event allowed us to

monitor the epigenetic status of IRER by collecting large amounts of proliferating cells

from early stage embryos and performing DNase I sensitivity assay and ChIP. The

relatively synchronized initiation of cellular differentiation around embryonic stage 12

was conceivably crucial in allowing us, using population- based biochemical methods, to

detect the change of DNA accessibility and epigenetic modification in IRER during the

proliferation-to-differentiation transition. Intriguingly, although late stage embryos are

resistant to irradiation-induced cell death, many cells in developing imaginal discs at

larval stage become sensitive to irradiation-induced pro-apoptotic gene expression and

apoptosis, suggesting they may have an open IRER. The larval tissues are composed

of cells from different lineages and/or at different differentiation statuses. Thus, it is

impossible to discern the chromatin conformation of IRER in such a tissue using the

same biochemical approaches we applied to embryos, i.e. DNase sensitivity assay or

ChIP. To solve this problem, we designed a strategy which integrated a fluorescent

reporter into the epigenetically regulated IRER. Our experimental verification indicated

that the fluorescent reporter could be used to monitor cell- and tissue-specific epigenetic

regulation and changes of DNA accessibility during animal development.

Materials and Methods

Generation of IRER{ubi-DsRed}

Two bacterial artificial chromosome clones (BACR35F04 and BACR04F07)

harboring the genomic region of 18,372,802…18,379,434 (coordinates of Genome

release 4.3) were obtained from the BACPAC Resources Center

(http://bacpac.chori.org/). The ~6.5kb targeting region is located in the middle region of

Page 40: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

40

IRER and is highly accessible to DNase I digestion. The targeting region was cut by

BamHI/XhoI, purified from agarose gel, cloned into the p[Endsout2] vector and verified

by Sanger sequencing. A ~3kb ubiquitin-DsRed cassette was prepared by polymerase

chain reaction (PCR) from fly genomic DNA with NdeI sites added to the PCR primers.

The NdeI-{ubi-DsRed}-NedI PCR product was then sub-cloned into an endogenous

NdeI site (18,375,553) within the targeting IRER fragment in the p[Endsout2] vector, to

generate the donor construct. Multiple transgenic strains carrying the donor construct

were generated with the p-element-based integration. An insertion on the X-

chromosome was selected as the donor strain for homologous recombination. The

ends-out homologous recombination was conducted as previously described (Gong and

Golic, 2003). One recombination event was recovered from about 6,000 progenies

based on autosomal inheritance. The successful recombination into the endogenous

IRER on the 3rd chromosome was verified by genetic mapping, inverse PCR, and

southern blot.

Fly Handling

Unless otherwise mentioned, all the crosses were done at 25 degree. Alleles used:

Ser-Gal4 (Bloomington #6791), Hdac[303], Hdac[326] and Hdac[328] (Dr. Mottus),

Su(var)3-9[1] (Bloomington #6209), Su(var)3-9[2] (Bloomington #6210), UAS-RNAi

strains (Vienna Drosophila RNAi Center).

DNase I Sensitivity Assay and Chromatin Immunoprecipitation (ChIP)

The DNase I sensitivity assay and ChIP assay on staged embryos were performed

as previously described (Zhang et al., 2008).

Page 41: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

41

Immunofluorescence

Embryo in situ hybridization was performed as stated (Zhang et al., 2008). For the

imaginal disc in situ hybridization: third instar larvae of appropriate genotype were

briefly dissected in DEPC-PBS to facilitate imaginal disc attachment to the larval body

wall. Larvae were immediately fixed with 4% paraformaldehyde for 30 minutes,

dehydrated with a graded methanol series (50%-75%-90%-100%), and stored at -20°C

till use. The in situ hybridization signal was amplified and detected by TMR-conjugated

tyramide (TSA kit from PerkinElmer Life). Imaginal discs were further dissected and

mounted onto glass slides. Fluorescent images were taken with a Leica upright

fluorescent microscope using OpenLab software.

Flourometric Measurement of DsRed Expression

For each genotype, five newly eclosed male flies (< 2 days) were collected into a

1.5mL microcentrifuge tube containing 250uL assay buffer (50mM NaH2PO4, 10mM

Tris-HCl Ph8.0, 200mM NaCl) on ice. The sample was homogenized by pestle, and

centrifuged at 200g for 1 minute to remove tissue chunks. 200uL of supernatant

containing DNA was transferred to a new tube. Meanwhile, a blank sample consisting

solely of 200uL of assay buffer was prepared. The samples were stained with

Hoeschst33342 to a final concentration of 10µg/mL for 5 minutes on ice, and 40uL was

immediately pipetted into a 96-well plate. Fluorometric measurement was done with the

Perkin Elmer Victor3V 1420 Multiabel Counter with the settings of DsRed, excitation

555±19nm, emission 590±5nm; H33342, excitation 355±19nm, emission 460±5nm. If

the blank reading was too high, it was subtracted from each sample reading to correct

for background fluorescence. For each sample, I divided the DsRed value by the

H33342 value, which reflects the expression level of DsRed relative to DNA content.

Page 42: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

42

The two-tailed, un-paired Student’s t-test was used for statistical analysis and a p-value

< 0.05 was considered significant.

Results

Inserting the ubi-DsRed Reporter into IRER via Homologous Recombination

The silencing of transgenes by local epigenetic regulation has been observed in

several contexts. For instance, the white gene in P element- insertions near the

centromere shows variegated expression in the eye, which indicates the marker gene is

silenced in some cells but not others (Konev et al., 2003; Sun et al., 2004). This

silencing is due to the spread of H3K9Me3 modification during the formation of the

pericentromeric heterochromatin. Similarly, the UAS-lacZ transgene contained in a P

insertion in the regulatory region of bithorax(bx) cannot be induced by ubiquitously

expressed Gal4 in the anterior segments, because the bx locus there is epigenetically

suppressed by PcG-mediated trimethylation of H3K27 (McCall and Bender, 1996). In

contrast, the expression of the UAS-lacZ can be induced by Gal4 in posterior segments

where the regulatory region is not epigenetically suppressed. These reported

observations prompted us to postulate that the expression pattern of a reporter gene

with a constitutively active promoter inserted into IRER may reflect the epigenetic status

of IRER in any cell at any developmental point.

Using the “Ends-out” homologous recombination developed by Gong & Golic

(Gong and Golic, 2003), we first generated a transgenic strain carrying the donor

construct p(LA-ubi-DsRed.T4-SV40PolyA-RA) in the X-chromosome (Figure 2-1A).

DsRed.T4 is a modified version of the original DsRed protein with a maturation half-time

of about 40 mins (Bevis and Glick, 2002). LA and RA are two ~3kb consecutive

segments of IRER linked by an endogenous Nde1 site at 18,375,553-558 on the 3rd

Page 43: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

43

chromosome (the coordinates in Chapter 2 are based on genome release 4.3 unless

otherwise stated). The ubi is a 2.2 kb regulatory region extracted from the endogenous

ubiquitin-p63E gene that includes the first non-coding exon and an intron. The same

regulatory fragment has been used to generate markers/reporters that give ubiquitous

expression pattern throughout development (Brown and Castelli-Gair Hombria, 2000;

Supatto et al., 2005). The reporter flanked by the homologous arms was released from

the X chromosome with the FLP recombinase and subsequently cut by the I-SceI

endonuclease as described by Gong and Golic (Gong and Golic, 2003). A homologous

recombination event was recovered from about 6,000 progeny screened for autosome-

based inheritance of the fluorescent reporter as well as a decrease of the fluorescent

signal. The successful recombination was then verified with genetic mapping, Inverse

PCR, and southern blot (Figure 2-1B). The recombined strain was named as IRER{ubi-

DsRed}.

The DsRed mRNA, measured by in situ hybridization, is ubiquitously expressed at

high levels in early stage embryos, which is in agreement with the fact that IRER is

open and accessible in proliferating cells at this stage (Figure 2-1 C-D). However,

beginning with later embryogenesis, DsRed mRNA expression becomes restricted to a

small percentage of cells (Figure 2-1 E-F). In newly hatched larvae, strong DsRed

fluorescent signal can be detected from scattered cells in the nerve cord (Figure 2-2B),

a place known for continued neuroblast proliferation and apoptosis in late

embryogenesis and early larval stages (Peterson et al., 2002). The pattern of cells

positive for DsRed during the larval stages is very dynamic. 2 days AEL (1st Instar

larvae), a group of cells at the posterior epidermis develop a distinct signal (Figure 2-

Page 44: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

44

2C). In 3rd instar larvae, most cells have little or no detectable DsRed. In addition, the

restricted expression of the DsRed reporter was also observed from the arterial tip of

the germarium (Figure 2-1 G-I) and the polar cells of egg chambers in the adult ovary

(Figure 2-1J).

Verifying the Responsiveness of the Reporter to Changes in DNA Accessibility

The restricted expression pattern of IRER{ubi-DsRed} suggests that it is subject to

epigenetic regulation. To verify whether the DNA accessibility of the reporter is co-

regulated with IRER, we monitored the DNase I-sensitivity of the DsRed gene before

and after the open-to-closed transition of IRER (Figure 2-3A). Previously, through

DNase I sensitivity assay and ChIP analysis, we have documented that IRER

undergoes an open-to-closed transition during embryonic stage 12 which is

accompanied by the formation of repressive histone marks (H3K27Me3, H3K9Me3) and

the binding of PcG proteins and HP1(Zhang et al., 2008). Here we found that like the

surrounding IRER, the DsRed locus became significantly more resistant to DNase I

treatment following the open-to-closed transition of IRER. The magnitude of the change

at the reporter locus (DsRed) was not as large as the neighboring genomic loci.

Consequently, the reporter locus was not as resistant to DNase I as the neighboring

IRER region (Figure 2-3B). This is likely due to the impact of the strong ubiquitin

enhancer/promoter on the local environment of histone modification and DNA

accessibility. However, this impact appears to be limited to the reporter locus without

affecting epigenetic suppression of IRER during the transition. The immediate

neighboring loci (18,374, 377) became as resistant to DNase I as the heterochromatin

control (H23), same as what was observed in wild type animals (Zhang et al., 2008).

Page 45: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

45

Even though the change of DNase sensitivity at the DsRed locus is not prominent

until the end of embryogenesis, it appears that the enrichment of suppressive histone

mark H3K27Me3 as well as binding of Polycomb group protein PSC (posterior sex

comb) is similar to the neighboring IRER loci (Figure 2-3C). Similarly, the accumulation

of H3K9Me3 and the binding of HP1 to the reporter are also comparable to that of the

neighboring loci and the H23 constitutive centromeric heterochromatin region (Figure 2-

3D). The formation of suppressive histone marks and the binding of PSC and HP1

appear to play a significant role in constraining the expression of the reporter. This may

be due to the fact that the accessibility to large complexes such as RNA Pol II can be

blocked long before the DNA becomes inaccessible to DNase I, a phenomenon that has

been reported previously and noticed by us in our early study of IRER (Zhang et al.,

2008).

By the time animals reach 2nd instar larval stage (~3-4 days AEL at 25 oC), few

cells have strong DsRed signal comparable to early embryogenesis. To verify whether

DsRed can only be expressed at a high level in cells with an open conformation of

IRER, we used fluorescence-activated cell sorting (FACS) to separate DsRed positive

(+) and negative (-) cells isolated from a mixture of 2nd and 3rd instar larvae. Using a

protocol adapted from Okataba et al. (Oktaba et al., 2008), we found that about 2% cells

dissociated from shredded larval tissues can be reliably separated as DsRed(+). ChIP

analysis of the sorted cells indicates that both the reporter and the IRER region are

enriched for H3K27Me3 and H3K9Me3 in DsRed(-) cells. There is also a significant

binding of PSC and HP1 to the reporter and IRER in these cells (data not shown). In

contrast, DsRed(+) cells have much lower levels of suppressive histone modifications,

Page 46: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

46

which is similar to what has been observed for undifferentiated cells during early

embryogenesis. Correspondingly, DsRed(+) cells are extremely sensitive to the stress

associated with tissue dissociation and FACS. Many DsRed(+) cells are dying or dead

following FACS, as monitored by DAPI staining via microscopy (data not shown). The

level of reaper in DsRed(+) cells is about 8 times as high as that in DsRed(-) cells

(Figure 2-3E).

The aforementioned evidence indicates that IRER{ubi-DsRed} is a reliable reporter

for the DNA accessibility of IRER. Higher levels of reporter expression correlate with an

open chromatin conformation in the IRER while lower levels of IRER{ubi-DsRed}

expression correlate with the formation of suppressive histone marks in IRER. Since the

ubiquitin promoter is presumably a constitutively active promoter, it may interfere with

the suppressive histone modifications in IRER. It has been reported that active

transcription can disrupt Polycomb-mediated silencing (Schmitt et al., 2005). However,

to our surprise, there is no significant delay of the open-to-closed transition of IRER loci

flanking the reporter (loci 374, 377 in Figure 2-3) compared to wild type. In addition,

there is no detectable delay of the sensitive-to-resistant transition of the responsiveness

of reaper and hid to irradiation. This is likely because the DsRed transcription is

terminated by the SV40 polyA and does not extend into the neighboring region. The

insertion of the ubi-DsRed cassette has no noticeable impact on animal viability or

fertility and the reporter strain has been stably kept for over 50 generations in the lab.

Monitoring Cell and Tissue Specific Epigenetic Changes Using IRER{ubi-DsRed}

The IRER{ubi-DsRed} reporter allowed us to monitor the dynamic changes of

epigenetic status of IRER in individual tissues and specific cells. While essentially all

cells in the early stage embryo have strong DsRed signal, cells in the developing larvae

Page 47: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

47

display differential levels of DsRed. Some of the observed differences in DsRed

expression may due to cell type and/or differentiation status. For instance, in the

transgenic fly carrying a p{ubi-GFP} insertion on the second chromosome, GFP is

expressed ubiquitously from the entire disc (Figure 2-4 A-A’’). However, it is clear that

cells at the D/V boundary as well as the border of the wing pouch have relatively higher

levels of GFP than other cells. Compared to p{ubi-GFP}, the striking feature of

IRER{ubi-DsRed} expression is the variegated expression (Figure 2-4 B-B’’). In the wing

discs of IRER{ubi-DsRed}, patches of cells have DsRed levels that are not detectable,

indicating the reporter is totally silenced in these cells. For cells with detectable level of

DsRed, the level of expression appears significantly lower than p-based, or phiC31-

mediated insertion of ubi-DsRed into an euchromatin locus.

The variegated expression of IRER{ubi-DsRed} is not specific to wing discs; all

imaginal discs from the transgenic strain display variegated expression of the reporter

(Figure 2-5 A-D). While the silenced, i.e. IRER{ubi-DsRed} -negative, clones can be

observed at most positions of the discs, it seems they are more frequently occurred in

regions with relatively condensed chromatin, as indicated by the level of DsRed

expression in surrounding cells. The occurrence of patched DsRed- positive or negative

clones indicates that their progenitor cells may contain distinct DNA accessibilities at the

IRER locus, and that the epigenetic status of IRER is, to a certain extent, mitotically

stable during subsequent proliferation. While all discs in IRER{ubi-DsRed} larvae reared

under normal condition have DsRed -negative clones, the size of the clones, as well as

the overall area covered by the clones varies among discs. This suggests that the

epigenetic silencing of the reporter could happen within a relatively long time window of

Page 48: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

48

disc development. We did notice some small DsRed-positive clones completely

surrounded by DsRed-negative cells, which seems to suggest that it is possible to

reverse from a silenced state to an open state during normal development.

To verify that the variegated expression of IRER{ubi-DsRed} is due to epigenetic

silencing of the reporter, i.e. through a similar mechanism as that described for position-

effect variegation (PEV), we sought to knock down Su(var)3-9 in part of the wing disc

tissues. Su(var)3-9, or Suppressor of variegation 3-9, encodes a histone

methyltransferase responsible for catalyzing suppressive heterochromatin marks

H3K9Me2 and H3K9Me3 (Ebert et al., 2004). We have shown previously that the

function of Su(var)3-9 is required for blocking IRER accessibility during embryogenesis

(Zhang et al., 2008). When a double strand hairpin RNA targeting Su(var)3-9 mRNA is

specifically expressed in Serrate(Ser)-expressing cells in the wing discs using the UAS-

Gal4 system (Figure 2-4C), essentially all cells in the Ser domain have dramatically

increased level of DsRed expression (Figure 2-4D). In addition, there is no DsRed-

negative clone in the Ser domain upon Su(var)3-9 knockdown, indicating variegation is

abolished via reduced levels of Su(var)3-9 and consequently the repressive histone

mark H3K9Me3. To investigate whether the de-repression of IRER is a cell autonomous

response to Su(var)3-9 knockdown, we simultaneously labeled the RNAi-expressing

cells with a GFP transgene, and noticed that the IRER{ubi-DsRed} is primarily induced

from GFP-positive cells, suggesting a cell-autonomous de-repression of IRER (Figure 2-

4F-F’’). To rule out the possibility that the change of DsRed expression is due to

abnormal activation of the ubiquitin promoter, we examined the responsiveness of

p{ubi-GFP} on the 2nd chromosome to the knockdown of Su(var)3-9. Interestingly,

Page 49: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

49

ectopic GFP signal is induced from wing discs, with an induction pattern totally different

from that of IRER{ubi-DsRed} and seems not to overlap with the Ser-domain.(Figure 2-

5). The induction of p{ubi-GFP}, unlike the induction of IRER{ubi-DsRed}, probably

reflects an independent response to Su(var)3-9 knockdown. This further indicates that

the induced DsRed expression in Su(var)3-9 knockdown cells is mostly attributable to

de-repression of IRER. Moreover, IRER{ubi-DsRed} expression is also significantly

induced when dsRNA targeting HDAC3 is expressed in the same experimental setup,

suggesting the epigenetic silencing of IRER requires multiple histone modifiers (Figure

2-7).

As we have shown previously, an open IRER is responsible for mediating

immediate (within 15-30 minutes) induction of reaper and hid in early stage embryos

following gamma-irradiation (Zhang et al., 2008). Following development stage 12,

reaper and hid become irresponsive to irradiation in most cells due to the formation of

heterochromatin-like structure in IRER. Intriguingly, reaper mRNA is readily detected

from Su(var)3-9 RNAi discs via in situ hybridization. This suggests that when the DNA

accessibility of IRER is intensely increased, it is sufficient to drive cells to undergo

apoptosis. Yet another possibility is that cells cannot survive when the level of a

fundamental histone modifier such as Su(var)3-9 is down-regulated or totally abolished.

Further studies are required to differentiate the two possibilities. However, they are not

mutually exclusive with each other because IRER may serve as a key regulatory region

to define the cell fate by mediating its epigenetic status.

Monitoring Epigenetic Status of IRER at Organismal Level

As afore mentioned, the IRER{ubi-DsRed} reporter allowed us to monitor the

epigenetic status of IRER from individual tissues and cells. However, this requires a

Page 50: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

50

considerable amount of tissue dissection work. Since the DNA accessibility of IRER is

reflected by the expression level of DsRed reporter, we are able to “quantify” the

accessibility of IRER by directly measuring the fluorescent intensity of DsRed from the

whole animal.

The reporter strain can be used to screen for histone modifiers required for the

appropriate epigenetic regulation of IRER. In the previous study, we have identified

several histone modifiers, including Hdac1(rpd3), Su(var)3-9, and Pc , which are

required for the epigenetic silencing of IRER during embryogenesis (Zhang et al., 2008).

To test whether they still perform such a role in adults, we crossed their mutant strains

individually with the IRER reporter strain and measured the DsRed expression from

their progeny. Among all the tested mutants, three alleles of Hdac1 mutant ([303], [326],

and [328]), one Pc mutant (Pc3), and two Su(var)3-9 mutants ([1] and [2]) consistently

showed a modest but significant increase of DsRed signal from IRER{ubi-

DsRed}/Mutant fly, when compared to the IRER{ubi-DsRed}/+ control (Figure 2-8A).

Therefore, we concluded that these histone modifiers are required for epigenetic

silencing of IRER, similar to what has been observed in embryos. We hypothesize that

the homozygous mutants should further release the condensed chromatin and induce

still higher levels of DsRed. However, none of the histone modifier mutants tested is

homozygous viable, presumably due to their essential roles of conducting proper

epigenetic regulation during normal development. This may explain the vigorous

induction of DsRed from the Su(var)3-9 RNAi wing discs but not from the wing discs

heterozygous for Su(var)3-9 (data not shown). Nonetheless, the stable increase of

DsRed observed from the histone modifier mutants is statistically significant. The

Page 51: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

51

IRER{ubi-DsRed} reporter can also be used as a convenient tool to measure the DNA

accessibility of IRER at the organismal level, and screen for any environmental factors

or drug treatments sufficient to change the epigenetic status of IRER.

One unique feature of epigenetic regulation is that it is dynamic, responsive to

environmental and dietary factors, and under many circumstances, reversible.

Interestingly, our preliminary data showed that dietary restriction (DR), a reduction in

food intake without malnutrition, significantly induced DsRed expression (Fig 2-8B),

suggesting that the epigenetically regulated IRER is de-repressed at the organismal

level.

Recent studies have provided extensive evidence to show that DR can extend life

span of diverse organisms, including yeast, flies, rodents, and monkeys (Anderson et

al., 2009). Accumµating evidence suggests that the biological effects of DR are closely

related to chromatin function, although the underlying mechanisms are still unclear

(Heydari et al., 2007). Here we linked the DR to an epigenetically regulated enhancer

region that is required for stress response. This work will help us to better understand

how epigenetic regulation is responsible for the effects on lifespan by dietary restriction.

Discussions

Histone modifications, by controlling the expression profiles of different sets of

genes, play a pivotal role in defining distinct cellular properties over the otherwise

identical genome. Not only are specific histone modifications important for maintaining

the differentiation potential of stem cells, but also the dynamic change of epigenetic

status of specific sets of genes is one of the main mechanisms of cellular differentiation.

Misregulation of epigenetic status has been implicated in the etiology of a variety of

diseases such as mental disorders and cancers (Isles and Wilkinson, 2008; Johnstone

Page 52: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

52

and Baylin, 2010). Traditional biochemical methods of monitoring epigenetic status rely

on homogenizing millions of cells. In this work, we showed that a fluorescent reporter

could be applied to monitor the epigenetic status of a given locus in individual cells.

This not only allowed us to monitor the cellular specificity of epigenetic regulation, but

also enabled us to assess the DNA accessibility of the targeted locus without tissue

dissection or sacrificing the animal.

Epigenetic Regulation with Cellular Resolution

In any given tissue, the epigenetic status of a particular gene could vary

significantly depending on the cell type and/or differentiation status. For the

development of certain diseases, such as cancer, it is conceivable that the causative

epigenetic change may be initially restricted to a very small number of cells or even a

single cell. Monitoring epigenetic status at the cellular level is thus essential for us to

understand the mechanisms of how misregulation of epigenetic status may arise as a

consequence of development and aging, or following environmental stresses.

Cell specific epigenetic regulation was first observed with the variegated

expression of the white gene when it was transposed close to the centromere (Henikoff,

1992; Karpen, 1994). A UAS-lacZ reporter inserted into the regulatory region of bithorax

was used successfully to show that this region became inaccessible to Gal4 and the

transcription apparatus in cells in the anterior segments, where it is silenced by PcG-

mediated silencing, but remained open in cells at posterior segments (McCall and

Bender, 1996). In canonic regulation of homeotic genes such as the bithorax, the

formation of facultative heterochromatin is determined by the position (and identity) of

the cell and is largely irreversible in later development. However, there are many loci

whose epigenetic status depends on the differentiation status of the cell, or is controlled

Page 53: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

53

by a particular signal transduction pathway, or changes in response to particular stimuli.

For instance, PcG-mediated silencing/repression of differentiation regulators in stem

cells could be relieved in differentiating cells (Reik, 2007). And the expression of

oncogenic Ras could specifically induce the silencing of pro-apoptotic genes such as

Fas (Gazin et al., 2007b). Our previous analysis indicated that, unlike canonic PcG-

mediated suppression of homeotic genes, epigenetic regulation of IRER is not cell

lineage –dependent (Zhang et al., 2008). The IRER{ubi-DsRed} reporter allowed us to

show that its regulation is rather dynamic during development and is also responsive to

environmental factors.

The cellular resolution offered by this fluorescent reporter-based strategy also

highlighted the stochastic nature of epigenetic regulation, i.e. cells of the same tissue

and similar differentiation status may display different DNA accessibility at a particular

locus. This feature has been very well documented before with variegated expression of

the white gene, and hence variegated eye pigmentation. However, fluorescent reporter

such as IRER{ubi-DsRed} allowed this stochastic feature to be monitored not just in the

eye, but in essentially all tissues. This should provide a rich opportunity for us to

investigate the mechanisms that control/affect the stochastic regulation of epigenetic

status.

Limitations of the Strategy

There are certainly limitations to this strategy that need to be addressed in future

studies. First, the ubiquitin promoter we used in the reporter line is not truly “ubiquitous”.

Even in the attB{ubi-DsRed} control line, the expression levels of DsRed are not totally

even in every cell in the imaginal discs. The unevenness of the reporter system in

different cells will compound the association between the expression level of the

Page 54: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

54

reporter and the epigenetic status. This limitation could be partially addressed by using

a better ubiquitously expressed driver. We are in the process of generating another

reporter line using the tubulin regulatory region as a driver, which has been reported to

direct ubiquitous transcription and is sensitive to chromosomal position effects

(O'Donnell et al., 1994). In the new version of reporter, GFP is used to replace the

DsRed as a reporter gene, in case DsRed itself may have a cell-specific expression. If

we can observe a similar expression pattern of the reporter signal from the IRER{tub-

GFP} line, it will be a strong and independent verification of our strategy.

More importantly, the current method relies on monitoring the fluorescent reporter

signal from dissected tissues or homogenized animals. In order to follow the epigenetic

changes in normal development, we need to access the reporter signal in live animals

without disturbing normal development. It is possible to quantify the DNA accessibility of

the targeted region in live animals with the adaptation of a quantitative fluorescence

measurement technology. We have collaborated with the research group of Dr. Huabei

Jiang in the department of biomedical engineering, University of Florida, and developed

a method to quantitatively measure the DNA accessibility in live animals via

fluorescence molecular tomography (FMT) (data not shown).

Page 55: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

55

Figure 2-1. Generation of IRER{ubi-DsRed}. A) Schematic drawing of the strategy for inserting the ubi-

DsRed reporter into IRER via “Ends-out” homologous recombination. A donor strain carrying the targeting construct P(LA-ubi-DsRed.T4-SV40PolyA-RA) on X-chromosome was firstly generated. LA and RA were two ~3kb homologous sequence of IRER(coordinates based on genome release 4.3). B) Southern blot verification of the recombination. Probe was corresponding to the red bar in A. Hybridization of the p32-labeled DNA probe to the BglII-digested genomic DNA specifically recognized the 8kb recombined fragment from the IRER{ubi-DsRed} strain, confirming the successful recombination. BglII sites were labeled as arrows in A. C-F) Monitor the DsRed expression in embryos by RNA in situ hybridization with a DsRed probe. C) DsRed was expressed ubiquitously at high level in stage 11 embryos, which was in agreement with the fact that IRER is open and accessible in proliferating cells at this stage. D) Stage 11 embryos from the wild type control did not show any signal, indicating the high specificity of the DsRed probe. E) Starting from stage 13, when IRER has formed a condensed chromatin structure, the overall DsRed level was dramatically decreased. Notice that cells in the midline were still labeled (red arrow head). F) By the end of embryogenesis, the general DsRed mRNA level was steadily at a reduced level; salivary gland showed strong hybridization signal (red arrow). G-J) The restricted DsRed expression in adult ovary by fluorescent microscopy. G) GFP was ubiquitously expressed in every single cell in the control ovary: w[1118];P{w[+mC]=Ubi- GFP(S65T)nls}2R/CyO. H) In the IRER{ubi-DsRed} ovary, the DsRed expression (red) was restricted to only a few locations. Nuclear DNA was counter-stained by DAPI (blue). I) Higher magnification picture of the ovary germarium, showing bright DsRed signal at the anterior tip. J) Higher magnification picture of the stage 10B egg chamber, showing DsRed was specifically expressed from the polar cells (white arrow head), with TOTO-3 DNA counter stain (green). Scale Bars, 25µm. The southern blot in B) was performed by Dr. Nianwei Lin (former graduate student in the Zhou laboratory); the egg chamber staining in J) was provided by Bingqing Zhang and Dr. Brian Calvi in Department of Biology at the Indiana University.

Page 56: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

56

Figure 2-2. The expression of IRER{ubi-DsRed} from embryos and larvae. A) In early embryos, the reporter was expressed ubiquitously at high levels. B) In newly hatched larvae, the strongest reporter expression was detected from nerve cord cells (blue arrow). C) In day2 larvae, a group of cells at the posterior epidermis have distinct DsRed signal (blue arrowhead). The larvae were counter stained with DAPI.

Page 57: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

57

Figure 2-3. Verification of the IRER{ubi-DsRed} reporter for DNA accessibility and epigenetic status of IRER. A) Schematic representation of the IRER locus and DsRed reporter. The positions of DNA amplicons for quantification of DNase I sensitivity and ChIP assay were shown below the IRER map. B) DNase I sensitivity in IRER and the reporter in early and late embryogenesis, when the reaper gene induction is sensitive or resistant to DNA damage, respectively. Both IRER and DsRed loci showed high sensitivity to DNase I at the sensitive stage, and reduced sensitivity in the resistant stage. Note that the insertion of the ubi-DsRed reporter did not disrupt the closed conformation of its surrounding environment (i.e. that of 374 & 377) at late embryogenesis (resistant stage). C) Enrichment of H3K27Me3 and PSC binding in the reporter locus in late stage embryos. ChIP data was normalized against the recovery rate of the positive control locus bxd. D) Enrichment of H3K9Me3 and HP1 binding at the reporter locus in later stage embryos. ChIP data was normalized against the positive control locus H23 (centromeric heterochromatin). E) DsRed (+) cells were very sensitive to the stress associated with tissue disassociation and FACS. The level of reaper, presented as RNA/DNA ratio, was much higher in DsRed (+) cells than that in (-) cells. Experiments in D-E) were performed by the Dr. Nianwei Lin.

Page 58: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

58

Figure 2-4. Monitoring the change of epigenetic status of IRER in wing discs. A-A’’) In

the control wing discs: w[1118];P{w[+mC]=Ubi- GFP(S65T)nls}2R/ CyO, GFP was ubiquitously expressed. B-B’’) The mosaic pattern of DsRed expression from IRER{ubi-DsRed} wing disc. The expression of DsRed was under the control of not only ubi-promoter, but also the local environment of IRER, and exhibited a variegated expression. C-D) Cell-specific knock-down Su(var)3-9 in the Ser-expressing domain in wing discs led to de-repression of IRER, shown by intensely increased DsRed signal and abolished mosaic pattern, verifying that the variegated expression of IRER{ubi-DsRed} is due to epigenetic silencing of the reporter. E) Upon Su(var)3-9 knock-down, the pro-apoptotic gene reaper was induced from the Ser-domain, shown by in situ hybridization using a reaper probe. F-F’’) DsRed was induced predominantly in Su(var)3-9 RNAi cells (see the overlapping between DsRed (red) and Ser>GFP (green)).

Page 59: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

59

Figure 2-5. The expression pattern of the ubi-DsRed reporter is largely controlled by the

local chromosome environment. When inserted into the epigenetically regulated IRER, the reporter shows a variegated expression from A) wing discs, B) eye discs and C) haltere discs in third instar larvae. D) The variegated expression pattern is also observed from imaginal discs at earlier developmental stage. When the ubi-DsRed construct was inserted into a relatively open locus on 2nd chromosome, the DsRed singal is fairly ubiquitous from all examined tissues. E) wing discs; F) eye discs; G) haltere discs.

Page 60: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

60

Figure 2-6. Su(var)3-9 knockdown-induced DsRed expression is not due to abnormal activation of the ubiquitin promoter. Using a Ser-Gal4 driver, the construct of UAS-Su(var)3-9 dsRNA was expressed from either IRER{ubi-DsRed} wing disc or Ctr{ubi-GFP} wing disc. A) Ser-expression pattern in wing disc was labeled by UAS-GFP. B) Upon Su(var)3-9 knockdown, IRER{ubi-DsRed} was primarily induced from Ser-expressing cells. Compare the DsRed signal in B with the GFP signal in A. C) Interestingly, ubi-GFP was also induced by Su(var)3-9 knockdown. The induced GFP signal was mainly distributed in the center of the wing porch and hinge, but not from Ser-expressing cells. This suggests a non-cell autonomous induction of ubi-GFP following Su(var)3-9 RNAi.

Page 61: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

61

Figure 2-7. The expression of IRER{ubi-DsRed} is responsible to HDAC. We tested a list of RNAi strains for their effect on alternating DsRed expression, which reflects the role on epigenetic regulation of IRER. Among the histone modifiers we have tested, B) Su(var)3-9- and C) HDAC3- RNAi caused induction of DsRed signal, suggesting that they are required for epigenetic silencing of IRER during development. We are still in the process of testing additional histone modifiers with more RNAi constructs for their potential role in regulating local DNA accessibility of IRER.

Page 62: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

62

Figure 2-8. Monitoring the epigenetic status of IRER in different genetic backgrounds

and in response to dietary restriction (DR). A) Flourometric measurement of DsRed was performed on males eclosed within 1 day. The DsRed value from the mutant strains IRER{ubi-DsRed}/mutant was normalized to that from the control strain IRER{ubi-DsRed}/w[1118]. Interestingly, DsRed was increased from several histone modifier mutants, including HDAC1, Pc, and Su(var)3-9, indicating their requirement for the reporter gene silencing. *, p<0.05 from unpaired student t-test. B) The IRER{ubi-DsRed} was induced by dietary restriction (DR) in both males(blue) and females(red). DsRed was measured from adults at different time points (0/3/6/9 day) after feeding with either DR food(10S0Y) or control food(SY). The DR-induced DsRed change was calculated by dividing the DsRed value from 10S0Y-feeding animal by that of a SY-feeding animal. Solid line, IRER{ubi-DsRed}, carrying an ubi-DsRed within IRER; Dash line, attB{ubi-DsRed}, carrying an ubi-DsRed in a different genomic locus. The DR-induced DsRed expression was only observed from IRER{ubi-DsRed} but not attB{ubi-DsRed}, indicating the DsRed induction requires the function of IRER.

Page 63: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

63

Table 2-1. List of RNAi lines used for the screen of epigenetic modifiers of IRER

RNAi target gene

VERC stock # Wing morphology IRER{ubi-DsRed} signal in wing disc

dHDAC3 20814 Small, curled wing Moderate increase

dHDAC1 30600 Small, curl a little Moderate increase

E(z) 107072 Normal No change

Lid 103830 normal size, curl a little No change

Hpo 104169 Severely curled No change

UTX 105986 Normal No change

Su(var)3-9 39378 Lethal Strong increase

Su(z)12 42423 Normal No change

CBP 104064 Normal No change

UAS-GFP (Ctr) Normal No change

In order to identify the factors required for establishing and/or maintaining the epigenetic status of IRER, individual UAS-RNAi lines were crossed with Ser-Gal4; IRER{ubi-DsRed} to knock down target genes from Ser-expressing cells in wing discs. The wing disc DsRed signal and wing morphology of the progenies were compared with that in the control (Ser-Gal4>UAS-GFP). Through this round of screening, Su(var)3-9. HDAC1 and HDAC3 were identified as potential key repressors for IRER.

Page 64: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

64

CHAPTER 3 AN EPIGENETICALLY REGULATED INTERGENIC REGION CONTROLS ORGAN

SIZE AND MEDIATES DMYC -INDUCED APOPTOSIS IN DROSOPHILA

Introduction

Cell autonomous apoptosis following Myc over-expression has been regarded as a

major tumor-suppression mechanism (reviewed by (Meyer et al., 2006)). Suppression of

apoptosis, by means such as overexpression of Bcl-2, is required for unleashing the

tumorigenic potential of Myc in mammalian cells (Letai et al., 2004; Pelengaris et al.,

2002; Strasser et al., 1990). Whether cells with elevated level of Myc undergo apoptosis

depends on the availability of growth factors such as IGF (insulin-like growth factors)

(Harrington et al., 1994). It has been postulated that Myc-induced cell autonomous

apoptosis reflects a fundamental mechanism that maintains tissue homeostasis by

inducing apoptosis when overproliferation is sensed. However, the mechanistic details

of this mechanism remain enigmatic.

One extensively studied pathway implicated in Myc-induced cell autonomous

apoptosis is the P53 –mediated activation of pro-apoptotic genes and/or suppression of

anti-apoptotic genes (reviewed by (Hoffman and Liebermann, 2008)). Many important

pro-apoptotic genes, such as apaf-1 and caspase 9, have been implicated in Myc-

induced apoptosis in cell culture systems. However, study with animal models

suggested that Myc-induced cell death can proceed in the absence of apaf-1 or

caspase 9 (Scott et al., 2004). It is also clear that at least under some circumstances,

Myc –induced cell death can proceed without the participation of P53.

Despite its enigmatic nature, the mechanism of Myc-induced apoptosis appears to

be highly conserved. Overexpression of dMyc, the only Myc ortholog in Drosophila, also

induces cell autonomous apoptosis (de la Cova et al., 2004; Montero et al., 2008).

Page 65: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

65

Intriguingly, while the level of dP53 (Drosophila ortholog of mammalian tumor

suppresser P53) mRNA is significantly increased following dMyc expression, the

function of dP53 appears to be dispensable for dMyc-induced cell death in Drosophila

(Montero et al., 2008). Ectopic expression of dMyc leads to increased cell size but fails

to result in significant hyperplasia on its own (Johnston et al., 1999). Moderate tissue

overgrowth was only observed after dMyc -induced apoptosis was blocked by co-

expression of the viral caspase inhibitor P35 (Montero et al., 2008). This indicates that,

similar to what was observed with mammalian tumorigenesis models, blockage of

apoptosis is required for dMyc –induced hyperplasia in Drosophila. In this study, we

showed that the induction of apoptosis following dMyc –induced overproliferation

requires a highly conserved intergenic regulatory control region in the RHG (reaper, hid,

and grim) genomic block.

The ~ 33kb intergenic region was originally named as IRER (irradiation responsive

enhancer region) since it was found to be required for mediating the induction of reaper,

hid, and sickle following ionizing irradiation of the embryo (Zhang et al., 2008). This

region contains a previously identified response element for Drosophila P53 (Brodsky et

al., 2000). Interestingly, the epigenetic status of this region undergoes a dramatic

change at embryonic development stage 12, when most cells enter into post-mitotic

differentiation. During the transition, this region becomes enriched for H3K27me3,

H3K9me3, and bound by Polycomb group (PcG) proteins as well as HP1

(Heterochromatin Protein 1). Consequently, the DNA in this region becomes as

inaccessible to DNase I as the pericentromeric heterochromatin region. This epigenetic

blocking of IRER specifically renders the pro-apoptotic genes irresponsive to ionizing

Page 66: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

66

irradiation while other branches of the DNA damage response, such as the DNA repair

pathway, remain active (Zhang et al., 2008).

In this work, we found that the IRER is required for mediating overproliferation -

induced apoptosis and plays a pivotal role in maintaining tissue homeostasis during

development. Animals lacking IRER are viable but display tissue hyperplasia in several

organs, indicating it is required for controlling cell numbers during development. The

functional significance of this regulatory control region is heightened in the context of

oncogenic stress. IRER is required for the induction of apoptosis associated with

oncogene induced overproliferation. While overexpression of dMyc alone failed to

induce hyperplasia, it led to significant overproliferation in cells lacking IRER.

Materials and Methods

Drosophila Strains and Culture

Flies were maintained on a standard corn-agar medium at 25°C, except otherwise

mentioned. The strains used in this study were described in supplementary table I.

Irradiation was performed as described previously (Zhang et al., 2008).

Clone Induction

The IRER mutant clones were generated with either the standard FLP/FRT system

or the MARCM system (Morata and Ripoll, 1975; Wodarz, 2000). Hsp-flp and ey-flp

were used to induce clones from imaginal discs and adult eyes, respectively. Clone size

was determined by measuring the 2-dimensional area of clones at 200X magnification.

Immunohistochemistry and Microscopy

Imaginal discs were dissected from third instar larvae in 1XPBS, fixed in 4%

paraformaldehyde for 25 min at room temperature, washed three times with 1XPBT for

5min each, counter-stained with DAPI (1ug/mL) and mounted on slides with Vectashield

Page 67: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

67

HardSet mounting medium (Vector laboratories). To detect the apoptotic cells, fixed

discs were incubated with blocking buffer (5% normal goat serum in PBT) for 30min~1hr

at room temperature, followed by incubation with the anticleaved Caspase 3 antibody

(Cell signaling, 1:200) for 2hr at room temperature or overnight at 4°C, and incubation

with the Alexa Fluor 488/568 Goat anti-Rabbit IgG secondary antibody (Molecular

Probes, 1:400) for 2hr at room temperature or overnight at 4°C.

In situ hybridization was carried out as described (Zhang et al., 2008). FISH signal

was detected by HRP-conjugated anti-DIG antibody (1:500, Roche) and subsequently

amplified by the Tyramid Signal Amplification Kit (PerkinElmer, Waltham, MA, USA).

Fluorescent images were taken with a Zeiss Axioplan imaging 2 microscope or a

Leica SP5 Confocal Microscope. The Open lab software was used to acquire the

images. Clone size was measured at 200X magnification in Photoshop (Adobe).

Scanning electron microscopy (SEM) was performed as described (Sullivan et al.,

2000). Images were taken at ×1000 magnification.

Wing Size Measurement

Flies were raised at a standard density (~20 flies per vial) and three replicate vials

were established for each genotype. Both wings were dissected from adult flies and

mounted onto slides in Permount mounting medium (Fisher Scientific); only one wing

from each fly was used for size measurement and the other one served as back up.

Wing images were acquired using the Leica DMLB light microscope and wing area was

determined using methods as described (Gilchrist and Partridge, 1999).

Gene Expression Analysis

RNA was extracted from ~20 wing discs or ~10 larvae of the desired genotypes

using the RNeasy Minikit (Qiagen). cDNA was synthesized with the high-capacity cDNA

Page 68: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

68

archive kit (Applied Biosystems). Quantitative PCR was run using the 7500 fast real-

time PCR system (Applied Biosystems) and signal was detected based on SYBR Green

intensity (SYBR Green Master Mix from Applied Biosystems). 10ng cDNA was used as

template for each quantitative PCR reaction in duplicates.

Statistics

Wilcoxon rank-sum test was used to compare the difference between clone size

and twin spots. In other cases, the data are subject to normal distribution and the

student’s t-tests were used to determine the statistical significance.

Bioinformatics

Hi-C chromatin interaction: The Hi-C data for D.melanogaster embryos (16-

18hr) was taken from a recent genome-wide study of three-dimensional chromatin

interactions (Sexton et al., 2012). The data in the 600kb region (Ch3L: 18,000,001-

18,600,000) with the resolution of 10kb bin was used to generate the heat-map. The

contact enrichment was calculated in the same way as described in Sexton et al. And

the heat-map was generated with R (R version 2.9.2, R Development Core Team,

2009).

dMyc and P53 binding sites: Due to the unavailability of the Myc and P53

binding motifs in Drosophila, we took the motifs for cMyc and P53 in vertebrates from

TransFac dataset (Release 2012.1) (Issigonis et al., 2009). We applied MATCH

algorithm (Kel et al., 2003) with the “minFP” (minimal false-positive) setting to identify

the potential binding sites for each motif in the RHG region. Then the identified binding

sites were pooled together for Myc (V$CMYC_01, V$CMYC_02) and P53 (V$P53_01,

V$P53_02, V$P53_03, V$P53_04, V$P53_05).

Page 69: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

69

Results

IRER Mediates DNA-damage-induced Pro-apoptotic Gene Expression in Post-embryonic Tissues

Our previous work revealed that IRER is required for mediating irradiation-

induced reaper, hid, and sickle expression in embryos before stage 12 (Zhang et al.,

2008) (Fig. 3-1A). In embryos deficient for this intergenic region, i.e. homozygotes of

Df(IRER), the transcriptional response of the three pro-apoptotic genes to DNA damage

is totally blocked. While the irradiation responsiveness of these three pro-apoptotic

genes is robust in stage 9-11 embryos, it is diminished in most cells in embryos past

developmental stage 12. This sensitive-to-resistant transition is due to a targeted

epigenetic regulation that requires the function of Polycomb group (PcG) proteins and

histone deacetylase (HDAC) (Zhang et al., 2008).

To test whether IRER is also required for DNA damage- induced cell death in post-

embryonic tissues, we measured irradiation -induced caspase activation and pro-

apoptotic gene expression in the developing imaginal discs of Df(IRER). It has been

well documented that exposure of Drosophila larvae to irradiation induces a rapid and

wide-spread apoptosis in imaginal discs (Brodsky et al., 2000; Ollmann et al., 2000).

Specifically, it has been shown that the significant increase of apoptotic cells in the wing

disc at 4 hours following irradiation is dependent on the function of dP53 (Wichmann et

al., 2006). We subjected third instar larvae to similar treatment, i.e. 40 Gy of γ-ray.

Indeed, we found that at 4 hours post irradiation, there is a substantial increase in the

amount of apoptosis in the wild type wing discs, preferentially at the wing margin (Fig. 3-

1C vs. B). However, in sharp contrast, there is little detectable increase of caspase

activation in discs from animals homozygous to Df(IRER) (Fig. 3-1E vs. D).

Page 70: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

70

We then measured the mRNA levels of the RHG genes by quantitative PCR. In a

time course analysis, we found that the induction of reaper and hid was highest

between 1 and 2 hours following irradiation (Fig. 3-1F). At this time interval, irradiation –

induced expression of reaper and hid was significantly lower in wing discs homozygous

to Df(IRER), when compared to the wild-type control (Fig. 3-1G). The levels of sickle

and grim mRNAs in the wing discs were rather low and barely detectable even after

irradiation. We noticed that, unlike the embryo, the induction of reaper and hid was not

completely blocked in the wing discs. It has been observed that a P53-indpendent

mechanism also contributes to irradiation/DNA damage -induced cell death in the wing

disc. In animals mutated for dp53 or chk2, such a mechanism could eventually

compensate for the loss of dP53 or Chk2 function and induce apoptosis at a much later

(12-16hr post IR) time point (Wichmann et al., 2006). It is possible that the incomplete

block of reaper/hid induction in the wing discs reflects the existence of such a

mechanism in wing disc cells. However, it is clear that the significant suppression of γ-

ray induced reaper/hid expression in Df(IRER) was sufficient to result in a significant

decrease of apoptotic cells, i.e. cells with activated caspase 3, at 4 hours post

irradiation. Therefore, we conclude that IRER also mediates DNA damage –induced

pro-apoptotic gene expression and cell death in post-embryonic tissue.

The cis Regulatory Function of IRER is Required for Tissue Homeostasis and Organ Size Control

During the process of analyzing γ-ray induced cell death in the developing

imaginal discs, we noticed that discs from homozygous Df(IRER) animals are

conspicuously larger than those with wild type IRER or heterozygous to the deficiency

(Fig. 3-2A-B & D-E). This is accompanied by a decreased level of apoptotic cells in the

Page 71: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

71

wing disc even without any irradiation treatment. A detailed statistical analysis revealed

that both eye and wing imaginal discs from homozygous Df(IRER) larvae are

significantly larger than those from their heterozygous littermates (Fig. 3-2C&F). Close

inspection of the discs indicated that the size of the cell in the Df(IRER) discs, reflected

both as the distance between DAPI –stained nuclei as well as density of cells within a

given area, was indistinguishable from that of the heterozygous discs. Thus it appears

that there are more cells in the developing discs lacking the function of IRER.

To make sure that the observed increase in imaginal disc size was not due to

changes in the timing of proliferation or developmental cell death, we further evaluated

the size of the adult wing. About 10-20% homozygous Df(IRER) animals survive to

adulthood. The wings of these Df(IRER) homozygous flies are significantly larger than

those of wild type animals raised in parallel or heterozygous animals developed in the

same vial (Fig. 3-3A’). When different compartments of the wing were monitored, it

appeared that both anterior and posterior compartments of the wing overgrew with the

change being relatively more pronounced in the posterior compartments (Fig. 3-4A-B).

The increased size of the wing was not due to increased cellular size but purely

due to the increased number of cells that contributed to the adult wing. When the

densities of the trichome (one from each wing cell) were measured, to our surprise, the

density was slightly higher in Df(IRER) wing than those of wild type or heterozygous to

Df(IRER). The difference was statistically significant for the posterior compartments

(Fig. 3-3B). When corrected for this difference in cell density, wings from homozygous

Df(IRER) animals have about 10% more cells than the wings of wild type animals.

Page 72: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

72

To rule out the possibility that the phenotype was due to background mutation on

the Df(IRER) chromosome, complementation test was performed with well-defined

deficiencies of this region (Fig. 3-3C). Both Df(ED224) and Df(ED225) were generated

by the DrosDel project with defined break points and in isogenic background (Ryder et

al., 2004). While both deficiencies are homozygous lethal, the transheterozygous with

Df(IRER) are viable. The size of the wings of Df(IRER)/Df(ED225) animal is about the

same as Df(IRER) homozygous. In contrast, the overgrown phenotype was partially

complemented by the Df(ED224), which lacks the transcribed regions of reaper, grim

and hid, but has the IRER region intact (Fig. 3-1A). This seems to suggest that either

the expression of sickle can contribute to the size control or that IRER can act in trans

to regulate the expression of reaper and/or hid. Since the genetic background of

Df(ED225) is independent of that of Df(IRER), we conclude that the observed

overgrown phenotype is due to the lack of the genomic region of IRER.

The abnormality of organ size control is due to the cis regulatory function of IRER.

There is no predicted coding or non-coding gene in the IRER genomic region

(Flybase.org). In addition, no transcript with coding potential has been detected in this

region by any RNA sequencing projects (based on data collected by Flybase or

ModENCODE). To rule out the possibility that the phenotypes associated with Df(IRER)

might be due to a low level of non-coding transcripts from IRER, we cloned the genomic

region encompassing the IRER as a potential rescue construct (Fig. 3-1A). A BAC clone

containing the genomic region Chr 3L:18391335–18432601, corresponding to the span

from just before the start of Reaper ORF to the stop codon of Sickle (Fig. 3-1A), was

used to make a transgenic fly via phiC31 integrase -mediated transformation. Animals

Page 73: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

73

carrying the 2nd chromosome that contains this reaper-sickle interval fragment appear to

be normal without any noticeable change in viability or organ size. When this

chromosome was recombined with the Df(IRER) chromosome, it failed to rescue the

partial lethality and increased wing size associated with homozygous Df(IRER) (Fig. 3-

3D). Homozygous Df(IRER) flies carrying this rescue construct have enlarged wings

indistinguishable from those without the rescue construct. From these evidences, we

concluded that the cis regulatory function of IRER is required for controlling tissue

homeostasis.

To monitor how the lack of IRER cis regulatory function affects pro-apoptotic gene

expression in development, we compared the expression of the four pro-apoptotic

genes, reaper, hid, grim, and sickle, between wild-type and homozygous Df(IRER)

larvae. We found that the level of reaper mRNA was significantly lower (≈ 50%) in

Df(IRER) mutant larvae. The level of sickle was reduced by about 60%, while the levels

of hid and grim were not significantly changed (Fig. 3-3E). The level of reaper mRNA in

Df(IRER) wing discs was reduced to similar extent as that in the whole larvae (Fig. 3-

3F). However, the level of sickle in the wing discs was too low to be reliably measured

with QPCR. Similarly, the level of hid mRNA in the wing discs was not significantly

changed. The level of grim was also too low to be detectable from the wing discs. The

measurement of RHG mRNA levels was normalized against housekeeping genes rp49

or GAPDH. The trends as well as the magnitude of changes do not seem to be

significantly affected by the choice of standard genes.

To observe whether IRER is also responsible for mediating developmental cell

death in other tissues, we monitored developmental cell death in the embryonic CNS.

Page 74: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

74

We noticed that about 10% of the homozygous embryos have an enlarged nervous

system (Fig.3-5 A-B). Comparing the nervous system phenotype of Df(IRER) with that

of H99 suggests that most RHG –mediated developmental cell death likely proceeds

normally without the function of IRER (White et al., 1994; Zhou et al., 1995). Within the

embryonic system, the programmed cell death of the midline glial cells has been shown

to be dependent on the EGF signals provided by the axons (Bergmann et al., 2002).

Using the slit1.0-lacZ reporter that labels the ensheathing midline glia, we noticed that in

stage 16-17 embryos homozygous to Df(IRER), there were extra slit1.0-lacZ -positive

cells present in abdominal segments (Fig. 3-5C-D). This suggests that IRER function is

also required for mediating this EGF –dependent cell death of the midline glia.

The pattern of cells expressing reaper and sickle in developing tissues is dynamic,

which is partly due to the fact that cells expressing these genes are quickly removed via

apoptosis. The expression pattern of these two genes, especially reaper, seems to be

sporadic in many developing tissues such as the wing disc. Since it is impossible to

predict which cells in the developing wing will express reaper, we turned to the embryo.

We noticed that in places where the expression of reaper is cell lineage -specific, such

as in neuroblasts in post stage 14/15 embryos, there is no detectable difference in

Df(IRER) homozygous embryos. In contrast, the segmentally repeated expression of

reaper at the epidermis was significantly weakened in animals lacking IRER (Fig. 3-6).

It’s interesting to note that it has been very nicely demonstrated that the number of

epidermal cells that can survive in the embryonic epidermis is strictly dependent on the

EGF signaling (Parker, 2006).

Page 75: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

75

Taken together, our observations indicate that the intergenic region of IRER plays

a fundamental role in regulating the appropriate number of cells in several tissue

compartments. This function is likely achieved by activating/increasing the expression of

pro-apoptotic genes in response to increased cell number and consequently decreased

availability of growth factors. The impact of losing IRER appears to be more significant

in tissues that have developmental cell death regulated by growth hormones as

opposed to tissues where developmental cell death is largely cell-lineage-specific.

IRER is Required for the Induction of Apoptosis following dMyc-induced Overproliferation

The fact that IRER is required for mediating appropriate cell number during tissue

development prompted us to see whether IRER is also involved in mediating the pro-

apoptotic response following dMyc -induced overproliferation. Similar to what has been

observed for c-Myc in mammalian systems, overexpression of dMyc in developing

Drosophila tissues leads to increased metabolism, increased cell size, and

overproliferation (reviewed in (Gallant, 2009)). The dMyc -induced overproliferation is

quickly followed by induction of apoptosis which essentially cancels out the impact on

final number of cells. For instance, when dMyc was expressed in differentiating eye

cells driven by GMR-Gal4, there was a significant increase of cell proliferation as

documented by incorporation of BrdU (Montero et al., 2008). However, the

overproliferation was quickly followed by increased TUNEL-signals among dMyc -

expressing cells. Overall, GMR –driven dMyc (GMM) has little noticeable effect on the

final number of cells in the eye. These reported observations were reconfirmed when

we established the system with GMR-Gal4 and UAS-dMyc (Fig. 3-7A&H).

Page 76: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

76

The cis regulatory function of IRER is required for overproliferation -induced

apoptosis in the GMM model. Under normal conditions, there is no detectable defect in

the eyes of surviving homozygous Df(IRER) adults (Fig. 3-7B), suggesting that either

there is a minimal level of developmental cell death in the eye, or the developmental cell

death can proceed in the absence of the regulatory function of IRER. As previously

reported (Montero et al., 2008), we found that co-expressing P35, the viral caspase

inhibitor, marginally enhanced the hyperplasia induced by GMM (Fig. 3F). However,

when dMyc was over-expressed in eye discs lacking IRER, there was a massive

increase of the size of the adult eye (Fig. 3-7C & I). The superfluous ommatidia caused

disorganization of eye, which was never seen with GMM alone. This enhancer effect

was noticeable even in the animals heterozygous to Df(IRER) (Fig. 3-7D). The dominant

enhancing effect of Df(IRER) was similar to that observed for heterozygous Df(ED225)

(Fig. 3-7E), a ~450kb deletion that covers the transcribed regions of grim, reaper,

sickle, and more than 10 other genes (Fig. 3-1A). These observations strongly suggest

that IRER plays a central role in regulating the expression of pro-apoptotic genes in the

context of GMM -induced overproliferation.

It has been shown cell autonomous apoptosis following dMyc –induced

overproliferation is accompanied by the induction of reaper and sickle (Montero et al.,

2008). Using fluorescent in situ hybridization (FISH), we found that reaper is

significantly induced in GMM eye disc. Cells with significantly elevated reaper mRNA

are posterior to the morphogenetic furrow (Fig. 3-7K), consistent with previously

reported observation of apoptotic cells in this model. This dMyc -induced cell

Page 77: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

77

autonomous expression of reaper was totally absent in discs homozygous to Df(IRER)

(Fig. 3-7L).

It has been documented that constitutively elevated level of dMyc introduced by

tubulin>dMyc (Tub-dMyc) leads to an increased size of the wing (de la Cova et al.,

2004), mostly due to the increased cellular size instead of cell numbers. When Tub-

dMyc was crossed into the Df(IRER) background, we found that the wing sizes were

significantly larger than either Df(IRER) or Tub-dMyc alone (Fig. 3-7M). Significantly

more cells survived when IRER is absent in dMyc- overexpressing wings.

To make sure that the observed effect of Df(IRER) on dMyc -induced cell death is

indeed cell-autonomous, we generated clones of cells deficient for IRER and

simultaneously over-expressing dMyc via the MARCM strategy (Morata and Ripoll,

1975) (Fig. 3-8A). Clones of Act-Gal4>UAS-dMyc;IRER(-/-)Gal80(-/-) cells (Fig. 3-8D-E)

were significantly larger than that of Act-Gal4>UAS-dMyc;IRER(+/+)Gal80(+/+) (Fig. 3-

8B-C). Taken together, our observations indicate that IRER is required for the induction

of apoptosis, and consequently the suppression of hyperplasia, following dMyc -induced

overproliferation. This is not limited to a particular tissue, but appears to be true for a

variety of tissues examined in this study.

Cells with Relatively Open IRER are More Sensitive to dMyc-induced Cell Death

Our previous analyses have shown that the accessibility of IRER in developing

embryos is subject to epigenetic regulation (Zhang et al., 2008). In cells with open

IRER, reaper, sickle, and hid can be dramatically induced within 15-30 minutes

following ionizing irradiation. In contrast, none of these three genes can be induced

when IRER forms a heterochromatin -like structure enriched with both H3K27me3 and

H3K9me3. To monitor the accessibility of IRER in individual cells, an ubiquitin-DsRed

Page 78: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

78

reporter was inserted into IRER through homologous recombination (Fig. 3-9A). This

reporter, IRER{ubi-DsRed}, gives relatively low and variegated expression in developing

wing discs (Fig. 3-10A). When Su(var)3-9, which is required for epigenetic blocking of

IRER during embryogenesis, was knocked down via RNAi, the reporter was

dramatically de-repressed (Fig. 3-10B). The expression of IRER{ubi-DsRed} is also

sensitive to environmental stress such as irradiation (Fig. 3-11) and nutrition.

It has been shown that dMyc driven by dpp-Gal4 at the anterior/posterior boundary

of the wing discs induces apoptosis within the dMyc -expressing zone as well as the

zone immediately anterior to the dMyc -expressing zone (de la Cova et al., 2004). We

reproduced the system in the presence of IRER{ubi-DsRed}. We noticed that, as

reported, the sizes of cells with dMyc overexpression are noticeably larger than the

neighboring cells, which is reflected by the diluted DAPI signal intensity (Fig. 3-9C-D).

Surprisingly, there is a conspicuous lack of DsRed positive cells in the dMyc expression

zone as well as the zone immediately anterior to it. In contrast, the variegated patterns

of DsRed in other regions of the dpp>dMyc wing discs are indistinguishable from those

in discs without dMyc overexpression (Fig. 3-9C’ vs. B’). The conspicuous lacking of

IRER{ubi-DsRed} expressing -cells is not specific to dpp>dMyc. Similar phenomenon

was observed when clones of dMyc -expressing cells were generated with Act>y>Gal4;

UAS-dMyc (Fig. 3-9E-E”).

To verify that cells with higher levels of DsRed, and presumably more accessible

IRER, are preferentially eliminated by dMyc -induced cell autonomous cell death, we co-

expressed Diap1 in those cells that have dpp-Gal4 expression. The variegated pattern

of DsRed signal in the dpp zone was largely restored by co-expressing of Diap1(Fig. 3-9

Page 79: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

79

D-D’’), indicating that the absence of DsRed -positive cells in this zone without Diap1

was due to preferential elimination of cells with relatively open IRER. This observation

supports the hypothesis that epigenetic regulation of IRER plays an important role in

controlling cellular sensitivity to overproliferation –induced cell death.

Cells Lacking the cis Regulatory Function of IRER have the Propensity to Overgrow and are Resistant to Stress-induced Cell Death.

To mimic the behavior of cells with completely blocked IRER in proliferating

tissues, we generated mosaic clones bearing IRER-deletions in the developing imaginal

discs. The heat shock -induced FLP system was titrated to generate about 1 clone per

disc, which allowed us great confidence in identifying and measuring the size of the

clones and the simultaneously generated twin spots. As shown in Figure 3-12, control

(wild type) clones and their twin spots grow to similar size in either the wing or the eye

discs (Fig. 3-12A & C). In contrast, clones lacking either the left section of IRER

(Df(IRER_left)) (Fig. 3-12B & D), or the entire IRER (Df(IRER)), have a great propensity

to significantly overgrow their respective twin spots. Although there is considerable

variation in how much the mutant clones overgrow their twin spots, the overall

differences are statistically significant (open bars in Fig. 3-12E & F; Fig. 3-13).

The propensity of IRER mutant clones to overgrow (wild-type counterparts) was

significantly heightened when cytotoxic stress was present. When growing animals were

subjected to a non-lethal dose of IR (10Gy) at 24 hrs following clone induction, the ratio

between the size of the IRER deficient clones and their respective twin spots was

greatly increased, both in the wing discs (Fig. 3-12E & G) and the eye discs (Fig. 3-12F

& Fig. 3-13A). Under this heightened stress condition, we noticed that in about 15% of

the cases, the wild-type twin spots had all been eliminated, leaving only the IRER

Page 80: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

80

mutant clones at the end of larval development. Although these instances were not

included in calculating the ratio of sizes between mutant clone vs. twin spot, the

overgrown propensity of both Df(IRER_left) and Df(IRER) clones was still statistically

significant (Fig. 3-12E-F, closed bars).

The overgrown propensity of cells lacking the cis regulatory function of IRER is

accompanied by resistance to stress -induced cell death. When irradiation- induced cell

death was monitored in discs containing Df(IRER) clones at 4hrs post γ-ray treatment

(40Gy), it is clear that, while there are abundant apoptotic cells in the twin spots or the

heterozygous tissues, there is a conspicuous lack of activated caspase 3 signal in the

clones deficient for IRER (or IRER_left) (Fig. 3-12H).

The RHG Genomic Regulatory Block Contains Consensus Myc Binding Sites

The RHG genes reside in a ~250kb synteny that is marked by unusually long

intergenic regions surrounding reaper and grim as well as an unusually high level of

conservation of nucleotide sequences within these intergenic regions (Lin et al., 2009).

It is interesting to note that when sequences from distantly related Drosophila species

are considered, nucleotide sequences within IRER are better conserved than those

underlying the reaper coding region (Zhang et al., 2008). IRER is enriched for highly

conserved non-coding elements (HCNEs) (Engstrom et al., 2007; Lin et al., 2009).

Functional studies in both vertebrates/mammals and insects have shown that HCNEs

often function as long range enhancers to regulate complex temporal and spatial

expression patterns of target genes (Pennacchio et al., 2006; Woolfe et al., 2005).

Direct evidence that the genomic region encompassing the RHG genes forms a

regulatory block was provided by recent whole genome Hi-C analysis (Sexton et al.,

2012). By analyzing the data set generated by Sexton et al, it became clear that the

Page 81: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

81

previously observed synteny correlates very well with a domain defined by

chromosomal contacts (Fig. 3-14A). Extensive chromosomal contacts exist within this

region, which for simplicity, we will refer to as the RHG genomic regulator block.

Independent of this whole-genome Hi-C analysis, 3C analysis conducted by us also

revealed that there are extensive interactions between the IRER and the RHG gene

promoters. However, what remains to be addressed is whether the pattern of

chromosomal interactions within the RHG genomic regulatory block is the same among

different cells, or likely to be dynamic depending on the cell type or differentiation status.

Bioinformatics analysis also revealed that there are binding sites for Myc and P53 in

IRER (Fig. 3-14B). Unfortunately, we were not able to experimentally verify the

predicted binding sites due to the unavailability the dMyc antibody suitable for ChIP.

The mechanism of interaction between dMyc and IRER (e.g. direct or indirect

interaction) needs to be explored in further study.

Discussions

Our study provided evidences indicating that the regulatory region IRER in the

RHG genomic regulatory block plays a pivotal role in controlling apoptosis to maintain

tissue homeostasis. Animals lacking this regulatory region are viable but have

overgrown organs, suggesting cells lacking IRER have decreased sensitivity to growth

factor levels or other intrinsic cell number controlling mechanisms. The regulatory

function of this region is also required for mediating the induction of apoptosis, and

suppression of hyperplasia, following dMyc overexpression in a variety of tissues. The

fact that the accessibility of IRER is subject to epigenetic control implies that cellular

sensitivity to proliferation induce apoptosis, and consequently the balance between

Page 82: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

82

proliferation and cell death, is subject to epigenetic mechanisms which in turn are

regulated by differentiation status and/or environmental factors.

IRER Serves as the Gatekeeper for Overproliferation-induced Apoptosis

A tenacious link between proliferation and apoptosis has long been observed in

both developmental and pathological (tumorigenesis) settings (Reviewed in (Hipfner

and Cohen, 2004)). While there is little dispute on the importance of this mechanism in

maintaining tissue homeostasis and preventing hyperplasia, the mechanistic details

remain enigmatic. Our data indicate that in Drosophila, IRER serves as the converging

point for induction of apoptosis following overproliferation.

First, we showed that the cis regulatory function of IRER is required for controlling

appropriate cell numbers in the developing tissues. The consequence of lacking IRER

seems to be more significant in tissues where cell survival is largely dependent on the

availability of growth factors. While the overall impact of Df(IRER) on the size of the

embryonic CNS is marginal, the survival of midline glial cells, which have been shown to

be dependent on the EGF signal provided by the neuronal axons (Bergmann et al.,

2002), are significantly impacted. In post embryonic tissues, cell death in the wing disc

has been shown to be strongly influenced by the BMP/dpp signaling (Gibson and

Perrimon, 2005; Martin-Castellanos and Edgar, 2002; Moreno and Basler, 2004;

Moreno et al., 2002a; Shen and Dahmann, 2005). Interestingly, wing discs are very

sensitive to ionizing radiation and heat shock –induced apoptosis (Perez-Garijo et al.,

2004; Ryoo et al., 2004). This sensitivity is at least in part mediated through regulatory

elements reside in IRER, since irradiation –induced cell death is significantly inhibited in

discs lacking this regulatory region (Fig. 3-1). It has been estimated that during normal

development, at any given time point, about 1.4% cells in the larval developing wing

Page 83: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

83

disc are TUNEL-positive (Milan et al., 1997). Since apoptotic cells are quickly removed

by phagocytosis, the overall number of cells that undergo apoptosis during the whole

development period of the wing disc is certainly to be much higher.

The consequence of a reduced level of cell death caused by the absence of IRER

is significantly different from that of blockage of cell death by Baculovirus caspase

inhibitor P35. Inhibiting the final demise of cells with P35 leads to distorted formation in

the wing disc and the adult wing (Perez-Garijo et al., 2004; Ryoo et al., 2004).

Apparently cells with higher levels of RHG genes are not only on the path to final

demise, but also secret growth signals to induce compensatory proliferation of

neighboring cells (Perez-Garijo et al., 2004; Ryoo et al., 2004). In contrast, although

animals lacking IRER have about 10% more cells, the structure of the wing is

essentially normal. While there is a perceivable decrease of apoptotic cells in Df(IRER)

discs (Fig. 3-1D), the overall pattern of apoptotic cells was not significantly different from

that of wild type discs. This suggests that lacking IRER tunes down the sensitivity of

wing disc cells to environmental constraints, such as growth factor availability, without

fully blocking cell death regulation.

The fact that about a 50%-60% reduction of selected RHG genes can have a

significant impact on the number of cells forming the wing is rather interesting. It should

be noted that the overgrowth phenotype we observed was not reported for animals

lacking the transcribed region of reaper (Moon et al., 2008), or grim (Wu et al., 2010), or

sickle (Tan et al., 2011) alone. This is strikingly reminiscent of what was observed for

development cell death of the embryonic neuroblasts, in which the deletion of a central

regulatory region (NBRR) gave a more prominent phenotype than deleting individual

Page 84: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

84

RHG gene (Tan et al., 2011). Large deletions that delete multiple RHG genes, such as

H99, are embryonic lethal, preventing the analysis of tissue growth at later

developmental stages. The relatively similar wing disc overgrown phenotype between

Df(IRER) homozygous and the Df(IRER)/Df(ED225) transheterozygous strongly

suggest that IRER plays a pivotal role in regulating RHG gene expression in response

to increased cell numbers.

In addition to its role in limiting cell number during development, we showed that

IRER is also required for mediating the induction of apoptosis following dMyc-induced

overproliferation. IRER is certainly required for the cell autonomous apoptosis following

dMyc overexpression, as have been demonstrated with the overgrown phenotype of

dMyc;IRER(-/-) clones (Fig. 3-8). However, we cannot exclude that potential inhibition of

super-competition- induced cell death in Df(IRER) might also contributed to the

hyperplasia observed for GMM in Df(IRER) animal. Indeed, additional analysis

indicates that IRER is required for competition -induced elimination of weaker cells.

However, the mechanism seems to be different and involves mainly the role of IRER in

regulating hid expression. In the context of dMyc- induced cell autonomous cell death,

both our observation and that of other’s (Montero et al., 2008) indicate that reaper and

sickle are likely responsible. The levels of these two genes, but not that of hid, were

also significantly decreased in Df(IRER) larvae that have overgrown tissue. Thus, it

appears that overproliferation -induced apoptosis is achieved through IRER –mediated

induction of reaper and/or sickle.

It remains to be seen as to which enhancer is responsible for mediating dMyc –

induced cell autonomous expression of reaper (and sickle), and whether the same

Page 85: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

85

enhancer(s) is(are) responsible for regulating these two genes in developing tissues.

Although there are consensus dMyc/cMyc binding sites within IRER, the lack of a

suitable antibody prevented us from verifying whether any of these sites indeed binds

with dMyc. Whole genome dMyc binding in the cultured Kc167 cells was previously

revealed via the DamID method (Orian et al., 2003), the published list did not contain

any binding site in the RHG genomic block. However, since the IRER is enriched for

suppressive histone modifications and has very low accessibility in the Kc167 cells, it

very likely does not reflect cells in vivo that have relatively open IRER.

Functional Significance of Epigenetic Regulation of IRER in Development and in Tumorigenesis

Epigenetic regulation of IRER during normal development likely plays an important

role in adjusting cellular sensitivity to apoptosis –inducing signals in accordance with

cell type and differentiation/proliferation status. During embryogenesis, IRER is open in

rapidly proliferating cells prior to stage 12, conferring hypersensitivity to environmental

stress (Zhang et al., 2008). It likely also confers sensitivity to growth factor signaling,

based on our observation that reaper expression in the developing epidermis is

significantly impeded in Df(IRER) embryos (Fig. 3-6A). Although it remains to be seen

whether the expression of reaper at the epidermis is directly regulated by the EGF

pathway, it has been shown that apoptosis in the epidermis, and the number of cells

surviving at each segment, is dependent on the EGF signaling (Parker, 2006). In

addition, overexpression of a Ras gain-of-function mutant has been found to suppress

hid expression in embryos specifically around this stage (5-7 hr post egg laying)

(Kurada and White, 1998). In post stage 12 embryos, when most cells enter into post-

mitotic differentiation, IRER is closed and renders cells resistant to stress such as

Page 86: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

86

irradiation. The formation of heterochromatin in IRER never reaches the proximal

enhancer and promoter regions of reaper, which is due to the presence of about a

200bp DNA fragment that functions as a chromatin barrier (Lin et al., 2011). This IRER -

specific epigenetic blocking specifically tunes down the sensitivity of RHG genes to

stress, while still allowing them to be expressed in cell lineage –specific patterns. For

instance, reaper (and sickle) is expressed in neuroblasts in stage 14-16 embryos to

mediate lineage specific apoptosis (Peterson et al., 2002; Tan et al., 2011).

Using the IRER{ubi-DsRed} reporter, we were able to observe that the

accessibility of IRER varies significantly even among neighboring cells in developing

wing or eye discs cells. The variegated pattern of IRER{ubi-DsRed} in developing

tissues suggests that there is a stochastic component of epigenetic regulation of IRER.

A direct consequence of this stochastic regulation is that there is a distribution of cells

with varying degree of IRER accessibility in a given tissue compartment. Our results

with dpp>dMyc indicate that cells with relatively open IRER are preferentially removed

when over-proliferation is sensed among the population of cells. A lasting puzzle of

apoptosis regulation in development has been that how can some cells chosen to

commit suicide while many of their sister cells remain live. Our analysis indicated that

stochastic epigenetic regulation, and the resulting varying degree of sensitivity, may be

an important mechanism to tier apoptotic response to lowering growth factors or

environmental stimuli.

Abnormal chromatin regulation has been hypothesized as a major oncogenic

mechanism (reviewed in (Baylin and Jones, 2011). In this work, we found that the

epigenetically regulated IRER is required for the induction of apoptosis following dMyc-

Page 87: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

87

induced overproliferation. It has been elegantly demonstrated in mammalian models

that the tumorigenic potential of Myc is only fully manifested when apoptosis is blocked

by ectopic expression of key anti-apoptotic regulators such as Bcl-2 (reviewed in (Meyer

et al., 2006)). In this study, we showed that higher levels of dMyc can lead to dramatic

tissue hyperplasia in Df(IRER) animals, where the sensitivity to stress –induced

apoptosis was impeded due to the absence of the regulatory region. The accessibility of

IRER is subject to epigenetic modification, and formation of heterochromatin in IRER is

capable of blocking this region to transcription factors (Zhang et al., 2008). Our

observations strongly suggest that over-expression of dMyc, coupled with epigenetic

blocking of IRER, could lead to tumorigenesis in the absence of any other genetic

mutation. Such a mechanism appears to be also present in mammals. It has long been

observed that Bmi-1, the mammalian ortholog of PcG protein PSC, corroborates with

cMyc in promoting tumorigenesis but suppressing apoptosis (Jacobs et al., 1999). It has

also been hypothesized that epigenetic silencing of pro-apoptotic genes by latent EBV

infection may be the underlying mechanism of endemic Burkitt’s lymphoma, which is

strongly associated with EBV infection and characterized by ectopic expression of Myc

(Allday, 2009). In addition to complete silencing of pro-apoptotic genes or regulatory

regions, it is also possible that disturbance of the stochastic distribution of epigenetic

statuses may lead to increased heterogeneity observed for cancerous cells (Pujadas

and Feinberg, 2012).

The mechanism that underlies the epigenetic regulation of IRER remains to be

fully elucidated. Interestingly, the chromatin barrier (IRER left barrier or ILB) that

prevents the spreading of suppressive histone modification from IRER to the promoter

Page 88: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

88

of reaper also functions as a potent silencing barrier when tested in a vertebrate system

(Lin et al., 2011). However, it’s not clear as to what is responsible for initiating the

formation of facultative heterochromatin in IRER. There is no discernable PRE

(Polycomb response element) in IRER and the formation of heterochromatin in IRER

seems to be independent of anterior/posterior position of the cell. Rather, observing the

expression of IRER{ubi-DsRed} throughout the development of various tissues

suggests that there is no simple cause-effect relationship between cellular differentiation

and/or proliferation status and the accessibility of IRER. It appears that epigenetic

regulation of IRER is dynamic during development, stochastic in tissue compartments,

and responsive to environmental conditions. This regulation may very well present as a

unique system for us to further explore how epigenetic mechanism controls the

fundamental balance between proliferation and apoptosis, and how dysregulation of this

mechanism can contribute to tumorigenesis.

Page 89: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

89

Table 3-1. Drosophila strains used in this study

strain source note

Df(IRER) & Df(IRER_left) (Zhang et al., 2008) IRER deletion

Df(IRER),FRT80B & Df(IRER_left), FRT80B

Generated in this work

Df(3L)ED224 Bloomington #8080 Chromosomal deletion: 75B1--75C6

Df(3L)ED225 Bloomington #8081 Chromosomal deletion: 75C1--75D4

Bac-IRER Generated in this work Targeting site: 51D *

UAS-p35 Bloomington #6298

IRER{ubi-DsRed}5

Generated in this work

dpp-Gal4,UAS-GFP A gift from Laura Johnston

Act-Gal4,UAS-GFP Generated in this work

tub>dMyc>Gal4 A gift from Eduardo Moreno

UAS-Su(var)3-9 RNAi VDRC #39378

Page 90: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

90

Figure 3-1. IRER mediates DNA damage –induced reaper and hid expression in the developing

wing discs. A) The four pro-apoptotic RHG genes (reaper, hid, grim, and sickle) are clustered in a ~ 280 kb genomic region. The four genes are transcribed to the same direction and the two intergenic regions surrounding reaper are unusually long (99 kb and 40kb). Regulatory regions, such as NBRR (Neuroblast Regulatory Region) (Tan et al., 2011) or IRER (Zhang et al., 2008), controls the expression of multiple RHG genes during development or in response to stress. The regions deleted in deficiency strains used in this study are indicated by dashed lines. The region cloned into the BAC-IRER rescue construct is indicated by the red bar on top of the DNA. The figure is not drawn to scale due to space limitation. Nucleotide coordinates are relative to the reaper TSS and based on Drosophila genome release 5.42. B-E) Caspase 3 staining of wing discs without IR (B and D), or at 4 hours after 40Gy of IR (C and E). Irradiation induced a widespread of apoptosis in wild-type wing discs (compare C with B) but not in Df(IRER) discs (compare E with D). Scale bar = 50μm. F) Time course of irradiation-induced reaper expression from whole larvae. Early third instar larvae were irradiated with 40Gy of IR and collected for quantitative RT-PCR (qRT-PCR) at various times after IR. The induction level of reaper, i.e. the ratio of IR vs. control (NT), appeared to be the highest between 1 and 2 hour after IR in both wild-type and Df(IRER) larvae. G) Both reaper and hid transcripts are greatly induced from wild-type wing discs at 1.5~2 hours following IR as shown by qRT-PCR. The induction magnitude is much smaller in Df(IRER) wing discs. For all the qRT-PCR data, means + s.d. are shown (n=3).

Page 91: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

91

Figure 3-2. IRER is required for the control of imaginal disc size. A-B) The

representative images of wing imaginal discs from third instar larvae either heterozygous or homozygous to IRER deletion. C) The size measurements of wing discs of 3rd in star larvae were summarized in the whiskers box plot. The top and bottom bars of the box plot corresponded to the max the min values, respectively. The homozygous Df(IRER) larvae had enlarged wing discs when compared to their heterozygous littermates (p = 0.0024, Student t-test; n(IRER+/-)=20, n(IRER-/-)=32). D-E) The representative images of third instar eye imaginal discs from the labeled genotypes. F) Similar to what was observed with the wing discs, the size of eye discs from homozygous Df(IRER) larvae was also significantly larger than that from heterozygous Df(IRER) larvae (p = 1.9e-5, Student t-test; n(IRER+/-)=23, n(IRER-/-)=27).

Page 92: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

92

Figure 3-3. The cis regulatory function of IRER is required for organ size control. A) A

representative image of the wild-type wing, with five longitudinal veins labeled as L1 through L5. The squares indicate the areas sampled to measure hair number. A’) Wings from IRER-/- adult flies (red) are larger than those from wild-type flies (black). Bar=100µm in A) and A’). B) The density of cells was estimated by counting the number of trichomes from fixed areas on the wing blade. Four 75px2 - areas (squares in A) were selected for trichome counting (L1-L2, L2-L3, L4-L5 and L5). In the posterior compartment, i.e. L4-5 and L5, the cell density is higher in IRER-/- wings (p= 0.0022 and 0.0002, respectively; n=12). There is no significant difference in cell density between the two genotypes in the anterior compartment. (p= 0.0566 and 0.0783, for L1-2 and L2-3, respectively; n=12). C) Quantification of adult wing sizes. As shown in A’, wings of Df(IRER) animals are larger than the wild-type controls (p = 2.6e-5). A similar wing size increase is observed with the transheterozygous Df(ED225)/Df(IRER) (p<0.0001 between ED225-/IRER- (n=20) and +/+ (n=25)), p=0.0581 between ED225-/IRER- and IRER-/- (n=26)). In contrast,

Page 93: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

93

the wing overgrown phenotype was partially rescued by Df(ED224) (p<0.0001 between ED224-/IRER- (n=27) and +/+, p=0.0236 between ED224-/IRER- and IRER-/-). The deletion regions in ED224 and ED225 are indicated in Fig 1A. D) Introducing the BAC-IRER to the second chromosome failed to rescue the increased wing size of IRER-/- (p< 0.0001 between Bac-IRER, IRER-/- (n=30) and +/+, p= 0.2669 between Bac-IRER, IRER-/- and IRER-/-. E) The basal levels expression of reaper and sickle are significantly reduced in IRER-/- larvae as measured by qRT-PCR. F) The expression of reaper is also decreased in the IRER-/- wing discs. The unpaired student t-test was used to calculate p-value.

Page 94: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

94

Figure 3-4. Differentially increased compartment size in Df(IRER) wings. A) Schematic

figure showed the A-P compartment of the Drosophila wing. Compartment size was measured as the number of pixels within the contour lines. B) The size of A/P compartment was measured separately for both wild type and Df(IRER) wings. When comparing to the corresponding compartment in wild type wings, A-compartment of Df(IRER) wings showed a ~10% size increase, and P-compartment of Df(IRER) wings showed a ~19% size increase. p= 0.0151 between A (+/+) and A (IRER-/-), p= 0.0012 between P (+/+) and P (IRER-/-), student t-test; n=7 for each genotype.

Page 95: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

95

Figure 3-5. Homozygous Df(IRER) embryos have enlarged central nervous systems. A-

B) About 10% of the embryos homozygous to IRER deletion contained an enlarged central nervous system (CNS) at the end of embryogenesis (st. 17). Anti-Elav staining was used to label differentiated neurons. C-D) Anti-β-galactosidase staining was performed to st.17 embryos bearing the P[slit1.0-lacZ reporter to label the midline glia cells. The heterozygous Df(IRER) embryos have approximately three labeled cells per segment, whereas the homozygous Df(IRER) embryos have 2~5 extra cells/segment.

Page 96: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

96

Figure 3-6. Decreased expression levels of reaper in homozygous Df(IRER) embryos.

A) We analyzed the transcriptional regulation of the pro-apoptotic gene reaper by performing whole-mount in situ hybridization (ISH) in both wild-type and IRER-deficient embryos. Compared to the wild-type embryos, IRER-deficient embryos at stage 10 -11 have much less ISH signal. There was no difference at later stage when reaper is mainly expressed in the neuroblasts. B) To quantify the reaper transcription level from staged embryos, st.9-12 embryos were collected and qRT-PCR was performed. In consistence with the ISH results, decreased levels of reaper mRNA was observed for both Df(IRER) and Df(IRER_left) embryos. The experiments in this figure were performed by Dr. Nianwei Lin.

Page 97: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

97

Figure 3-7. IRER is required for dMyc –induced cell death. A) Consistent with previously

reported results (Montero et al., 2008), expression of dMyc under the control of the GMR-Gal4 driver (GMM) induced both overproliferation and apoptosis and had little impact on the size of the adult eye. B) There is no significant overgrow of eyes in IRER-/- adults. C) Expression of dMyc in the homozygous Df(IRER) animals results in significant overgrown phenotype, with some eyes containing protruding cell mass (black arrow). D) The enhancer effect of Df(IRER) on GMM –induced hyperplasia is detectable in animals heterozygous to Df(IRER). E-F) A similar enhancing effect was observed from flies heterozygous to Df(ED225), or flies over-expressing the viral caspase inhibitor P35. G-I) The hyperplasia of GMM,IRER-/- was evident

Page 98: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

98

when observed with scanning electron micrographs (SEM). Note that the ommatidia from GMM,IRER-/- were disorganized. J-L) Fluorescent in situ hybridization (FISH) was performed to the third-instar eye discs to detect reaper transcripts. Corresponding with the eye phenotype, we found that reaper was significantly induced by GMM in the GMR-domain in wild-type eye discs, and this induction was absent in IRER-/- eye discs. M) Either lack of IRER (IRER-/-) or overexpression of dMyc (provided by tub>dmyc) alone is sufficient to increase wing size (p < 0.0001 between +/+ (n=30) and IRER-/- (n=26), p< 0.0001 between +/+ and tub>dmyc (n=20), p=0.8818 between IRER-/- and tub>dmyc). When combined together (tub>dmyc,IRER-/-)(n=19), the wing size is increased even further, suggesting the synergy between Df(IRER) and dMyc-overexpression (p=0.0012 between IRER-/- and tub>dmyc,IRER-/-, p=0.0027 between tub>dmyc and tub>dmyc,IRER-/-). N) The representative wing images of tub>dmyc (upper panel) and tub>dmyc,IRER-/- (lower panel, merged layer of tub>dmyc,IRER-/- and tub>dmyc).

Page 99: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

99

Figure 3-8. Cell-autonomous role of IRER regulates dMyc-induced overgrowth. A)

Schematic representation of the MARCM strategy used to induce dMyc expression from IRER-mutant clones. Compared to wild-type clones with dMyc-expression (B-B’ & C-C’), IRER-mutant clones (D-D’ & E-E’) with dMyc-expression are significantly larger in size. Clones were monitored at 72hours after heat-shock. The percentage occupancy of clones was examined by calculating the ratio between the GFP-positive area and the entire disc area. The dMyc,IRER-/- clones occupy a much larger area than dMyc-expressing clones in both eye disc F) and wing disc G).

Page 100: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

100

Figure 3-9. Cells with an open IRER are more sensitive to dMyc -induced cell death. A) An

ubiquitin-DsRed reporter was inserted, via homologous recombination, into the middle of IRER, a locus that showed strongest resistance to DNase I in late stage embryos (Zhang et al., 2008). The strain was referred to as “IRER{ubi-DsRed}” and served as a reporter for the epigenetic status of IRER. The IRER{ubi-DsRed} reporter signal is variegated in the developing wing discs. B-B’’). Expression of GFP (green) in the dpp domain does not change the variegated expression pattern of IRER{ubi-DsRed}. C-C’’) When dMyc was co-expressed with GFP, there was a conspicuous lack of DsRed- positive cells in the dpp zone (yellow arrows). D-D’’) When cell death in the dpp zone was blocked by co-expressing UAS-diap1, many DsRed -positive cells were rescued (yellow arrows). E-E’’) dMyc-overexpression clones, marked by GFP (dashed lines), were generated by the Act>y>Gal4 flip-out in conjugation with UAS-GFP and UAS-dMyc transgene expression. Note cells in the dMyc-overexpressing clones have lower levels of IRER{ubi-DsRed} signal comparing to neighboring cells. Figure E-E’’) was provided by Dr. Sergio Casas Tintó (Cajal Institute, Spanish Research Council (CSIC), Madrid, Spain) and Dr. Eduardo Moreno (Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain).

Page 101: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

101

Figure 3-10. Su(var)3-9 is required for the epigenetic silencing of IRER in wing discs. A)

The expression of the reporter IRER{ubi-DsRed} was variegated in the developing wing discs (red). B) The reporter activity can be significantly de-repressed when Su(var)3-9 was knocked down from Ser-expressing cells. Cross: Ser-Gal4; IRER{ubi-DsRed} X Su(var)3-9 RNAi. The expression pattern of Ser-Gal4 was visualized using a nuclear-localized GFP (green-GFP; blue-DAPI). Cross: Ser-Gal4 X UAS-nlsGFP. The de-repression is not exclusively cell-autonomous, potentially due to secondary stress to neighboring cells.

Page 102: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

102

Figure 3-11. Epigenetic silencing of IRER is responsive to stress. A-A’’) Low and

variegated expression of IRER{ubi-DsRed} in the wing disc. B-B’’) & C-C’’) Irradiation led to an increase of the reporter signal. Wing discs were dissected from third instar larvae at either 4hrs or 24hrs after a 40Gy x-ray treatment.

Page 103: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

103

Figure 3-12. Cells lacking IRER have the propensity to overgrown and are resistant to

stress –induced cell death. A-D) Clones were induced using the FLP/FRT-mediated mitotic recombination and labeled by the absence of GFP (white lines). The sibling wild-type twin spots are marked by 2XGFP (brighter green). While neutral clones in either wing- or eye- imaginal discs generally grow to similar size as the simultaneously induced twin spots, homozygous IRER-deficient clones tends to overgrow (A & B, wing disc; C & D, eye disc, 72hr following clone induction). E-F) While there is a considerable variation in how much the Df(IRER) clone overgrows, the overgrown phenotype is significant (open bars in E & F. wing disc: p=0.017 between Ctr and Df(IRER-left), p<0.001 between Ctr and Df(IRER); eye disc: p<0.001 between Ctr and Df(IRER-left), p=0.002 between Ctr and Df(IRER)). Wilcoxon rank-sum test was used to for p-value calculation. When animals are subject to a sub-lethal dose (10Gy) of x-ray at 24 hours following clone induction, the size difference between Df(IRER) clones and the twin spots is significantly increased (closed bars in E & F. wing disc: p=0.021 between Ctr and Df(IRER-left), p=0.002

Page 104: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

104

between Ctr and Df(IRER); eye disc: p=0.027 between Ctr and Df(IRER-left), p=0.002 between Ctr and Df(IRER)). Size was measured at 72~96hr after clone induction and the ratio of clone size vs. twin spot size was calculated for different groups. G) Scatter plot of clone size and twin spot size measured at 72~96hr after clone induction. Red dots and green dots represent the clones induced with or without IR, respectively. H) Animals were irradiated (40Gy) at 48 hr post clone induction. Apoptosis was measured by Caspase 3 staining at 4hr following IR. There is a lack (or reduced level) of apoptotic cells in the Df(IRER) clones. The size difference between the mutant clone and the twin spot in this setting is not significant due to the relatively short time following clone induction and x-ray treatment.

Page 105: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

105

Figure 3-13. Df(IRER) clones overgrow their twin spots in the eye discs. A) The clone

size from eye discs was summarized in the 2D plot. B-D) Mitotic clones were induced in adult eyes (clone-red; twin spot-white). Both Df(IRER-left) and Df(IRER) clones grew larger than their twin spots.

Page 106: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

106

Figure 3-14. High order chromatin organization brings IRER into the proximity of

multiple RHG gene promoters. A) Hi-C data for D.melanogaster embryos was taken from a recent genome-wide study (Sexton et al., 2012). The region demarcated by the black lines indicates the predicted physical interaction domain, which is corresponding to the synteny block and enriched for HCNE (highly conserved noncoding elements). The HCNE data is derived from Ancora (Lin et al., 2009). The region containing IRER is enlarged in panel B). There are multiple predicted binding sites for both cMyc/dMyc and P53 within IRER. The bioinformatics was done by Guangyao Li.

Page 107: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

107

Figure 3-15. Diagram summarizing the mechanism of IRER. It has been known that

ectopic expression of dMyc leads to overproliferation and oncogenic potential, which is quickly followed by reaper/sickle-dependent apoptosis to remove the oncogenic cells. We here describe the regulatory elements within IRER function as the central regulatory element for the induction of apoptosis following overproliferation. It is still not clear whether dP53 is involved in the process and how dMyc induces apoptosis through IRER (e.g. direct binding or indirect targeting into IRER) and requires further studies. In addition, epigenetic regulation of IRER defines cellular sensitivity to developmental constraints and oncogenic stress. In general, cells with an open IRER are more sensitive to stress-induced apoptosis and have less potential to over-proliferate; whereas cells with a closed IRER are more resistant to stress and consequently, easier to undergo over-proliferation when they are confronted with over-expression of oncogene such as Myc.

Page 108: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

108

CHAPTER 4 A STRESS- RESPONSIVE REGULATORY REGION IS REQUIRED FOR

COMPETITION- INDUCED APOPTOSIS

Introduction

In multicellular organisms, cellular communication plays an essential role in

determining cell fates and growth decisions, e.g. whether a cell can survive to contribute

to the adult tissue. Cell competition is one of the sophisticated cellular phenomenon that

was originally identified in Drosophila in the 1970s, when fly geneticists were studying a

class of mutations of genes that encode ribosomal proteins (Morata and Ripoll, 1975).

These dominant mutations, named Minutes, are homozygous lethal to cells but are able

to survive in heterozygotes (M+/-), although it takes much longer for the organism to

grow and reach normal size. Surprisingly, when clones of M+/- cells are induced from

the wing discs containing otherwise wild type cells, they are actively eliminated and are

not recovered in the adult wing tissue. Therefore, the intrinsically viable cells could be

eliminated when surrounded by cells with higher fitness, a phenomenon called cell

competition (Simpson, 1979; Simpson and Morata, 1981).

Subsequent studies have shown that cell competition is not restricted to the

Minute condition and can be triggered by a variety of other genes. One of the decisive

regulators in cell competition is the transcription factor dMyc, which is the single

Drosophila orthologue of the mammalian proto-oncogene c-Myc (Johnston et al., 1999).

Through a still unclear mechanism, cells can compare dMyc levels with their neighbors

in the same tissue compartment and establish a win/lose status: cells with relatively

lower levels of dMyc will be removed while cells with relatively higher levels of dMyc will

expand at the expense of the loser cells, through compensatory proliferation (de la

Cova et al., 2004; Moreno and Basler, 2004). Since both dMyc and Minute mutations

Page 109: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

109

affect ribosomal biogenesis and protein synthesis, it is likely that the local differences of

ribosome activity are the factors leading to cell competition in this scenario (de Beco et

al., 2012).

Interestingly, recent studies have highlighted the role of a group of polarity genes

in the process of cell competition. This group of polarity genes, including scribble (scrib),

lethal giant larvae (lgl), and disc large (dlg), are evolutionarily conserved neoplastic

tumor suppressors. They play an important role in apical-basal polarity control and are

vital for epithelial integrity (Bilder, 2004; Hariharan and Bilder, 2006). Flies homozygous

for scrib mutation develop normally till the end of larvae stage, when the maternally

deposited scrib protein has been exhausted at that point. Their imaginal discs keep

proliferating without terminal differentiation, and the animals eventually die as a giant

larvae with multilayered epithelial structures (Wodarz, 2000). Interestingly, when clones

of scrib-mutant cells are surrounded by wild type tissues, they are actively eliminated

through cell competition instead of overgrowing their neighbors, like one might expect

(Brumby and Richardson, 2003; Pagliarini and Xu, 2003). Their elimination requires the

existence of the neighboring wild type cells, which is a hallmark of cell competition.

Since cell competition leads to and relies on the removal of suboptimal cells, it is

essential to fully elucidate the molecular mechanisms used to conduct their death. Here

we used both dMyc- and scrib-induced cell competition models to show that an

epigenetically regulated, stress responsive region-- IRER, is required for the induction of

pro-apoptotic genes in response to competition stress. Loss of IRER leads to inhibition

of pro-apoptotic gene expression and increased survival of disadvantaged cells.

Furthermore, the epigenetic status of IRER can be regulated by competition stress,

Page 110: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

110

which can possibly change the susceptibility of cells to apoptosis. It seems to be

worthwhile to better recognize the consequences of epigenetic regulation in cell

competition.

Materials and Methods

Drosophila Strains and Culture

All crosses were done at 25°C and maintained on a standard corn-agar medium

unless otherwise mentioned. Canton S Drosophila strain was used as wild-type control

in this study. Defined deletions of IRER were generated previously as described (Zhang

et al., 2008).

Clone Induction

Wild-type clones in a tub>dMyc background were induced from larvae of genotype

hsp70-flp; tub>dmyc>Gal4; UAS-GFP as previously described (Moreno and Basler,

2004). Wild-type clones in the tub>dMyc,Df(IRER) background were induced from

larvae of genotype hsp70-flp; tub>dMyc>Gal4/UAS-GFP; Df(IRER) upon the same

condition.

Microscopy and Immunohistochemistry

Imaginal disc dissection and immunostaining were performed as described in

Chapter 3.

Gene Expression Analysis

RNA was extracted from 8-10 larvae of the desired genotype using the RNeasy

Minikit (Qiagen). cDNA synthesis and quantitative PCR were performed as described in

Chapter 3.

Page 111: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

111

Results

IRER is Required for Super Competition-induced Elimination of Loser Cells

In the fly wing imaginal discs, the dMyc-induced cell competition is extraordinarily

influenced by the neighboring cells. For example, the wild type cells are recognized as

winner cells when surrounded by the dMyc-mutant cells, but become losers when facing

the challenge from dMyc-overexpressing cells, e.g., driven by the tubulin promoter.

Here we used the previously reported tubulin>dMyc>Gal4 system to study the role of

IRER in mediating super competition-induced apoptosis (Moreno and Basler, 2004). In

this system, all cells have elevated levels of dMyc due to the ubiquitous and strong

tubulin promoter. However, FLP-mediated excision of FRT -flanked dMyc will generate

clones of cells that have wild-type levels of dMyc protein. The cells with lower levels of

dMyc compared to surrounding cells, marked by GFP, will eventually be eliminated

through cell competition –induced apoptosis. When this system was recombined with

Df(IRER) or Df(IRER_Left), we noticed that the elimination of wild type clones was

significantly inhibited. While most GFP -positive clones were eliminated at 72 hours post

clone induction in wild-type discs (Fig. 4-1A), many clones survived, with larger size, in

the wing discs that lacked an IRER (or IRER_left) (Fig. 4-1B).

IRER is Required for Super Competition-induced hid Expression in Loser Cells

When the level of RHG genes was monitored, we noticed a dramatic induction of

hid between 24-48hrs following the generation of wild type clones by heat shock (Fig. 4-

2A). Parallel heat shock experiments conducted with animals that have hs-FLP but lack

tubulin>dMyc>Gal4 indicated that there was no detectable difference of hid mRNA

levels in heat-shocked or mocked animals between 24-48 hours following the treatment

Page 112: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

112

(data not shown). This strongly suggests that the induction of hid observed in hs-FLP;

tubulin>dMyc>Gal4 animals was due to competition -induced cell death in this system.

To our surprise, this induction of hid following wild type clone induction in

tubulin>dMyc>Gal4 animals was totally blocked in animals lacking IRER (Fig. 4-2A).

Even more intriguingly, we found that although reaper (and sickle) was not usually

induced in this competition –induced apoptosis system, the level of reaper mRNA was

nonetheless significantly increased in Df(IRER) animals (Fig. 4-2B). The induction of

reaper and sickle may be due to the accumulation of these cell death genes in the

“undead” cells in the Df(IRER) background. This may explain why the wild type clones

are eventually eliminated in the Df(IRER) animal.

Taken together, our observations indicate that IRER is required for competition -

induced cell death in a dMyc overexpression system, by mediating the pro-apoptotic hid

expression.

IRER is Required for the Elimination of scrib Knockdown Cells

Cell competition -induced apoptosis has been observed for cells mutated for cell

polarity genes. Cells mutated for scrib fail to exit cell cycle; and under many

circumstances the mutant cells are eliminated by JNK-mediated apoptosis (Bilder et al.,

2000; Brumby and Richardson, 2003; Pagliarini and Xu, 2003). Since both scrib (97B-

97C) and IRER (75C) are located on the 3rd chromosome, it is very difficult to generate

clones deficient for both scrib and IRER. To overcome this obstacle, we decided to

perform tissue-specific RNA interference (RNAi) to knock down scrib expression in eye

progenitor cells using the shRNA strategy (Ni et al., 2011).

In the subsequent experiments, ey1x-Gal4 (Bloomington 8228) was used to drive

the expression of both the GFP reporter gene and the shRNAs targeting scrib. The

Page 113: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

113

expression profile of the GFP signal indicated that the ey1x-Gal4 was active in eye disc

cells starting from late embryogenesis. In third in star larvae, the GFP signal was mostly

restricted to cells behind the morphorgenic furrow (Fig. 4-3A-A’). When scrib was

depleted by RNAi, a subset of scrib knockdown cells in the eye discs failed to complete

differentiation as indicated by Elav staining (Fig. 4-4 B-B’’’). The adult eyes of ey-

scribRNAi were smaller in size and had rough surface. Interestingly, for some eyes there

were necrotic spots in the middle of the eye (Fig. 4-3C1-C2). Similar phenotypes were

observed for the two shRNA strains, JF03229 and HMS01490, with the later resulting a

relatively more severe phenotype than the former (Fig. 4-5). When ey-scribRNAi was

introduced to the IRER heterozygous background, more GFP positive cells were

detectable in the eye discs in third instar larvae and some were even observed

migrating towards the antenna disc (Fig. 4-4 C-C’’’). However, no GFP positive cell

could be observed in the antenna disc, and adult eyes displayed a very similar small &

rough phenotype as the ey-scribRNAi flies in wild type background (data not shown).

Strikingly, when ey-scribRNAi was introduced to the homozygous Df(IRER) genetic

background, GFP positive cells were frequently found in the antenna disc in third instar

larvae (Fig. 4-3D’). Correspondingly, the overall morphology of the discs and the

monolayered epithelial organization was disrupted (Fig. 4-3 D). Some adults of ey-

scribRNAi in Df(IRER) background (~30%for HMS01490) had ectopic pigment cells in the

head (Fig. 4-3 D1). About 70% of adults had extra antennae –like structures mixed with

an undifferentiated tissue mass (Fig. 4-3 D2).

The majority of cells in the undifferentiated cell mass in the head were GFP

positive (Fig. 4-3 D1’ & D2’), indicating that the eyeless promoter remained active in

Page 114: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

114

these cells, whereas it was turned off in most differentiated cells. When the expression

of reaper and hid was monitored by fluorescent in situ hybridization (FISH) in the ey-

scribRNAi discs, the mRNA levels of both were elevated (Fig 4-6 A-A’’, C-C’’). However,

this elevation of reaper and hid in the ey-scribRNAi discs was absent in Df(IRER) animals

(Fig. 4-6 B-B’’ & D-D’’).

Disruption of Cell Polarity led to de-Repression of IRER

Previous studies have found that the DNA accessibility of IRER is subject to PcG-

mediated epigenetic regulation. The epigenetic status of IRER dictates cellular

sensitivity to developmental constraints and multiple stresses. In general, cells with an

open IRER are more sensitive to stress-induced apoptosis and tend to be preferentially

eliminated from the tissue (Zhang et al., 2008). Using a ubi-DsRed construct inserted

into IRER as a reporter, we were able to monitor the epigenetic status of IRER with a

cellular resolution. Interestingly, we found that when Scrib was knocked down in Ser-

expressing cells of the wing disc, there was a significant induction of the IRER{ubi-

DsRed} signal specifically in the Ser-domain. Therefore, disruption of cell polarity, via

down-regulation of the polarity-control gene scrib, leads to derepression of IRER and

thus increased sensitivity to stress-induced cell death. The increased DNA accessibility

of IRER in scrib knockdown cells may contribute to their elimination from the tissue, a

hypothesis which requires further evidence. Nonetheless, the epigenetic response of

IRER to decreased scrib activity suggests a link between epigenetic regulation and

polarity control.

Page 115: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

115

Discussions

Myc-induced Cell Autonomous Apoptosis vs. Cell Non-autonomous Apoptosis

It has been long known that ectopic expression of dMyc in the GMM context is

able to induce apoptosis in a cell autonomous manner (Montero et al., 2008). In Chapter

3, we identified IRER as a central regulatory region for dMyc-induced apoptosis in the

GMM. The fact that IRER is also required for competition –induced cell death in the

tub>dMyc>Gal4 system was quite a surprise to us. It should be noted that IRER is not

required for competition –induced cell death in the Minute +/- system (Li and Baker,

2007)(personal communication with Dr. Nicolas Baker). The main commonality between

dMyc –induced autonomous cell death and competitive cell death is that both are

affected by the availability of growth factors. However, detailed analysis indicated that

the two processes, although both requiring cis regulatory functions that reside in the

same intergenic region of IRER, likely involve different mechanisms.

First, it has been shown that dMyc -induced cell autonomous apoptosis is

accompanied by an increased expression of reaper and sickle, but not hid (Montero et

al., 2008). This was also confirmed by us in Chapter 3. And it appears that the effect of

IRER deletion on dMyc –induced cell autonomous cell death is through reducing the

responsiveness of reaper (and sickle). In contrast, we found that hid is the only RHG

gene strongly induced in the tub>dMyc>Gal4 system between 24-48 hours following

clone induction, a period when a majority of the elimination of wild type clones (with

lower levels of dMyc) occurs (Moreno and Basler, 2004). This correlates with previous

findings that the function of hid is required for dMyc super-competitor –induced cell

death (de la Cova et al., 2004).

Page 116: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

116

Secondly, we found that although the induction of hid in the tub>dMyc>Gal4

system was completely blocked in animals lacking IRER, the level of reaper mRNA,

which usually is not induced in this system, was then found to be significantly increased

(Fig. 4-2B). This phenomenon is rather intriguing and is to some extent reminiscent of

what was observed in an embryonic midline system. In the CNS midline glial cells that

are destined to die, usually the expression of reaper is not detectable while the

expression of hid is prominent. However, in the hid mutant background, there is a

dramatic accumulation of reaper (Zhou et al., 1997). This could be due to the fact that

delayed or inhibited cell death in the absence of hid induction allows the accumulation

of reaper mRNA, which would usually be too low to be detected. This is unlikely to be

the case in the tub>dMyc>Gal4 larvae since reaper levels in the larvae were reliably

detectable. It may also reflect a redundant mechanism built into the coordinated

regulation of RHG genes, which allows induction of alternative genes when the first

preference is not available or fails to respond. The nature of such a mechanism, as well

as the identification of distinctive elements required for cell-autonomous vs. competition

–induced RHG genes expression, will have to await further mechanistic analysis of the

IRER.

Epigenetic Regulation and Cell Competition

Since cell competition leads to and relies on the elimination of loser cells via

apoptosis, a fundamental question that remains to be answered is how the differences

of cellular fitness are recognized and what mechanisms are used to kill the loser cells.

Recent studies using gene expression arrays have brought some exciting insights into

the process of specification of winner/loser cells. It appears that this distinction involves

the differential display of the integral membrane protein Flower (Fwe), which is highly

Page 117: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

117

conserved in multi-cellular organisms (Rhiner et al., 2010). Fwe is alternatively spliced

in disadvantaged cells, and has been proposed to be the code that separates them from

better adapted cells. However, how the Fwe code transmits the death signal, as well as

which genes are involved in the process, is not yet clear.

In Chapter 4, we report that the stress responsive region IRER is required for cell

competition-induced elimination of loser cells, presumably by mediating the

transcriptional induction of hid upon competitive stress. Intriguingly, IRER is

epigenetically regulated and the DNA accessibility within this region defines cellular

sensitivity to stress-induced apoptosis (Zhang et al., 2008). The finding that IRER is

derepressed in cells with lower levels of scrib is rather interesting and links epigenetic

regulation and cell competition. One potential hypothesis is that decreased levels of

scrib leads to higher DNA accessibility of IRER, which allows transcription of pro-

apoptotic gene hid and activation of cell death from scribRNAi cells. Further experiments

would need to be done to study whether indeed the scribRNAi cells with open IRER

undergo apoptosis, e.g. by staining the wing discs of Ser>Scrib RNAi, IRER{ubi-DsRed}

with cleaved Caspase 3 antibody. If this proves to be true, it would be interesting to

further investigate the relationship between Fwe and IRER. For example, does

derepression of IRER in loser cells happen before, at the same time, or after the

splicing of the Fwe(lose) isoform. Furthermore, it is worth to test whether a similar

epigenetic response of IRER also exists in cell competition induced by other genes (e.g.

dMyc or Minute).

Interestingly, recent work from Bondar and Medshitov presented that cell

competition also exists between mammalian hematopoietic stem cells and is mediated

Page 118: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

118

by the relative activity of P53: cells with higher levels of P53 activity were outcompeted

by cells with lower levels of P53 activity. Since P53 activation is a critical cellular

response to DNA damage, the P53-dependent cell competition likely reflects a fail-safe

mechanism to select the progenitor cells with less damage (Bondar and Medzhitov,

2010). Intriguingly, there are several putative P53 binding sites within IRER, including

the previously identified dP53 response element (P53RE). The dP53-induced apoptotic

response following DNA damage is totally abolished when IRER is epigenetically

blocked in late stage embryos, suggesting the essential role of IRER in dP53-induced

cell death. The involvement of both P53 and IRER in cell competition highlights the

connection of cell competition, stress response and homeostatic maintenance.

Current knowledge indicates that dMyc-induced cell competition removes the loser

cells via hid-dependent apoptosis and likely the activation of JNK (de la Cova et al.,

2004; Moreno and Basler, 2004). A substantial amount of experimental data strongly

suggests that the Eiger/JNK-induced apoptosis plays a key role in eliminating the

polarity-deficient clones of scrib-mutant cells. The stress-responsive JNK signal is

upregulated from loser cells and triggers the ultimate execution of their apoptosis

(Brumby and Richardson, 2003; de la Cova et al., 2004; Igaki et al., 2006; Moreno and

Basler, 2004; Pagliarini and Xu, 2003). Eiger, the sole Drosophila tumor necrosis factor

(TNF), has been identified as an upstream regulator of JNK activity (Igaki et al., 2002;

Moreno et al., 2002b). Eiger is taken up by loser cells through endocytosis, which in turn

activates downstream JNK signaling and induces apoptosis from the scrib-mutant

clones. Indeed, either abolishing Eiger expression or down-regulating the endocytosis

Page 119: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

119

activity is sufficient to lead to tumor progression from epithelium containing polarity-

deficient clones (Igaki et al., 2009).

It is still unclear how the activated JNK pathway leads to the induction of pro-apoptotic genes as well as the execution of cell death in out-competed cells. Further experiments

need to be done to investigate the role of IRER in mediating JNK-induced apoptosis. The results from this study will hopefully set up a link between JNK activation and

apoptotic consequence in the loser cells.

Page 120: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

120

Figure 4-1. IRER is required for super competition -induced elimination of loser cells. In

transgenic flies carrying tubulin>dMyc>Gal, tub>Gal4 clones are generated by inducing expression of hs-FLP. These tub>Gal4 clones (marked by GFP) have wild-type levels of dMyc and are eliminated through cell competition –induced apoptosis because they are surrounded by cells expressing higher levels of dMyc (Moreno and Basler, 2004). A) By 72 hours after clone induction, majority of tub>Gal4 clones have been eliminated. B) However, the elimination of Tub>Gal4 clones is strongly inhibited in Df(IRER) discs. The data was provided by Dr. Sergio Casas Tintó (Cajal Institute, Spanish Research

Council (CSIC), Madrid, Spain) and Dr. Eduardo Moreno (Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain).

Page 121: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

121

Figure 4-2. IRER is required for super competition -induced hid expression in loser cells.

A) The pro-apoptotic gene hid is significantly induced by cell competition from the larvae at 24~48 hours after heat shock. However, the hid induction is almost totally blocked in IRER-deficient larvae. B) Another pro-apoptotic gene reaper is not responsive to cell competition in the presence of IRER, but is highly induced by cell competition in homozygous Df(IRER).

Page 122: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

122

Figure 4-3. IRER is required for the elimination of scrib knockdown cells. A-A’) & A1-A2)

Eye-antenna discs containing GFP expression driven by ey1x-Gal4. The animals develop normal eye, and antenna. B-B’) & B1-B2) Expression of ey1x>GFP in the homozygous Df(IRER) animals. These animals also develop normal eye and antenna, suggesting that the loss of the regulatory function of IRER by itself does not affect the eye-antenna development. C) When scrib is knocked down from ey1-expressing cells (marked by GFP) in eye discs, the affected disc cells lose the mono-layered organization and become folded as revealed by DAPI staining. C’) Some scribRNAi cells are misplaced and undergo local migration. C1) The eyes of ey1x>scribRNAi adults are small in size and have rough surface, with some eyes showing necrotic spots. C2) Antenna development is not affected. D-D’) When inducing scrib RNAi in IRER-deficient background, the ey1x>scribRNAi cells (marked by GFP) are detectable from antenna discs. D1-D2) Development of eye and antenna is severely affected: extra eyes (or eye-like tissues) and antennae (or antenna-like tissues) are frequently observed. D1’-D2’) The GFP-labeled scribRNAi cells can survive to adult stage and aremainly detected in the undifferentiated tissues in the head. D1’ and D2’ are the GFP channel of D1 and D2, respectively.

Page 123: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

123

Figure 4-4. Scrib knockdown leads to differentiation defects of photoreceptor cells. A-

A’’’) Wild-type eye discs with ey1x>GFP expression were stained with anti-Elav antibody to label the differentiated photoreceptor cells. All the cells behind the morphogenetic furrow (MF) were differentiated and positive for both GFP expression and Elav staining. B-B’’’) Upon scrib knockdown, some cells posterior to the MF were absent for Elav staining, suggesting their differentiation was affected (B’). C-C’’’) Similarly, a subset of scribRNAi cells failed to undergo terminal differentiation in heterozygous Df(IRER) background. Furthermore, some scribRNAi, IRER+/- cells tend to migrate within eye disc.

Page 124: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

124

Figure 4-5. Scrib knockdown phenotype in eyes. A) Flies carrying UAS-scribRNAi

constructs have normal eye development. B-C) In order to knock down the scrib expression from developing eyes, two different UAS-scribRNAi lines ordered from TRiP were used: the Long Hairpin JF03229 and the Short Hairpin HMS01490. They both cause a small, rough eye phenotype with frequently obversed necrotic spots in the eye. For the rest of the experiments, we chose the short hairpin HMS01490 for scrib knockdown because it appeared to have higher efficacy of gene targeting, and induced a stronger phenotype.

Page 125: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

125

Figure 4-6. IRER is required for scrib RNAi-induced reaper and hid expression from eye

discs. The transcription levels of reaper and hid mRNA were monitored by FISH. Indeed, ey1x>scribRNAi led to a strong induction of both reaper and hid in the wild type eye discs A-A’’) & C-C’’), but not from the homozygous Df(IRER) discs B-B’’) & D-D’’).

Page 126: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

126

Figure 4-7. Disruption of cell polarity leads to de-repression of IRER. A-A’’) Low and

variegated expression of IRER{ubi-DsRed} in the wing disc. B-B’’) Knocking down scrib (cross: Ser-Gal4; IRER{ubi-DsRed} X UAS-scrib RNAi) led to de-repression of IRER{ubi-DsRed} from the Ser-expression domain (Check Fig. 2-4C for the expression pattern of Ser-Gal4 in wing discs).

Page 127: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

127

CHAPTER 5 DISCUSSION AND PERSPECTIVE

Coordinated Transcriptional Regulation of the RHG Genes by Locus Control Regions

The RHG genes as a whole are required for almost all development cell death

during embryogenesis in Drosophila Melanogaster. Embryos homozygous for the H99

deletion, which removes hid, grim, and reaper, fail to hatch and have a CNS that

contains about 3 times more cells than that in wild type embryos (White et al., 1994).

However, how individual cells are chosen to die during embryogenesis, or in later

developmental period, is far from clear. With the exception of hid, the other three genes

(reaper, grim, and sickle) seem to be exclusively expressed in cells that are to be

eliminated shortly after one or a multiple of these genes is expressed (Jiang et al., 1997;

Lohmann et al., 2002; Steller, 2008; Zhou et al., 1995). Transcriptional regulation of

these genes thus plays a pivotal role in determining the life/death decision for an

individual cell.

Centralized regulation, i.e. one regulatory region which coordinates the expression

of multiple genes, appears to be the general theme of transcriptional regulation of the

RHG genes. Previously, we have found that the intergenic region between reaper and

sickle (i.e. IRER) is responsible for mediating radiation -induced expression of not one,

but three pro-apoptotic genes (reaper, hid, and sickle) (Zhang et al., 2008). Data from

Kristin White’s group has shown that a ~22kb intergenic region between grim and

reaper (i.e. neuroblast regulatory region or NBRR) is required for appropriate

developmental expression of reaper, grim, and sickle in neuroblasts at the end of

embryogenesis (Tan et al., 2011). While IRER is required for stress (DNA damage) -

induced expression of RHG genes, it is not required for the expression of RHG genes in

Page 128: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

128

neuroblasts during late embryogenesis. Rather, it appears that cell lineage- specific

expression of these genes in neuroblasts is controlled by the NBRR. Although it

remains to be seen as to whether this observed separation of task will hold true for cell

death regulation in other cell lineages or in response to different kinds of stress, it is

now clear that transcriptional regulations of the RHG pro-apoptotic genes are

coordinated by locus control regions such as the NBRR and IRER.

Both IRER and NBRR, situated upstream and downstream of reaper, respectively,

are highly conserved intergenic regions enriched for HCNEs (Fig. 3-14A). Characterized

HCNEs in both vertebrates and insects genomes often regulate target genes over long

distance to guide tissue or developmental stage –specific expression patterns (Kikuta et

al., 2007). The extensive chromosomal interactions revealed by the whole genome Hi-C

analysis within this region (Fig. 3-14A) strongly suggest that regions such as NBRR and

IRER can coordinate the expression of RHG genes through long range chromosomal

interactions. However, the detailed interaction map of the RHG genomic regulatory

block, as well as the potential dynamics of the chromosomal interactions, remains to be

fully understood.

Epigenetic Regulation and Cancer

Traditionally cancer has been considered as a genetic disease, which can be

dated back to the observation of aneuploidy associated with cancer cells. It becomes

more clear nowadays that epigenetic mechanisms play an important role in cancer

development (Jones and Baylin, 2002). Recently, several large scale transcriptome /

exome studies aimed at genomic analysis of tumor genetic abnormalities have again

revealed the importance of chromatin regulation in tumorigenesis (Dalgliesh et al., 2010;

Gui et al., 2011; van Haaften et al., 2009). For example, histone modifiers, such as the

Page 129: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

129

histone H3K27 demethylase UTX, were repeatedly identified to be mutated in a variety

of cancers. Besides protein complexes that directly modify histones, other protein

complexes, such as chromatin barriers, also play a role in affecting chromatin status

and oncogenesis. It has been reported that the loss of CTCF binding, which is an

insulator protein, will lead to the spreading of facultative heterochromatin into the

promoters and/or transcribed regions of tumor-suppressor genes p16 (Witcher and

Emerson, 2009) and p53 (Soto-Reyes and Recillas-Targa, 2010), resulting in the

ectopic silencing of these tumor suppressors.

The dysregulation of chromatin modification associated with tumorigenesis could

manifest as a global change in a particular modification, such as the global reduction of

H4K16 acetylation and H4K20 trimethylation observed in a skin carcinogenesis model

(Fraga et al., 2005). However, it is more common for epigenetic silencing to be

restrained to specific groups of genes, while the overall levels of suppressive histone

modification do not significantly change. For example, our data suggests that epigenetic

silencing of a group of pro-apoptotic genes promotes dMyc overexpression-induced

hyperplasia as well as scribRNAi-induced neoplastic growth. This finding is fairly

interesting to our understanding of cancer progression. Traditionally, cancer

development is considered as a multi-hit process, which requires accumulation of

multiple mutations to create the tumor potential. It has been well demonstrated that

transgenic mice expressing oncegenic Myc fail to rapidly develop tumors unless a

secondary mutation impeding apoptosis (e.g. mutation of P53, or overexpression of

BCL-2) is introduced to the animals. Our data suggests that in the presence of the

primary mutation, e.g., oncogene overexpression or loss of a tumor suppressor gene,

Page 130: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

130

aberrant regulation of epigenetic machinery is somewhat sufficient to promote tumor

progression without secondary mutations.

Is Cell Competition Relevant to Cancer?

Although many efforts have been made to understand the process of cell

competition, its impact on normal development and disease condition still remains to be

further elucidated. Cell competition has been implicated as an intrinsic mechanism to

control organ size. De la Cova et al. showed that competition between dMyc-expressing

cells and wild-type cells is essential to achieve the proper wing size, because dMyc-

induced overgrowth can be compensated by elimination of the wild type cells. In fact,

expression of dMyc from every single cell leads to an enlarged wing. When introducing

wild type clones to the disc to induce cell competition, the wing is restored to normal

size (de la Cova et al., 2004). Moreover, the recent finding of competition in the

Drosophila stem cell niche suggests its role in influencing progenitor cell selection

(Issigonis et al., 2009). Therefore, it is becoming more popular to consider cell

competition as a surveillance machinery that optimizes the fitness of progenitor cells

during normal development. From an evolutionary angle, the animals benefit from

selection of the most adaptive cells to maximize the fitness of the organism.

However, the continuous selection for super-competitors (e.g. cells with higher

levels of dMyc) may promote tumor growth and has been postulated to be the initial

stage of carcinogenesis (Baker and Li, 2008; Moreno, 2008). There are several reasons

for implicating cell competition in tumor formation. First, many genes causing cell

competition are well-defined oncegenes (e.g. dMyc) or tumor suppressor genes (e.g.

scrib, Hpo-Sal-Wts). These genes are highly conserved from flies to mammals and their

mammalian orthologues are intensively studied in cancer biology. Additionally, tumor

Page 131: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

131

development is a process of accumulating mutations that favor tumor growth or inhibit

developmental constraints, including both genetic mutations and epigenetic aberrances.

The selection process between cells with different genotypes (mutations) may be

conducted through cell competition. As aforementioned, competition between dMyc

super-competitor and normal cells results in a local expansion of super-competitors

without affecting organ size or epithelial morphology. The phenomenon is strikingly

similar to a clinical term called field cancerization, which describes the clonal expansion

of pre-tumorous cells in a particular field in the early stage of tumor development

(Dakubo et al., 2007). Additional genetic and epigenetic alterations of these cells may

transform them into malignant tumors. One major difficulty of cancer therapy is lack of

efficient detection of early stage tumors, due to their normal morphology and relatively

small size. If cell competition is really involved in the initial steps of tumor progression,

understanding the underlying mechanisms may benefit early disease prognosis. It has

to be noted that not all the genes involved in cell competition can transform cells into

super-competitors or contribute to tumor progression. For example, increasing copy

numbers of the Minute gene does not help to kill the surrounding wild type cells (Moreno

et al., 2002a; Simpson, 1979).

On the other hand, recent studies of cell competition induced by loss of neoplastic

tumor suppressors (e.g. scrib) strongly indicate its role in tumor suppression. The

precancerous scrib-mutant cells are eliminated by cell competition, which may

otherwise maldifferentiate into neoplastic tumor. So far there is no hard evidence of

whether cell competition is a cancer-promoting or cancer–inhibiting mechanism. It is

possible that different types of competitions contribute distinctly to carcinogenesis.

Page 132: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

132

These puzzles will become easier to decipher as more knowledge of cell competition is

uncovered.

Unraveling Epigenetic Regulation and Tumorigenesis in Drosophila

Although fruit flies have long served as a great model for genetic studies, they

seem to not attract much attention as a model for cancer research. One major reason is

the fact that flies rarely develop cancer in nature. Because cancer is a multi-hit process

and requires accumulation of mutations during cell proliferation, the relative short

lifespan of flies does not allow for this to occur. However, I personally consider

Drosophila as a beautiful model to study cancer due to the following reasons. First,

although flies don’t get cancer on their own, we can alternate their genome with the

powerful genetic tools available specifically for flies to induce cancer. For example, by

mutating the neoplastic tumor suppressor genes in the scrib-lgl-dlg gene family, the

imaginal discs undergo neoplastic overgrowth in larvae (Bilder et al., 2000).

Furthermore, the imaginal discs in Drosophila provide a perfect system to study cancers

of epithelial cells or carcinomas, which are the most commonly observed cancer. The

imaginal discs are a sac of epithelial cells that undergo continuous proliferation in larvae

stage and eventually differentiate into most of the adult tissues. Most of the tumor

suppressor genes identified in flies were screened with imaginal tissues. Finally, many

of the intensively studied genes are conserved between flies and humans. Indeed,

many key discoveries of cancer research were made with Drosophila, such as the

identification and characterization of Hippo pathway to control organ size, and the

socialized cellular interactions such as compensatory proliferation and cell competition.

Using Drosophila as a model, we and others also unraveled the role of epigenetic

regulation in tumorigenesis. For example, It has been noticed for some time that clones

Page 133: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

133

of cells mutated for PRC-1 components Psc-Su(z)2 or Polyhomeotic (Ph) in the

developing wing disc and eye disc display a tumor-like overgrowth phenotype, by either

up-regulating the cell cycle mediator CycB (Oktaba et al., 2008), or abnormally

activating the JAK-STAT pathway (Classen et al., 2009), or the Notch pathway

(Martinez et al., 2009).

The phenotype observed for mutant clones lacking PcG genes varies and largely

depends on the targeting genes and affected cell/tissue types. In some other cases,

there is evidence that enhancement of PcG-mediated silencing may lead to tumor-like

hyperplasia in Drosophila. For example, clones of cells mutated for dUTX have

increased levels of H3K27Me3, and significantly overgrow compared to their sister

clones (Herz et al., 2010). The overgrowth phenotype of these clones is significantly

reduced or blocked in animals heterozygous for either Pc or E(Z) mutations, indicating

that it is, at least partially, due to increased silencing of PcG-targeted genes (Herz et al.,

2010). Interestingly, in addition to homeotic genes, several Notch pathway genes had

increased H3K27Me3 modifications and reduced mRNA levels in dUTX heterozygous

animals. The interaction between dUTX and the Notch signaling pathway was also

genetically verified. Intriguingly, several other histone demethylases, such as dLSD1

and Lid (litter imaginal discs) also interact with the Notch signaling pathway (Di Stefano

et al., 2011; Mulligan et al., 2011), suggesting histone regulations play an important role

in defining the pleiotropic Notch pathway.

In this thesis study, we have reported the role of epigenetic regulation of apoptosis

in tumorigenesis. The epigenetically regulated IRER functions as the central regulatory

region for the induction of apoptosis following overproliferation and cell competition. The

Page 134: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

134

epigenetic status of IRER defines cellular sensitivity to oncogenic and competitive

stress. Cells with an accessible IRER have higher susceptibility to stress and are

preferentially eliminated; whereas cells with a condensed chromatin structure of IRER

are more resistant to stress and have higher possibilities to overgrow. Our work

highlights the importance of epigenetic mechanisms in mediating cellular sensitivity to

oncogenic stress and indicates that dysregulation of epigenetic status may contribute to

tumorigenesis.

The utility of Drosophila as a model for unraveling the role of epigenetic regulation

in tumorigenesis is emerging. There is no doubt that mechanistic studies in fruit fly will

continually provide insight on the fundamental mechanisms of chromatin regulation. In

addition, the fruit fly could also serve as valuable systems for addressing key questions

related to tumorigenesis, such as how PcG-mediated suppression is targeted to specific

genes and what pathway(s) controls epigenetic regulation of P53-targeted tumor

suppressor genes.

Perspectives

In my dissertation study, I used Drosophila as a model to investigate the role of

epigenetics, by transcriptional regulation of a group of pro-apoptotic genes, in normal

development and tumorigenesis. Our data suggests that an evolutionarily conserved

IRER is required to control the appropriate levels of apoptosis, which in turn mediates

the cell number in a given tissue and controls organ size during normal development. In

the context of tumor progression, because of, for example, overexpression of oncogene

dMyc or loss of tumor suppressor gene scrib, IRER-mediated apoptosis plays a key role

in keeping the cancerous cells in check. Furthermore, IRER is subject to epigenetic

regulation, which is dynamic during development and is responsive to environmental

Page 135: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

135

factors or genomic alterations. The dynamic and reversible epigenetic regulation

provides more plasticity to the static genome. More efforts need to be put forward to

understand the molecular mechanisms of IRER.

What Mechanism(S) Controls the Epigenetic Regulation of IRER?

The ChIP assay performed with late stage embryos detected a substantial

enrichment of repressive histone mark H3K27me3 in IRER, suggesting the epigenetic

blocking of IRER is mediated by PcG-mediated silencing. Canonical epigenetic

silencing of homeotic genes is mediated by the Polycomb Responsive Element (PRE).

However, we were not able to identify any PRE in IRER with the most updated PRE-

prediction software (Fiedler and Rehmsmeier, 2006). Instead, there are two non-coding

RNAs within IRER predicted by EST (Zhang et al., 2008). Interestingly, one exciting

breakthrough made over the past few years is to uncover long noncoding RNAs

(lncRNAs) (e.g. HOTAIR and XIST) associated with PRC binding and heterochromatin

silencing (Rinn and Chang, 2012). Thus, it is totally possible that the epigenetic

silencing of IRER is regulated by non-coding RNA. A quick and rough way to test is to

knock down the two non-coding RNAs respectively and examine its impact on the

epigenetic blocking of IRER in late stage embryos. However, it is also possible that the

epigenetic silencing is directed by other cis regulatory elements or even the lncRNAs

function in trans, which requires an unbiased screen to identify.

Which Signaling Pathways Regulate the Function IRER?

Our study showed that IRER is responsible for apoptosis following several stress

conditions including radiation and cell competition. However, little is known about the

signaling pathway that functions upstream of IRER, or the transcription factors that

response to upstream signal and bind to IRER to mediate RHG gene transcription. It

Page 136: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

136

has been well demonstrated that the immediate apoptotic response to irradiation is P53-

dependent (Brodsky et al., 2004), whereas activation of JNK plays a dominant role in

inducing apoptosis in the context of cell competition (Brumby and Richardson, 2003; de

la Cova et al., 2004; Igaki et al., 2006; Moreno and Basler, 2004; Pagliarini and Xu,

2003). Both P53 and JNK are well studied stress-responsive pathways that are highly

conserved from flies to humans. Therefore, it is necessary to investigate the relationship

between IRER and P53/JNK, for example, by examining the P53/JNK-induced cell

death in IRER-deficient background. In addition, we noticed that the absence of IRER

seems to have a more prominent effect on the cells whose survival is growth factor-

sensitive instead of cell lineage-dependent (Chapter 3). It is also worthwhile to test the

role of IRER on growth factor withdrawal-induced cell death.

What is the Relevance of our Study to Human Research?

It is important to transform and apply the knowledge we obtained in Drosophila to

humans. Up to now, we have not identified a sequence homologous sequence of IRER

in humans. However, the epigenetic mechanism established from the study on IRER

could be conserved and applicable to human research. In Chapter 3, our data indicates

a synergy between epigenetic blocking of IRER and overexpression of dMyc-induced

tumorigenesis. A similar mechanism has long been identified in mammalian cancer

research. It has been known that the mammalian ortholog of PcG protein PSC, Bmi-1,

correlates with c-Myc in promoting tumorigenesis by suppressing apoptosis (Jacobs et

al., 1999). Moreover, it has been shown that Adenovirus is able to specifically silence

P53-targeting genes to block the host response to viral infection (Soria et al., 2010b).

These observations all support the mechanistic themes of epigenetic regulation on

stress response. After we have further dissected the cis regulatory elements

Page 137: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

137

responsible for the silencing machinery of IRER, we could attempt to search for their

homologous sequence in the human genome. In addition, it will be interesting to

examine whether a similar locus control region exists in human, which can cooperatively

mediate the expression of a group of related genes, or which is subject to epigenetic

regulation.

Page 138: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

138

LIST OF REFERENCES

Abrams, J.M., White, K., Fessler, L.I., and Steller, H. (1993). Programmed cell death during Drosophila embryogenesis. Development 117, 29-43.

Adachi-Yamada, T., Fujimura-Kamada, K., Nishida, Y., and Matsumoto, K. (1999). Distortion of proximodistal information causes JNK-dependent apoptosis in Drosophila wing. Nature 400, 166-169.

Agger, K., Cloos, P.A.C., Christensen, J., Pasini, D., Rose, S., Rappsilber, J., Issaeva, I., Canaani, E., Salcini, A.E., and Helin, K. (2007). UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature 449, 731-734.

Akasaka, T., Kanno, M., Balling, R., Mieza, M.A., Taniguchi, M., and Koseki, H. (1996). A role for mel-18, a Polycomb group-related vertebrate gene, during theanteroposterior specification of the axial skeleton. Development 122, 1513-1522.

Allday, M.J. (2009). How does Epstein-Barr virus (EBV) complement the activation of Myc in the pathogenesis of Burkitt's lymphoma? Semin Cancer Biol 19, 366-376.

Anderson, R.M., Shanmuganayagam, D., and Weindruch, R. (2009). Caloric restriction and aging: studies in mice and monkeys. Toxicol Pathol 37, 47-51.

Baehrecke, E.H. (2002). How death shapes life during development. Nat Rev Mol Cell Biol 3, 779-787.

Baker, N.E., and Li, W. (2008). Cell competition and its possible relation to cancer. Cancer Res 68, 5505-5507.

Baumgartner, R., Poernbacher, I., Buser, N., Hafen, E., and Stocker, H. (2010). The WW domain protein Kibra acts upstream of Hippo in Drosophila. Dev Cell 18, 309-316.

Baylin, S.B., and Jones, P.A. (2011). A decade of exploring the cancer epigenome - biological and translational implications. Nat Rev Cancer 11, 726-734.

Bennett, F.C., and Harvey, K.F. (2006). Fat cadherin modulates organ size in Drosophila via the Salvador/Warts/Hippo signaling pathway. Curr Biol 16, 2101-2110.

Bergmann, A., Agapite, J., McCall, K., and Steller, H. (1998). The Drosophila gene hid is a direct molecular target of Ras-dependent survival signaling. Cell 95, 331-341.

Bergmann, A., Tugentman, M., Shilo, B.Z., and Steller, H. (2002). Regulation of cell number by MAPK-dependent control of apoptosis: a mechanism for trophic survival signaling. Dev Cell 2, 159-170.

Page 139: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

139

Bevis, B.J., and Glick, B.S. (2002). Rapidly maturing variants of the Discosoma red fluorescent protein (DsRed). Nat Biotechnol 20, 83-87.

Bilder, D. (2004). Epithelial polarity and proliferation control: links from the Drosophila neoplastic tumor suppressors. Genes Dev 18, 1909-1925.

Bilder, D., Li, M., and Perrimon, N. (2000). Cooperative regulation of cell polarity and growth by Drosophila tumor suppressors. Science 289, 113-116.

Bondar, T., and Medzhitov, R. (2010). p53-mediated hematopoietic stem and progenitor cell competition. Cell Stem Cell 6, 309-322.

Breen, T.R., and Harte, P.J. (1991). Molecular characterization of the trithorax gene, a positive regulator of homeotic gene expression in Drosophila. Mechanisms Dev 35, 113-127.

Brodsky, M.H., Nordstrom, W., Tsang, G., Kwan, E., Rubin, G.M., and Abrams, J.M. (2000). Drosophila p53 binds a damage response element at the reaper locus. Cell 101, 103-113.

Brodsky, M.H., Weinert, B.T., Tsang, G., Rong, Y.S., McGinnis, N.M., Golic, K.G., Rio, D.C., and Rubin, G.M. (2004). Drosophila melanogaster MNK/Chk2 and p53 regulate multiple DNA repair and apoptotic pathways following DNA damage. Mol Cell Biol 24, 1219-1231.

Brown, S., and Castelli-Gair Hombria, J. (2000). Drosophila grain encodes a GATA transcription factor required for cell rearrangement during morphogenesis. Development 127, 4867-4876.

Brumby, A.M., and Richardson, H.E. (2003). scribble mutants cooperate with oncogenic Ras or Notch to cause neoplastic overgrowth in Drosophila. EMBO J 22, 5769-5779.

Cao, R., Tsukada, Y.-i., and Zhang, Y. (2005). Role of Bmi-1 and Ring1A in H2A ubiquitylation and Hox gene silencing. Mol Cell 20, 845-854.

Cao, R., Wang, L., Wang, H., Xia, L., Erdjument-Bromage, H., Tempst, P., Jones, R.S., and Zhang, Y. (2002). Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 298, 1039-1043.

Chandraratna, D., Lawrence, N., Welchman, D.P., and Sanson, B. (2007). An in vivo model of apoptosis: linking cell behaviours and caspase substrates in embryos lacking DIAP1. J Cell Sci 120, 2594-2608.

Chen, C.L., Gajewski, K.M., Hamaratoglu, F., Bossuyt, W., Sansores-Garcia, L., Tao, C., and Halder, G. (2010). The apical-basal cell polarity determinant Crumbs regulates Hippo signaling in Drosophila. Proc Natl Acad Sci U S A 107, 15810-15815.

Page 140: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

140

Chen, C.L., Schroeder, M.C., Kango-Singh, M., Tao, C., and Halder, G. (2012). Tumor suppression by cell competition through regulation of the Hippo pathway. Proc Natl Acad Sci U S A 109, 484-489.

Chen, P., Nordstrom, W., Gish, B., and Abrams, J.M. (1996). grim, a novel cell death gene in Drosophila. Genes Dev 10, 1773-1782.

Chen, P., Rodriguez, A., Erskine, R., Thach, T., and Abrams, J.M. (1998). Dredd, a novel effector of the apoptosis activators reaper, grim, and hid in Drosophila. Dev Biol 201, 202-216.

Cho, E., Feng, Y., Rauskolb, C., Maitra, S., Fehon, R., and Irvine, K.D. (2006). Delineation of a Fat tumor suppressor pathway. Nat Genet 38, 1142-1150.

Christich, A., Kauppila, S., Chen, P., Sogame, N., Ho, S.I., and Abrams, J.M. (2002). The damage-responsive Drosophila gene sickle encodes a novel IAP binding protein similar to but distinct from reaper, grim, and hid. Curr Biol 12, 137-140.

Classen, A.K., Bunker, B.D., Harvey, K.F., Vaccari, T., and Bilder, D. (2009). A tumor suppressor activity of Drosophila Polycomb genes mediated by JAK-STAT signaling. Nat Genet 41, 1150-1155.

Conlon, I., and Raff, M. (1999). Size control in animal development. Cell 96, 235-244.

Core, N., Bel, S., Gaunt, S.J., Aurrand-Lions, M., Pearce, J., Fisher, A., and Djabali, M. (1997). Altered cellular proliferation and mesoderm patterning in Polycomb-M33-deficient mice. Development 124, 721-729.

Czermin, B., Melfi, R., McCabe, D., Seitz, V., Imhof, A., and Pirrotta, V. (2002). Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites. Cell 111, 185-196.

Dakubo, G.D., Jakupciak, J.P., Birch-Machin, M.A., and Parr, R.L. (2007). Clinical implications and utility of field cancerization. Cancer Cell Int 7, 2.

Dalgliesh, G.L., Furge, K., Greenman, C., Chen, L., Bignell, G., Butler, A., Davies, H., Edkins, S., Hardy, C., Latimer, C., et al. (2010). Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463, 360-363.

de Beco, S., Ziosi, M., and Johnston, L.A. (2012). New frontiers in cell competition. Dev Dyn 241, 831-841.

de la Cova, C., Abril, M., Bellosta, P., Gallant, P., and Johnston, L.A. (2004). Drosophila myc regulates organ size by inducing cell competition. Cell 117, 107-116.

Page 141: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

141

de Napoles, M., Mermoud, J.E., Wakao, R., Tang, Y.A., Endoh, M., Appanah, R., Nesterova, T.B., Silva, J., Otte, A.P., Vidal, M., et al. (2004). Polycomb group proteins Ring1A/B link ubiquitylation of histone H2A to heritable gene silencing and X inactivation. Dev Cell 7, 663-676.

del Mar Lorente, M., Marcos-Gutierrez, C., Perez, C., Schoorlemmer, J., Ramirez, A., Magin, T., and Vidal, M. (2000). Loss- and gain-of-function mutations show a polycomb group function for Ring1A in mice. Development 127, 5093-5100.

Di Stefano, L., Walker, J.A., Burgio, G., Corona, D.F.V., Mulligan, P., Näär, A.M., and Dyson, N.J. (2011). Functional antagonism between histone H3K4 demethylases in vivo. Genes Dev 25, 17-28.

Ebert, A., Schotta, G., Lein, S., Kubicek, S., Krauss, V., Jenuwein, T., and Reuter, G. (2004). Su(var) genes regulate the balance between euchromatin and heterochromatin in Drosophila. Gene Dev 18, 2973-2983.

Engstrom, P.G., Ho Sui, S.J., Drivenes, O., Becker, T.S., and Lenhard, B. (2007). Genomic regulatory blocks underlie extensive microsynteny conservation in insects. Genome Res 17, 1898-1908.

Fan, Y., and Bergmann, A. (2008). Apoptosis-induced compensatory proliferation. The Cell is dead. Long live the Cell! Trends Cell Biol 18, 467-473.

Fiedler, T., and Rehmsmeier, M. (2006). jPREdictor: a versatile tool for the prediction of cis-regulatory elements. Nucleic Acids Res 34, W546-550.

Fraga, M.F., Ballestar, E., Villar-Garea, A., Boix-Chornet, M., Espada, J., Schotta, G., Bonaldi, T., Haydon, C., Ropero, S., Petrie, K., et al. (2005). Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet 37, 391-400.

Friedberg, E.C., G. C. Walker, and W. Siede. (1995). DNA repair and mutagenesis. ASM Press, Washington, DC.

Gallant, P. (2009). Drosophila Myc. Adv Cancer Res 103, 111-144.

Gazin, C., Wajapeyee, N., Gobeil, S., Virbasius, C.-M., and Green, M.R. (2007a). An elaborate pathway required for Ras-mediated epigenetic silencing. Nature 449, 1073-1077.

Gazin, C., Wajapeyee, N., Gobeil, S., Virbasius, C.M., and Green, M.R. (2007b). An elaborate pathway required for Ras-mediated epigenetic silencing. Nature 449, 1073-1077.

Genevet, A., Wehr, M.C., Brain, R., Thompson, B.J., and Tapon, N. (2010). Kibra is a regulator of the Salvador/Warts/Hippo signaling network. Dev Cell 18, 300-308.

Page 142: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

142

Gibson, M.C., and Perrimon, N. (2005). Extrusion and death of DPP/BMP-compromised epithelial cells in the developing Drosophila wing. Science 307, 1785-1789.

Gilchrist, A.S., and Partridge, L. (1999). A comparison of the genetic basis of wing size divergence in three parallel body size clines of Drosophila melanogaster. Genetics 153, 1775-1787.

Gong, W.J., and Golic, K.G. (2003). Ends-out, or replacement, gene targeting in Drosophila. Proc Natl Acad Sci U S A 100, 2556-2561.

Goyal, L., McCall, K., Agapite, J., Hartwieg, E., and Steller, H. (2000). Induction of apoptosis by Drosophila reaper, hid and grim through inhibition of IAP function. EMBO J 19, 589-597.

Grether, M.E., Abrams, J.M., Agapite, J., White, K., and Steller, H. (1995). The head involution defective gene of Drosophila melanogaster functions in programmed cell death. Genes Dev 9, 1694-1708.

Grewal, S.I., and Moazed, D. (2003). Heterochromatin and epigenetic control of gene expression. Science 301, 798-802.

Grzeschik, N.A., Parsons, L.M., Allott, M.L., Harvey, K.F., and Richardson, H.E. (2010). Lgl, aPKC, and Crumbs regulate the Salvador/Warts/Hippo pathway through two distinct mechanisms. Curr Biol 20, 573-581.

Gui, Y., Guo, G., Huang, Y., Hu, X., Tang, A., Gao, S., Wu, R., Chen, C., Li, X., Zhou, L., et al. (2011). Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet advance online publication.

Gupta, R.A., Shah, N., Wang, K.C., Kim, J., Horlings, H.M., Wong, D.J., Tsai, M.-C., Hung, T., Argani, P., Rinn, J.L., et al. (2010). Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464, 1071-1076.

Halder, G., and Johnson, R.L. (2011). Hippo signaling: growth control and beyond. Development 138, 9-22.

Hamaratoglu, F., Willecke, M., Kango-Singh, M., Nolo, R., Hyun, E., Tao, C., Jafar-Nejad, H., and Halder, G. (2006). The tumour-suppressor genes NF2/Merlin and Expanded act through Hippo signalling to regulate cell proliferation and apoptosis. Nat Cell Biol 8, 27-36.

Hariharan, I.K., and Bilder, D. (2006). Regulation of imaginal disc growth by tumor-suppressor genes in Drosophila. Annu Rev Genet 40, 335-361.

Harrington, E.A., Bennett, M.R., Fanidi, A., and Evan, G.I. (1994). c-Myc-induced apoptosis in fibroblasts is inhibited by specific cytokines. EMBO J 13, 3286-3295.

Page 143: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

143

Henikoff, S. (1992). Position effect and related phenomena. Curr Opin Genet Dev 2, 907-912.

Herz, H.M., Madden, L.D., Chen, Z., Bolduc, C., Buff, E., Gupta, R., Davuluri, R., Shilatifard, A., Hariharan, I.K., and Bergmann, A. (2010). The H3K27me3 demethylase dUTX is a suppressor of Notch- and Rb-dependent tumors in Drosophila. Mol Cell Biol 30, 2485-2497.

Heydari, A.R., Unnikrishnan, A., Lucente, L.V., and Richardson, A. (2007). Caloric restriction and genomic stability. Nucleic Acids Res 35, 7485-7496.

Hipfner, D.R., and Cohen, S.M. (2004). Connecting proliferation and apoptosis in development and disease. Nat Rev Mol Cell Biol 5, 805-815.

Hoffman, B., and Liebermann, D.A. (2008). Apoptotic signaling by c-MYC. Oncogene 27, 6462-6472.

Hong, S., Cho, Y.-W., Yu, L.-R., Yu, H., Veenstra, T.D., and Ge, K. (2007). Identification of JmjC domain-containing UTX and JMJD3 as histone H3 lysine 27 demethylases. Proc Natl Acad Sci USA 104, 18439-18444.

Igaki, T., Kanda, H., Yamamoto-Goto, Y., Kanuka, H., Kuranaga, E., Aigaki, T., and Miura, M. (2002). Eiger, a TNF superfamily ligand that triggers the Drosophila JNK pathway. EMBO J 21, 3009-3018.

Igaki, T., Pagliarini, R.A., and Xu, T. (2006). Loss of cell polarity drives tumor growth and invasion through JNK activation in Drosophila. Curr Biol 16, 1139-1146.

Igaki, T., Pastor-Pareja, J.C., Aonuma, H., Miura, M., and Xu, T. (2009). Intrinsic tumor suppression and epithelial maintenance by endocytic activation of Eiger/TNF signaling in Drosophila. Dev Cell 16, 458-465.

Isles, A.R., and Wilkinson, L.S. (2008). Epigenetics: what is it and why is it important to mental disease? Br Med Bull 85, 35-45.

Issigonis, M., Tulina, N., de Cuevas, M., Brawley, C., Sandler, L., and Matunis, E. (2009). JAK-STAT signal inhibition regulates competition in the Drosophila testis stem cell niche. Science 326, 153-156.

Jacobs, J.J., Scheijen, B., Voncken, J.W., Kieboom, K., Berns, A., and van Lohuizen, M. (1999). Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev 13, 2678-2690.

Jassim, O.W., Fink, J.L., and Cagan, R.L. (2003). Dmp53 protects the Drosophila retina during a developmentally regulated DNA damage response. EMBO J 22, 5622-5632.

Page 144: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

144

Jenuwein, T., and Allis, C.D. (2001). Translating the Histone Code. Science 293, 1074-1080.

Jiang, C., Baehrecke, E.H., and Thummel, C.S. (1997). Steroid regulated programmed cell death during Drosophila metamorphosis. Development 124, 4673-4683.

Johnston, L.A., Prober, D.A., Edgar, B.A., Eisenman, R.N., and Gallant, P. (1999). Drosophila myc regulates cellular growth during development. Cell 98, 779-790.

Johnstone, S.E., and Baylin, S.B. (2010). Stress and the epigenetic landscape: a link to the pathobiology of human diseases? Nat Rev Genet 11, 806-812.

Jones, P.A., and Baylin, S.B. (2002). The fundamental role of epigenetic events in cancer. Nat Rev Genet 3, 415-428.

Jurgens, G. (1985). A group of genes controlling the spatial expression of the bithorax complex in Drosophila. Nature 316, 153-155.

Karpen, G.H. (1994). Position-effect variegation and the new biology of heterochromatin. Curr Opin Genet Dev 4, 281-291.

Kel, A.E., Gossling, E., Reuter, I., Cheremushkin, E., Kel-Margoulis, O.V., and Wingender, E. (2003). MATCH: A tool for searching transcription factor binding sites in DNA sequences. Nucleic Acids Res 31, 3576-3579.

Kerr, J.F., Wyllie, A.H., and Currie, A.R. (1972). Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26, 239-257.

Kikuta, H., Laplante, M., Navratilova, P., Komisarczuk, A.Z., Engstrom, P.G., Fredman, D., Akalin, A., Caccamo, M., Sealy, I., Howe, K., et al. (2007). Genomic regulatory blocks encompass multiple neighboring genes and maintain conserved synteny in vertebrates. Genome Res 17, 545-555.

Kimura, K., Kodama, A., Hayasaka, Y., and Ohta, T. (2004). Activation of the cAMP/PKA signaling pathway is required for post-ecdysial cell death in wing epidermal cells of Drosophila melanogaster. Development 131, 1597-1606.

Konev, A.Y., Yan, C.M., Acevedo, D., Kennedy, C., Ward, E., Lim, A., Tickoo, S., and Karpen, G.H. (2003). Genetics of P-element transposition into Drosophila melanogaster centric heterochromatin. Genetics 165, 2039-2053.

Kornbluth, S., and White, K. (2005). Apoptosis in Drosophila: neither fish nor fowl (nor man, nor worm). J Cell Sci 118, 1779-1787.

Kurada, P., and White, K. (1998). Ras promotes cell survival in Drosophila by downregulating hid expression. Cell 95, 319-329.

Page 145: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

145

Lee, M.G., Norman, J., Shilatifard, A., and Shiekhattar, R. (2007). Physical and functional association of a trimethyl H3K4 demethylase and Ring6a/MBLR, a Polycomb-like protein. Cell 128, 877-887.

Letai, A., Sorcinelli, M.D., Beard, C., and Korsmeyer, S.J. (2004). Antiapoptotic BCL-2 is required for maintenance of a model leukemia. Cancer Cell 6, 241-249.

Levine, S.S., Weiss, A., Erdjument-Bromage, H., Shao, Z., Tempst, P., and Kingston, R.E. (2002). The core of the Polycomb repressive complex is compositionally and functionally conserved in flies and humans. Mol Cell Biol 22, 6070-6078.

Lewis, E.B. (1978). A gene complex controlling segmentation in Drosophila. Nature 276, 565-570.

Lewis, P.H. (1947). D.melanogaster new mutants: Report of Pamela H. Lewis. Drosophila Inform Ser. 21, 69.

Li, W., and Baker, N.E. (2007). Engulfment is required for cell competition. Cell 129, 1215-1225.

Lin, N., Li, X., Cui, K., Chepelev, I., Tie, F., Liu, B., Li, G., Harte, P., Zhao, K., Huang, S., et al. (2011). A barrier-only boundary element delimits the formation of facultative heterochromatin in Drosophila melanogaster and vertebrates. Mol Cell Biol 31, 2729-2741.

Lin, N., Zhang, C., Pang, J., and Zhou, L. (2009). By design or by chance: cell death during Drosophila embryogenesis. Apoptosis 14, 935-942.

Ling, C., Zheng, Y., Yin, F., Yu, J., Huang, J., Hong, Y., Wu, S., and Pan, D. (2010). The apical transmembrane protein Crumbs functions as a tumor suppressor that regulates Hippo signaling by binding to Expanded. Proc Natl Acad Sci U S A 107, 10532-10537.

Lohmann, I., McGinnis, N., Bodmer, M., and McGinnis, W. (2002). The Drosophila Hox gene deformed sculpts head morphology via direct regulation of the apoptosis activator reaper. Cell 110, 457-466.

Luo, X., Puig, O., Hyun, J., Bohmann, D., and Jasper, H. (2007). Foxo and Fos regulate the decision between cell death and survival in response to UV irradiation. EMBO J 26, 380-390.

Lyko, F., Ramsahoye, B.H., and Jaenisch, R. (2000). DNA methylation in Drosophila melanogaster. Nature 408, 538-540.

Martin-Castellanos, C., and Edgar, B.A. (2002). A characterization of the effects of Dpp signaling on cell growth and proliferation in the Drosophila wing. Development 129, 1003-1013.

Page 146: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

146

Martinez, A.M., Schuettengruber, B., Sakr, S., Janic, A., Gonzalez, C., and Cavalli, G. (2009). Polyhomeotic has a tumor suppressor activity mediated by repression of Notch signaling. Nat Genet 41, 1076-1082.

McCall, K., and Bender, W. (1996). Probes of chromatin accessibility in the Drosophila bithorax complex respond differently to Polycomb-mediated repression. EMBO J 15, 569-580.

McEwen, D.G., and Peifer, M. (2005). Puckered, a Drosophila MAPK phosphatase, ensures cell viability by antagonizing JNK-induced apoptosis. Development 132, 3935-3946.

McNamee, L.M., and Brodsky, M.H. (2009). p53-independent apoptosis limits DNA damage-induced aneuploidy. Genetics 182, 423-435.

Meyer, N., Kim, S.S., and Penn, L.Z. (2006). The Oscar-worthy role of Myc in apoptosis. Semin Cancer Biol 16, 275-287.

Michalopoulos, G.K., and DeFrances, M.C. (1997). Liver regeneration. Science 276, 60-66.

Milan, M., Campuzano, S., and Garcia-Bellido, A. (1997). Developmental parameters of cell death in the wing disc of Drosophila. Proc Natl Acad Sci U S A 94, 5691-5696.

Mills, A.A. (2010). Throwing the cancer switch: reciprocal roles of polycomb and trithorax proteins. Nat Rev Cancer 10, 669-682.

Montero, L., Muller, N., and Gallant, P. (2008). Induction of apoptosis by Drosophila Myc. Genesis 46, 104-111.

Moon, N.S., Di Stefano, L., Morris, E.J., Patel, R., White, K., and Dyson, N.J. (2008). E2F and p53 induce apoptosis independently during Drosophila development but intersect in the context of DNA damage. PLoS Genet 4, e1000153.

Morata, G., and Ripoll, P. (1975). Minutes: mutants of drosophila autonomously affecting cell division rate. Dev Biol 42, 211-221.

Moreno, E. (2008). Is cell competition relevant to cancer? Nat Rev Cancer 8, 141-147.

Moreno, E., and Basler, K. (2004). dMyc transforms cells into super-competitors. Cell 117, 117-129.

Moreno, E., Basler, K., and Morata, G. (2002a). Cells compete for decapentaplegic survival factor to prevent apoptosis in Drosophila wing development. Nature 416, 755-759.

Page 147: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

147

Moreno, E., Yan, M., and Basler, K. (2002b). Evolution of TNF signaling mechanisms: JNK-dependent apoptosis triggered by Eiger, the Drosophila homolog of the TNF superfamily. Curr Biol 12, 1263-1268.

Muller, H.J. (1932). Further studies on the nature and causes of gene mutations. Proc 6th Int Congr Genet 1, 213-255.

Müller, J., Hart, C.M., Francis, N.J., Vargas, M.L., Sengupta, A., Wild, B., Miller, E.L., O'Connor, M.B., Kingston, R.E., and Simon, J.A. (2002). Histone methyltransferase activity of a Drosophila Polycomb group repressor complex. Cell 111, 197-208.

Mulligan, P., Yang, F., Di Stefano, L., Ji, J.-Y., Ouyang, J., Nishikawa, J.L., Toiber, D., Kulkarni, M., Wang, Q., Najafi-Shoushtari, S.H., et al. (2011). A SIRT1-LSD1 corepressor complex regulates Notch target gene expression and development. Mol Cell 42, 689-699.

Nanty, L., Carbajosa, G., Heap, G.A., Ratnieks, F., van Heel, D.A., Down, T.A., and Rakyan, V.K. (2011). Comparative methylomics reveals gene-body H3K36me3 in Drosophila predicts DNA methylation and CpG landscapes in other invertebrates. Genome Res 21, 1841-1850.

Ni, J.Q., Zhou, R., Czech, B., Liu, L.P., Holderbaum, L., Yang-Zhou, D., Shim, H.S., Tao, R., Handler, D., Karpowicz, P., et al. (2011). A genome-scale shRNA resource for transgenic RNAi in Drosophila. Nat Methods 8, 405-407.

O'Donnell, K.H., Chen, C.T., and Wensink, P.C. (1994). Insulating DNA directs ubiquitous transcription of the Drosophila melanogaster alpha 1-tubulin gene. Mol Cell Biol 14, 6398-6408.

Ohm, J.E., McGarvey, K.M., Yu, X., Cheng, L., Schuebel, K.E., Cope, L., Mohammad, H.P., Chen, W., Daniel, V.C., Yu, W., et al. (2007). A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet 39, 237-242.

Oktaba, K., Gutierrez, L., Gagneur, J., Girardot, C., Sengupta, A.K., Furlong, E.E., and Muller, J. (2008). Dynamic regulation by polycomb group protein complexes controls pattern formation and the cell cycle in Drosophila. Dev Cell 15, 877-889.

Ollmann, M., Young, L.M., Di Como, C.J., Karim, F., Belvin, M., Robertson, S., Whittaker, K., Demsky, M., Fisher, W.W., Buchman, A., et al. (2000). Drosophila p53 is a structural and functional homolog of the tumor suppressor p53. Cell 101, 91-101.

Orian, A., van Steensel, B., Delrow, J., Bussemaker, H.J., Li, L., Sawado, T., Williams, E., Loo, L.W., Cowley, S.M., Yost, C., et al. (2003). Genomic binding by the Drosophila Myc, Max, Mad/Mnt transcription factor network. Genes Dev 17, 1101-1114.

Page 148: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

148

Orlando, V., and Paro, R. (1995). Chromatin multiprotein complexes involved in the maintenance of transcription patterns. Curr Opin Genet Dev 5, 174-179.

Pagliarini, R.A., and Xu, T. (2003). A genetic screen in Drosophila for metastatic behavior. Science 302, 1227-1231.

Pan, D. (2010). The hippo signaling pathway in development and cancer. Dev Cell 19, 491-505.

Parker, J. (2006). Control of compartment size by an EGF ligand from neighboring cells. Curr Biol 16, 2058-2065.

Pazdera, T.M., Janardhan, P., and Minden, J.S. (1998). Patterned epidermal cell death in wild-type and segment polarity mutant Drosophila embryos. Development 125, 3427-3436.

Pearson, J.C., Lemons, D., and McGinnis, W. (2005). Modulating Hox gene functions during animal body patterning. Nat Rev Genet 6, 893-904.

Pelengaris, S., Khan, M., and Evan, G.I. (2002). Suppression of Myc-induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell 109, 321-334.

Pennacchio, L.A., Ahituv, N., Moses, A.M., Prabhakar, S., Nobrega, M.A., Shoukry, M., Minovitsky, S., Dubchak, I., Holt, A., Lewis, K.D., et al. (2006). In vivo enhancer analysis of human conserved non-coding sequences. Nature 444, 499-502.

Perez-Garijo, A., Martin, F.A., and Morata, G. (2004). Caspase inhibition during apoptosis causes abnormal signalling and developmental aberrations in Drosophila. Development 131, 5591-5598.

Peterson, C., Carney, G.E., Taylor, B.J., and White, K. (2002). reaper is required for neuroblast apoptosis during Drosophila development. Development 129, 1467-1476.

Pirrotta, V. (1997). Chromatin-silencing mechanisms in Drosophila maintain patterns of gene expression. Trends Genet 13, 314-318.

Pujadas, E., and Feinberg, A.P. (2012). Regulated noise in the epigenetic landscape of development and disease. Cell 148, 1123-1131.

Quinn, L.M., Dorstyn, L., Mills, K., Colussi, P.A., Chen, P., Coombe, M., Abrams, J., Kumar, S., and Richardson, H. (2000). An essential role for the caspase dronc in developmentally programmed cell death in Drosophila. J Biol Chem 275, 40416-40424.

Reik, W. (2007). Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447, 425-432.

Page 149: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

149

Rhiner, C., Lopez-Gay, J.M., Soldini, D., Casas-Tinto, S., Martin, F.A., Lombardia, L., and Moreno, E. (2010). Flower forms an extracellular code that reveals the fitness of a cell to its neighbors in Drosophila. Dev Cell 18, 985-998.

Rinn, J.L., and Chang, H.Y. (2012). Genome Regulation by Long Noncoding RNAs. Annu Rev Biochem 81, 145-166.

Robinson, B.S., Huang, J., Hong, Y., and Moberg, K.H. (2010). Crumbs regulates Salvador/Warts/Hippo signaling in Drosophila via the FERM-domain protein Expanded. Curr Biol 20, 582-590.

Ryder, E., Blows, F., Ashburner, M., Bautista-Llacer, R., Coulson, D., Drummond, J., Webster, J., Gubb, D., Gunton, N., Johnson, G., et al. (2004). The DrosDel collection: a set of P-element insertions for generating custom chromosomal aberrations in Drosophila melanogaster. Genetics 167, 797-813.

Ryoo, H.D., Gorenc, T., and Steller, H. (2004). Apoptotic cells can induce compensatory cell proliferation through the JNK and the Wingless signaling pathways. Dev Cell 7, 491-501.

Salvesen, G.S., and Duckett, C.S. (2002). IAP proteins: blocking the road to death's door. Nat Rev Mol Cell Biol 3, 401-410.

Santos-Rosa, H., Schneider, R., Bannister, A.J., Sherriff, J., Bernstein, B.E., Emre, N.C.T., Schreiber, S.L., Mellor, J., and Kouzarides, T. (2002). Active genes are tri-methylated at K4 of histone H3. Nature 419, 407-411.

Saurin, A.J., Shao, Z., Erdjument-Bromage, H., Tempst, P., and Kingston, R.E. (2001). A Drosophila Polycomb group complex includes Zeste and dTAFII proteins. Nature 412, 655-660.

Schmitt, S., Prestel, M., and Paro, R. (2005). Intergenic transcription through a polycomb group response element counteracts silencing. Genes Dev 19, 697-708.

Schwartz, Y.B., and Pirrotta, V. (2007). Polycomb silencing mechanisms and the management of genomic programmes. Nat Rev Genet 8, 9-22.

Scott, C.L., Schuler, M., Marsden, V.S., Egle, A., Pellegrini, M., Nesic, D., Gerondakis, S., Nutt, S.L., Green, D.R., and Strasser, A. (2004). Apaf-1 and caspase-9 do not act as tumor suppressors in myc-induced lymphomagenesis or mouse embryo fibroblast transformation. J Cell Biol 164, 89-96.

Sexton, T., Yaffe, E., Kenigsberg, E., Bantignies, F., Leblanc, B., Hoichman, M., Parrinello, H., Tanay, A., and Cavalli, G. (2012). Three-dimensional folding and functional organization principles of the Drosophila genome. Cell 148, 458-472.

Page 150: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

150

Shao, Z., Raible, F., Mollaaghababa, R., Guyon, J.R., Wu, C.-t., Bender, W., and Kingston, R.E. (1999). Stabilization of chromatin structure by PRC1, a Polycomb complex. Cell 98, 37-46.

Shen, J., and Dahmann, C. (2005). Extrusion of cells with inappropriate Dpp signaling from Drosophila wing disc epithelia. Science 307, 1789-1790.

Shi, Y. (2007). Histone lysine demethylases: emerging roles in development, physiology and disease. Nat Rev Genet 8, 829-833.

Silva, E., Tsatskis, Y., Gardano, L., Tapon, N., and McNeill, H. (2006). The tumor-suppressor gene fat controls tissue growth upstream of expanded in the hippo signaling pathway. Curr Biol 16, 2081-2089.

Simpson, P. (1979). Parameters of cell competition in the compartments of the wing disc of Drosophila. Dev Biol 69, 182-193.

Simpson, P., and Morata, G. (1981). Differential mitotic rates and patterns of growth in compartments in the Drosophila wing. Dev Biol 85, 299-308.

Smith, C.L., and Peterson, C.L. (2005). ATP-dependent chromatin remodeling. Curr Top Dev Biol 65, 115-148.

Sogame, N., Kim, M., and Abrams, J.M. (2003). Drosophila p53 preserves genomic stability by regulating cell death. Proc Natl Acad Sci U S A 100, 4696-4701.

Song, Z., and Steller, H. (1999). Death by design: mechanism and control of apoptosis. Trends Cell Biol 9, M49-52.

Soria, C., Estermann, F.E., Espantman, K.C., and O'Shea, C.C. (2010a). Heterochromatin silencing of p53 target genes by a small viral protein. Nature 466, 1076-1081.

Soria, C., Estermann, F.E., Espantman, K.C., and O'Shea, C.C. (2010b). Heterochromatin silencing of p53 target genes by a small viral protein. Nature 466, 1076-1081.

Soto-Reyes, E., and Recillas-Targa, F. (2010). Epigenetic regulation of the human p53 gene promoter by the CTCF transcription factor in transformed cell lines. Oncogene 29, 2217-2227.

Sparmann, A., and van Lohuizen, M. (2006). Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer 6, 846-856.

Srinivasula, S.M., Datta, P., Kobayashi, M., Wu, J.W., Fujioka, M., Hegde, R., Zhang, Z., Mukattash, R., Fernandes-Alnemri, T., Shi, Y., et al. (2002). sickle, a novel Drosophila death gene in the reaper/hid/grim region, encodes an IAP-inhibitory protein. Curr Biol 12, 125-130.

Page 151: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

151

Steller, H. (2008). Regulation of apoptosis in Drosophila. Cell Death Differ 15, 1132-1138.

Strahl, B.D., and Allis, C.D. (2000). The language of covalent histone modifications. Nature 403, 41-45.

Strasser, A., Harris, A.W., Bath, M.L., and Cory, S. (1990). Novel primitive lymphoid tumours induced in transgenic mice by cooperation between myc and bcl-2. Nature 348, 331-333.

Struhl, G. (1983). Role of the esc+ gene product in ensuring the selective expression of segment-specific homeotic genes in Drosophila. J Embryol Exp Morphol 76, 297-331.

Sullivan, W., Ashburner, M., and Hawley, R.S. (2000). Drosophila Protocols, 1 edn (Cold Spring Harbor Laboratory Press).

Sun, F.L., Haynes, K., Simpson, C.L., Lee, S.D., Collins, L., Wuller, J., Eissenberg, J.C., and Elgin, S.C. (2004). cis-Acting determinants of heterochromatin formation on Drosophila melanogaster chromosome four. Mol Cell Biol 24, 8210-8220.

Supatto, W., Debarre, D., Moulia, B., Brouzes, E., Martin, J.-L., Farge, E., and Beaurepaire, E. (2005). In vivo modulation of morphogenetic movements in Drosophila embryos with femtosecond laser pulses. PNAS 102, 1047-1052.

Tan, Y., Yamada-Mabuchi, M., Arya, R., St Pierre, S., Tang, W., Tosa, M., Brachmann, C., and White, K. (2011). Coordinated expression of cell death genes regulates neuroblast apoptosis. Development 138, 2197-2206.

Tyler, D.M., Li, W., Zhuo, N., Pellock, B., and Baker, N.E. (2007). Genes affecting cell competition in Drosophila. Genetics 175, 643-657.

Urist, M., Tanaka, T., Poyurovsky, M.V., and Prives, C. (2004). p73 induction after DNA damage is regulated by checkpoint kinases Chk1 and Chk2. Genes Dev 18, 3041-3054.

van Haaften, G., Dalgliesh, G.L., Davies, H., Chen, L., Bignell, G., Greenman, C., Edkins, S., Hardy, C., O'Meara, S., Teague, J., et al. (2009). Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet 41, 521-523.

Vaux, D.L., and Silke, J. (2005). IAPs, RINGs and ubiquitylation. Nat Rev Mol Cell Biol 6, 287-297.

Vignali, M., Hassan, A.H., Neely, K.E., and Workman, J.L. (2000). ATP-dependent chromatin-remodeling complexes. Mol Cell Biol 20, 1899-1910.

Page 152: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

152

Wang, H., Wang, L., Erdjument-Bromage, H., Vidal, M., Tempst, P., Jones, R.S., and Zhang, Y. (2004). Role of histone H2A ubiquitination in Polycomb silencing. Nature 431, 873-878.

Wang, S.L., Hawkins, C.J., Yoo, S.J., Muller, H.A., and Hay, B.A. (1999). The Drosophila caspase inhibitor DIAP1 is essential for cell survival and is negatively regulated by HID. Cell 98, 453-463.

Werz, C., Lee, T.V., Lee, P.L., Lackey, M., Bolduc, C., Stein, D.S., and Bergmann, A. (2005). Mis-specified cells die by an active gene-directed process, and inhibition of this death results in cell fate transformation in Drosophila. Development 132, 5343-5352.

White, K., Grether, M.E., Abrams, J.M., Young, L., Farrell, K., and Steller, H. (1994). Genetic control of programmed cell death in Drosophila. Science 264, 677-683.

Wichmann, A., Jaklevic, B., and Su, T.T. (2006). Ionizing radiation induces caspase-dependent but Chk2- and p53-independent cell death in Drosophila melanogaster. Proc Natl Acad Sci U S A 103, 9952-9957.

Willecke, M., Hamaratoglu, F., Kango-Singh, M., Udan, R., Chen, C.L., Tao, C., Zhang, X., and Halder, G. (2006). The fat cadherin acts through the hippo tumor-suppressor pathway to regulate tissue size. Curr Biol 16, 2090-2100.

Wing, J.P., Karres, J.S., Ogdahl, J.L., Zhou, L., Schwartz, L.M., and Nambu, J.R. (2002). Drosophila sickle is a novel grim-reaper cell death activator. Curr Biol 12, 131-135.

Witcher, M., and Emerson, B.M. (2009). Epigenetic silencing of the p16(INK4a) tumor suppressor is associated with loss of CTCF binding and a chromatin boundary. Mol Cell 34, 271-284.

Wodarz, A. (2000). Tumor suppressors: linking cell polarity and growth control. Curr Biol 10, R624-626.

Wolff, T., and Ready, D.F. (1991). Cell death in normal and rough eye mutants of Drosophila. Development 113, 825-839.

Woolfe, A., Goodson, M., Goode, D.K., Snell, P., McEwen, G.K., Vavouri, T., Smith, S.F., North, P., Callaway, H., Kelly, K., et al. (2005). Highly conserved non-coding sequences are associated with vertebrate development. PLoS Biol 3, e7.

Wu, J.N., Nguyen, N., Aghazarian, M., Tan, Y., Sevrioukov, E.A., Mabuchi, M., Tang, W., Monserrate, J.P., White, K., and Brachmann, C.B. (2010). grim promotes programmed cell death of Drosophila microchaete glial cells. Mech Dev 127, 407-417.

Page 153: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

153

Xu, D., Li, Y., Arcaro, M., Lackey, M., and Bergmann, A. (2005). The CARD-carrying caspase Dronc is essential for most, but not all, developmental cell death in Drosophila. Development 132, 2125-2134.

Xu, D., Wang, Y., Willecke, R., Chen, Z., Ding, T., and Bergmann, A. (2006). The effector caspases drICE and dcp-1 have partially overlapping functions in the apoptotic pathway in Drosophila. Cell Death Differ 13, 1697-1706.

Yu, J., Zheng, Y., Dong, J., Klusza, S., Deng, W.M., and Pan, D. (2010). Kibra functions as a tumor suppressor protein that regulates Hippo signaling in conjunction with Merlin and Expanded. Dev Cell 18, 288-299.

Zhang, Y., Lin, N., Carroll, P.M., Chan, G., Guan, B., Xiao, H., Yao, B., Wu, S.S., and Zhou, L. (2008). Epigenetic blocking of an enhancer region controls irradiation-induced proapoptotic gene expression in Drosophila embryos. Dev Cell 14, 481-493.

Zhao, B., Tumaneng, K., and Guan, K.L. (2011). The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nat Cell Biol 13, 877-883.

Zhou, L., Hashimi, H., Schwartz, L.M., and Nambu, J.R. (1995). Programmed cell death in the Drosophila central nervous system midline. Curr Biol 5, 784-790.

Zhou, L., Schnitzler, A., Agapite, J., Schwartz, L.M., Steller, H., and Nambu, J.R. (1997). Cooperative functions of the reaper and head involution defective genes in the programmed cell death of Drosophila central nervous system midline cells. Proc Natl Acad Sci U S A 94, 5131-5136.

Page 154: © 2012 Can Zhang - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/45/63/00001/ZHANG_C.pdfDIAP Drosophila inhibitor of apoptosis protein H3K9me3 Trimethylated histone 3 lysine

154

BIOGRAPHICAL SKETCH

Can was born in Jining, Shandong province, P.R. China in 1983. She received her

Bachelor of Science in biological sciences from the Ocean University of China

(Qingdao) in 2005 and immediately started the Ph.D. study in marine biology also from

the Ocean University of China. One year later, her husband Bing Yao was admitted to

the Ph.D. program at the University of Florida in US. Can made a lifetime decision of

giving up her Ph.D. career in China and traveling to US to join her husband. After she

came to the US, Can volunteered in Dr. Lei Zhou’s laboratory for one year and gained

some valuable research experience. To pursue further education, she started her Ph.D.

study in the interdisciplinary program in biomedical sciences at the University of Florida

in 2007. She then officially joined Zhou laboratory in 2008 and passed her qualifying

examination in October 2009. She received her Ph.D. degree in August 2012.

After graduation, Can will continue her research career as a postdoctoral fellow in

Dr. Kenneth Moberg’s laboratory at the Emory University. Outside of the science life,

Can enjoys watching soccer games with her husband, and relaxes through reading,

sleeping, and traveling.