chaf1a regulates prc2-mediated epigenetic memory · pdf filechromatin and epigenetics ... gene...

154
CHAF1A Regulates PRC2-mediated Epigenetic Memory by Mina Rafiei A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Laboratory Medicine and Pathobiology University of Toronto © Copyright by Mina Rafiei 2014

Upload: trinhmien

Post on 06-Feb-2018

220 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

CHAF1A Regulates PRC2-mediated Epigenetic Memory

by

Mina Rafiei

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Department of Laboratory Medicine and Pathobiology University of Toronto

© Copyright by Mina Rafiei 2014

Page 2: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

ii

CHAF1A Regulates PRC2-mediated Epigenetic Memory

Mina Rafiei

Doctor of Philosophy

Department of Laboratory Medicine and Pathobiology University of Toronto

2014

Abstract

During DNA replication both genetic and epigenetic information on DNA and nucleosomes

needs to be faithfully reproduced. Although DNA replication is well described, the mechanism

of nucleosome and epigenetic replication is not well understood. We studied this issue in the

context of the IFN!-responsive genes (ISGs). Previous work in our lab showed that in the

BRG1-deficient SW13 adenocarcinoma cells, the Polycomb Repressive Complex 2 (PRC2)

blocks IFN!-induction of CIITA and a subset of other ISGs. We detected the repressive PRC2–

mediated epigenetic mark, H3K27me3, across the CIITA locus and at the promoters of other

PRC2-regulated ISGs. We hypothesized that other unknown factors collaborate with PRC2 in

repression of ISGs. To identify the components of this repressive apparatus, we performed an

siRNA screen using a BAC-CIITA vector containing all the remote elements crucial for CIITA

regulation. The screen yielded an unexpected novel link between the histone chaperone,

CHAF1A, and PRC2. We show that siCHAF1A rescues PRC2-repressed genes in a cell cycle

dependent manner. Interestingly, siCHAF1A does not lead to a loss but a redistribution of

H3K27me3. Therefore, we believe that CHAF1A coordinates repressive epigenetic memory by

regulating the genomic positions at which PRC2 methylates histone H3K27 during DNA

Page 3: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

iii

synthesis. Our study thus, for the first time provides important clues regarding how the

epigenetic memory of this important PRC2-mediated mark is maintained in mammalian cells.

Page 4: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

iv

Acknowledgments

I would like to sincerely thank to Dr. Rod Bremner and the department of Laboratory Medicine

and Pathobiology, at the University of Toronto for giving me the opportunity to study and learn

science. I am grateful for the financial support provided by CIHR and other sources at Dr. Rod

Bremner’s lab. I am indebted to my PhD committee, Drs. Jeff Wrana, Andrew Emili and Gerold

Schmitt-Ulms for their invaluable advice and support. I appreciate all the help from Dr. Marek

Pacal and my fellow students and co-workers at the lab of Dr. Rod Bremner. I am thankful for all

the help from Dr. Kevin Brown for analyzing the screen data and from Alexandro Datti, Thomas

Sun and Frederick Sagayaraj Vizeacoumar at S.M.A.R.T., the Mount Sinai robotic facility. Last

but not least, I am grateful for the all understanding and support I received from my family.

Page 5: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

v

Table of Contents

!"#$%#$&'

Acknowledgments.......................................................................................................................... iv!

Table of Contents ............................................................................................................................ v!

List of Figures ................................................................................................................................. x!

List of Appendices ......................................................................................................................... xi!

List of Abbreviations .................................................................................................................... xii!

Chapter 1 ......................................................................................................................................... 1!

1! General Introduction .................................................................................................................. 1!

1.1! Gene regulation ................................................................................................................... 2!

1.1.1! Gene regulation at the transcriptional level ............................................................ 2!

1.1.1.3! Chromatin and epigenetics...................................................................................... 4!

1.2! Gene regulation by PRC2 and TrxG................................................................................... 6!

1.2.1! PcG complexes........................................................................................................ 7!

1.2.2! TrxG........................................................................................................................ 9!

1.3! IFN! .................................................................................................................................. 11!

1.4! CIITA regulation ............................................................................................................... 11!

1.5! Identifying new factors in CIITA regulation ..................................................................... 12!

1.5.1! Replication Fork proteins, epigenetic memory and CIITA regulation ................. 13!

1.5.2! Histone Chaperones, epigenetics and cancer ........................................................ 15!

1.5.3! Nucleosome replication: Conservative vs. Semiconservative .............................. 15!

1.5.4! Chaperones regulate transport, modification, and deposition of new histones..... 17!

1.5.5! Chaperones recycle old histones during replication ............................................. 20!

1.5.6! Chaperones and Epigenetic Memory: The Example of CAF1 ............................. 24!

Page 6: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

vi

1.5.7! Other potential roles for CAF1 in maintaining repressive chromatin states......... 29!

1.5.8! CAF1 and Cancer.................................................................................................. 29!

1.5.9! ASF1, HIRA and cancer ....................................................................................... 30!

1.5.10! DEK, DAXX, H3.3 and Cancer............................................................................ 31!

1.5.11! Future directions ................................................................................................... 34!

1.6! Hypothesis......................................................................................................................... 35!

Chapter 2 ....................................................................................................................................... 36!

2! RNAi Screens to Identify new components regulating IFN! responsive genes (ISGs)........... 36!

2.1! Abstract ............................................................................................................................. 37!

2.2! Introduction....................................................................................................................... 38!

2.3! Material and Methods ....................................................................................................... 42!

2.3.1! Cell culture and adenovirus .................................................................................. 42!

2.3.2! HTP siRNA screening........................................................................................... 42!

2.3.3! BAC construction and reporter assays .................................................................. 43!

2.3.4! Statistical Methods for Hit Selection .................................................................... 43!

2.4! Results............................................................................................................................... 44!

2.4.1! The HTP siRNA screen optimization ................................................................... 44!

2.4.2! The primary WT and Suppressor screens ............................................................. 52!

2.4.3! The secondary WT and Suppressor screens for hits validation ............................ 55!

2.4.4! Tertiary Validation on Endogenous CIITA ........................................................... 57!

2.5! Discussion ......................................................................................................................... 58!

Chapter 3 ....................................................................................................................................... 60!

3! CHAF1A Regulates the Distribution of PRC2-mediated epigenetic mark, H3K27me3, During Replication ................................................................................................................... 60!

3.1! Abstract ............................................................................................................................. 61!

3.2! Introduction....................................................................................................................... 62!

Page 7: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

vii

3.3! Material and methods........................................................................................................ 64!

3.3.1! Cell culture and siRNA transfection ..................................................................... 64!

3.3.2! RNA extraction and reverse transcription (RT).................................................... 64!

3.3.3! Western Blotting ................................................................................................... 64!

3.3.4! Chromatin immunoprecipitation (ChIP). .............................................................. 65!

3.3.5! Quantitative Real-Time PCR (qPCR)................................................................... 65!

3.3.6! Plasmid construction............................................................................................. 66!

3.3.7! Immunostaining .................................................................................................... 66!

3.3.8! Cell cycle block..................................................................................................... 66!

3.4! Results............................................................................................................................... 67!

3.4.1! CHAF1A and PCNA repress CIITA induction ..................................................... 67!

3.4.2! CIITA rescue by CHAF1A KD is BRM independent ........................................... 71!

3.4.3! The effect of siCHAF1A on known regulators of IFN! signaling........................ 73!

3.4.4! Effects of siCHAF1A on cell survival and DNA damage .................................... 75!

3.4.5! Depleting other components linked to histone deposition, DNA replication or gene silencing does not rescue CIITA ................................................................... 78!

3.4.6! CHAF1A represses multiple PRC2-regulated ISGs ............................................. 81!

3.4.7! CHAF1A is required for PRC2-mediated chromatin modification ...................... 83!

3.4.8! General role for CHAF1A in promoting PRC2-mediated gene silencing ............ 85!

3.4.9! CHAF1A-mediated repression requires S-phase .................................................. 87!

3.4.10! SiCHAF1A causes a dramatic redistribution of H3K27me3 without changing PRC2 localization ................................................................................................. 89!

3.5! Discussion ......................................................................................................................... 92!

3.5.1! A novel direct link between CHAF1 and PRC2-mediated gene repression during replication .................................................................................................. 92!

3.5.2! CHAF1A is required for printing the H3K27me3 mark at the proper targets ...... 93!

4! Discussion and Future Directions ............................................................................................ 97!

Page 8: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

viii

4.1! Discussion ......................................................................................................................... 98!

4.1.1! CHAF1A maintains H3K27me3 during replication ............................................. 98!

4.1.2! CHAF1A may guide the H3K27me3 deposition indirectly.................................. 99!

4.1.3! CHAF1A may activate PRC2 ............................................................................. 101!

4.2! Future directions ............................................................................................................. 102!

4.2.1! The global effect of depleting CHAF1A on PRC2 regulated genes and redistribution of H3K27me3 ............................................................................... 102!

4.2.2! DNA methylation and histone modification ....................................................... 102!

4.2.3! Identifying the components involved in the CHAF1A-dependent maintenance of H3K27me3...................................................................................................... 103!

4.2.4! Structure-Function analyses of CHAF1A........................................................... 104!

4.2.5! Using aniPOND to screen for additional factors co-operating with CHAF1A at the replication forks ............................................................................................ 105!

4.3! Concluding remarks ........................................................................................................ 105!

References................................................................................................................................... 107!

Appendices.................................................................................................................................. 131!

Page 9: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

ix

Page 10: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

x

List of Figures

Fig 1.1 Histone chaperone functions during DNA replication…………………………………23

Fig 1.2 Multiple roles for CAF1 at heterochromatin…………………………………………...28

Fig 2.4.1 Optimizing siRNA transfection and viral transduction conditions…………………..47

Fig 2.4.2 Optimizing the timing and order of the screen steps………………………………....48

Fig 2.4.3 Automated HTP version of the optimized protocol…………………………………..49

Fig 2.4.4 Testing the “B score” method of hit analysis in the optimized protocol …………….50

Fig 2.4.5 A. Plot of B scores from the pilot Wt screen………………………………………….51

Fig 2.4.6 Schematic diagram of the primary siRNA screen …………………………………...53

Fig 2.4.7 Schematic diagram of secondary validation screen…………………………………..56

Fig 3.4.1 CHAF1A and PCNA repress endogenous CIITA induction………………………….69

Fig 3.4.2 CIITA rescue by siCHAF1A is BRM-independent……………………………………72

Fig 3.4.3 The effect of siCHAF1A on the level of IRF1, p-STAT1 or STAT1 proteins………..74

Fig 3.4.4 Effects of siCHAF1A on cell growth, apoptosis and DNA damage. …………………76

Fig 3.4.5 The role of other known CHAF1A interacting proteins in CIITA silencing…………..80

Fig 3.4.6 The effect of CHAF1A KD on PRC2/BRG1dependent ISGs…………………………82

Fig 3.4.7 The effect of CHAF1A KD on H3K27me3 at PRC2/BRG1 regulated ISGs………….84

Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene silencing ……………………….86

Fig 3.4.9 The role of CHAF1A in gene silencing is cell cycle dependent………………………88

Fig 3.4.10 CHAF1A KD alters H3K27me3 distribution………………………………………...90

Page 11: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

xi

List of Appendices

Apx1. List of primers for RT-PCR……………………………………………………………..128

Apx 2. List of SiRNAs………………………………………………………………………….130

Apx 3. List of Abs………………………………………………………………………………131

Apx 4. List of activator and inhibitor hits form primary and secondary screens…………….....132

Page 12: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

xii

List of Abbreviations

Ago: argonaut

aniPOND: accelerated native iPOND

BAF: BRG associated factor

BAC: bacterial artificial chromosome

BRG1: Brahma related gene1

BRM: Brahma

Bromodomain: binds acetylated histones

CHD: chromodomain and helicase-like domain

CIITA: MHC ClassII transactivator

ChIP: chromatin immunoprecipitation

ChIP-chip: immunoprecipitation coupled with DNA tiling array

Chromodomain: binds methylated histones

CpG: cytosine Guanine nucleotide

CBX: chromobox homolog

CHAF1A: chromatin assembly factor1

COSMIC: somatic mutation in cancer

CSE1l: CSE1 chromosome segregation 1-like

DNMT: DNA (cytosine-5-)-methyltransferase

ETM: epithelial-mesenchymal transition

EZH: zeste homolog 2

EED: embryonic ectoderm development

EHMT2: euchromatic histone-lysine N-methyltransferase 2

FXYD1: FXYD domain containing ion transport regulator 1

GBP1: guanylate-binding protein1

H3K4ac: acetylated lysine4 at histone3

Page 13: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

xiii

H3K9me3: tri-methylated lysine 9 at histone H3

H3K27me3: tri-methylated lysine 27 at histone 27

HIRA: HIR histone cell cycle regulation defective homolog A

HDAC: histone deacetylase

HP1: heterochromatin protein1

HOX: homeobox

HOTAIR: HOX transcript antisense RNA

HTTP: high throughput

IFI2: interferon, gamma-inducible protein

IFITM3: interferon-induced transmembrane protein 3

IFN: interferon

IRF1: interferon regulatory factor1

IP: immunoprecipitation

iPOND: isolation of proteins on nascent DNA

ISWI: imitation SWI

IFNGR1: interferon gamma receptor 1

JAK: Janus family protein tyrosine kinase

JARID: Jumonji AT-rich interactive domain

K: lysine

LSD1: lysine specific demethylase 1

Luc: luciferase

LincRNA: intergenic non-coding RNA

MLL: Mixed lineage leukemia

MAPK13: mitogen-activated protein kinase 13

MHC: major histocompatibility complex

MBD: methyl-CpG binding domain protein 1

MeCP2: methyl CpG binding protein 2

MMS: methyl methane-sulfonate

Page 14: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

xiv

ncRNA: non-coding RNA

PIV: promoter IV of CIITA

PLRG1: pleiotropic regulator 1

PITX2: paired-like homeodomain transcription factor2

PcG: Polycomb protein group

PCNA: proliferative cell nuclear antigen

POLE: polymerase (DNA directed), epsilon

POLD: polymerase (DNA-directed), delta

PRC: Polycomb Repressive Complex

PRPF8: PRP8 pre-mRNA processing factor 8 homolog

qPCR: real-time quantitative PCR

RT: reverse transcription

R: arginine

RING: ring finger protein

ROR1: receptor tyrosine kinase-like orphan receptor 1

RBBP: retinoblastoma associated protein

RISC: RNA-induced silencing

SD: standard deviation

SET: SuVar39, Enhancer of Zeste, and Trithorax

SLIDE: SANT-Like ISWI domain

SOCS: suppressor of cytokine signaling

SNAIL: snail zinc finger

STAT1: signal transducer and activator of transcription

SUV39H: suppressor of variegation 3-9 homolog

SUZ12: suppressor of zeste

SWI/SNF: mating type switching and sucrose non-fermenting

Page 15: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

xv

SUMO: sumoylation

TET: ten-eleven translocations

TrxG: trithorax group

Ub1: mono-ubiquitylation

Wt: wild type

XCI: X chromosome inactivation

Page 16: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

1

Chapter 1

1 General Introduction

Page 17: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

2

1.1 Gene regulation

Gene regulation in eukaryotes is a complex and sophisticated process of fundamental importance

for all biological events. The expression of genes in cells can be regulated on several levels in

response to internal and external stimuli. First, the DNA regulatory elements and promoters and

proteins that bind to these elements can control the rate of transcription. Second, mRNA splicing

and stability affect the amount of mRNA available. Finally, if relevant, the dynamics of mRNA

translation and post-translational protein modifications will crucially influence the final amount

of gene product generated [1, 2]. Here we focus on transcriptional level of gene regulation.

1.1.1 Gene regulation at the transcriptional level

The control of gene expression at the transcriptional level is the result of three critical

components. (1) DNA binding of transcriptional factors (TFs) that either increase (activators) or

decrease (repressors) the interaction and stabilization of RNA polymerase at the promoter of the

genes. (2) The presence of co-regulators which collaborate with TFs and can induce (co-

activators) or reduce (co-repressors) the transcriptional rate of genes. (3) The activity of factors

which affect chromatin structure by altering the accessibility of the DNA to the transcriptional

machinery. These latter factors can be histone modifying factors or chromatin remodeling

complexes (see 1.2.2.1 for a detailed discussion) [3-8].

1.1.1.1 Transcription Factors

TFs are proteins that bind to regulatory DNA sequences and either enhance or inhibit

transcription [9]. TFs are a large family of proteins that can be classified based on their: 1)

mechanism of action, 2) spatial and temporal regulatory function and 3) DNA binding domain

structure.

There are two major classes of TFs based on their mechanism of action. The first class

includes the so called general TFs such as TFIIA, TFIIB, TFIID, TFIIE, TFIIF and TFIIH that

are required for the basal level of transcription. General TFs form pre-initiation complexes at

gene promoters which are essential for the assembly of RNA polymerase II machinery and

initiation of transcription [10-14]. The second class includes TFs (e.g., c-MYC, OCT-1, SP-1)

that bind upstream of the initiation sites to induce or repress transcription.

Page 18: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

3

TFs can also be classified based on the spatial and temporal regulatory function. For

example, some of the transcription factors are constitutively expressed in all cell types (e.g.,

general TFs). However a large family of transcription factors is activated in specific cell types at

different developmental stages or in response to internal or external stimuli. During embryonic

development, the expression of cell specific transcriptional factors is tightly controlled to switch

on or off cell type specific genes. For example, homeobox (Hox) genes control the body plan of

the embryo and determine the type of segment structures (e.g. legs, antennae, and wings in fruit

flies or the different vertebrate ribs in humans) that will form on a given segment [15]. TFs also

respond to intercellular (e.g., TGF", interferons) and environmental (e.g., heat shock, hypoxia)

signals. For example, in the TGF" signaling pathway, the binding of ligand to a receptor leads to

phosphorylation of receptor-regulated SMADs (R-SMADs) which then bind the coSMAD

SMAD4. R-SMAD/coSMAD complexes translocate to the nucleus where they act as TFs

regulating target gene expression [16-18].

Finally, based on DNA binding domains, TFs can be classified to 5 major super classes:

1) Basic domain, 2) Zinc-coordinating DNA binding domains, 3) Helix-turn-helix, 4) beta-

scaffold factors with minor groove 5) Others. Each super class contains different sub classes and

families. Not surprisingly, altogether this is a large number of proteins. Indeed, TFs make up at

least 10% of genes in the genome, which makes this family the single largest family of human

proteins [19].

1.1.1.2 Transcriptional co-regulators

The regulatory functions of TFs as activators or repressors are achieved in collaboration with co-

regulators (co-activators or co repressors). Co-regulators do not typically have DNA binding

domains and are recruited to the DNA sequences by TFs. There are different classes of co-

regulators. For example repression of E-cadherin, which is an epithelial marker and is observed

in epithelial-mesenchymal transition (EMT), is achieved by collaboration of TFs activated by

different signaling pathways such as TGF", Notch, Wnt, as well as hypoxia and co-regulators

including Snail, Slug, Twist, ZEB1 and so on [20-22]. Co-regulators can be histone modifying

enzymes such as histone acetyltransferases (HATs) that act as co-activators or histone

deacetylases and histone methylases of lysine K9 and K27 which are co-repressors [13, 23-29].

Page 19: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

4

A special class of co-regulators includes ATP-dependent chromatin remodeling

complexes such as SWI/SNF that can be either co-activators by making the chromatin more

accessible to transcription machinery or repressors by making the chromatin more condensed and

not accessible for transcription [30-35]. These will be discussed in detail in the section 1.2.2.1.

1.1.1.3 Chromatin and epigenetics

DNA is packed into a compact structure called chromatin so that it can fit into the cell nucleus

[36]. Chromatin is a dynamic and highly organized system made of repeating units referred to as

nucleosomes [36-40]. In eukaryotic cells about 147 base pairs (bps) of DNA are wrapped around

a histone octamer (two of H3/H4 and two of H2A/H2B) to form a nucleosome [39-41]. The H1

histone, which is called the linker histone, binds the DNA segments between nucleosomes and

helps wrap nuleosomes into a higher conformation [39, 40]. Chromatin is classified into two

forms, based on its structure. The first, euchromatin, is less condensed, gene-rich and replicates

early. The second, heterochromatin, is highly condensed, mostly not transcribed and replicates

late in the S phase of the cell cycle [39, 40, 42-44].

Chromatin structure functions as a barrier for transcription and needs to be rendered

accessible for transcription factors and transcriptional machinery. This can be done, in

eukaryotic cells in two ways. The first, carried out by ATPase chromatin remodeling complexes

which alter nucleosome structure using energy derived from ATP hydrolysis [45-47]. The

second, performed by histone modifying enzymes, relaxes histone-histone and histone-DNA

interactions through covalent histone modifications.

There are different types of histone modifications such as acetylation, phosphorylation,

methylation, ubiquitination and sumoylation that are context-dependent and can either repress or

activate gene transcription. Two important and well known histone modifications associated

with gene repression are the methylated lysine 9 and 27 of histone H3 (H3K9 and H3K27). The

former is mediated by several different histone methyltransferases (see below) and the latter is

mediated by Polycomb Group (PcG) protein complexes [48].

Page 20: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

5

In addition to histone modifications, DNA can also be methylated which usually

correlates with gene repression. The DNA and histone modifications together are referred to as

“epigenetic code” (epi = over, beyond) which defines the status of gene expression as either

silent or active without changes in DNA sequence [49]. A strict definition of epigenetics only

includes chromatin events that are inherited from one cell or organism generation to the next.

However, the term is also used more broadly to refer to chromatin modifications in any cell, even

post-mitotic cells.

1.1.1.3.1 DNA methylation

DNA methylation is an epigenetic modification carried out by DNA methyltransferases that

covalently add methyl groups to the cytosine nucleotides of DNA. It occurs in those parts of

DNA sequences where a cytosine nucleotide sits next to a guanine nucleotide; such sequence is

referred to as CpG [50]. Unmethylated CpG clusters are called CpG islands, which are usually

associated with gene promoters in normal cells [51-53]. Abnormal hypermethylation of CpG

islands is often seen at the promoters of tumor suppressors, leading to their inactivation, in

different types of cancers such as colon cancer, glioma and leukemia [54, 55]. DNA methylation

attracts methyl-CpG-binding proteins such as MBD1 and MeCP2, which in turn promote binding

of histone modifying enzymes such as histone deacetylases [56, 57]. The combination of DNA

and histone modification leads to chromatin compaction and gene repression [58].

DNA methylation/demethylation is mediated by different classes of DNA methyl

enzymes. One class of DNA methyltransferases is the DNMT, DNA (cytosine-5-)-

methyltransferase family. One member of DNMT family is DNMT1 which maintains the DNA

methylation pattern during replication [59]. DNMT1 recognizes methylated CpGs at parental

DNA strands and transfers methyl groups to the correct CpG sites on the daughter DNA [60].

DNMT1 also interacts with Proliferative Cell Nuclear Antigen (PCNA) at the replication forks,

which may be important in the maintenance of DNA methylation during replication [58, 61].

Other DNMT family members such as DNMT3a and DNMT3b are involved in de novo

methylation of DNA during development [59]. Recent studies indicate that Ten-Eleven

Translocation (TETs) DNA methylase enzymes, which oxidize 5-methylcytosine (5mC) to 5-

hydroxymethylcytosine (5hmC), serve to demethylate DNA [62]. DNA

methylation/demethylation is a dynamic event and has crucial impact on gene regulation. The

Page 21: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

6

cross talk between DNA and histone epigenetic modifications leads to gene activation or

repression.

1.1.1.3.2 Histone methylation

Both histones H3 and H4 can be methylated and these marks can be either activating or

repressive. Methylation of H3 is well studied, and both lysine (K) and arginine (R) residues can

be methylated to a different degree: mono (me1), di (me2) or tri-methylation (me3). The

methylation of K 4 or 36 of histone H3 is linked to gene activation while the methylation of K 9,

27, or 79 of this histone lead to gene repression [48, 63]. SET1 and Mixed lineage leukemia

(MLL) family of histone methyl transferases are involved in H3K4 methylation [64]. Histone

methylation is reversible and can be erased by specific histone demethylases. For example

Lysine-specific histone demethylase1 (LSD1) and Jumonji AT-rich interactive domain 1

(JARID1) are responsible for erasing methyl groups from histone H3K4 [65].

The trimethylation of K 9 of histone H3 (H3K9me3) and of H3 lysine 27 (H3K27me3)

are two major and well-studied repressive marks that have been conserved throughout eukaryotic

evolution [66]. These latter histone modifications are mediated by several different types of

histone methyltrasferases such as Suv39 family containing SET domain-containing enzymes [48,

67, 68]. The methylation of H3K9 provides docking sites for proteins such as heterochromatin

protein 1(HP1) which induces chromatin condensation and maintains gene silencing [69-73].

The H3K27me3 modification is mediated by PcG proteins. Components of PcG complex

contain chromodomains which bind H3K27me3 and mediate gene silencing. We will discuss

these in more detail below.

1.2 Gene regulation by PRC2 and TrxG

The DNA sequences and genetic information are shared by all cells but what distinguishes their

identity and fate is their different gene expression patterns. These patterns (i.e., active or silent)

are established during development. Next, the memory of the transcriptional state needs to be

maintained during cell division and DNA replication [74]. Trithorax group (TrxG) and PcG

complexes are known to maintain the memory of the gene status as active and repressed,

respectively [75, 76].

Page 22: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

7

PcG proteins were identified more than 50 years ago in Drosophila as negative HOX

genes regulators, revealed by mutations of HOX gene activators [74, 77]. Later, TrxG proteins

were identified via mutations that rescued the phenotype observed in PcG protein mutations [75,

76]. Therefore, PcG and TrxG show antagonistic effects on gene regulation [78, 79]. The

function of PcG and TrxG are evolutionarily conserved. In vertebrates, TrxG as PcG are also

crucial gene regulators but have been also implicated in other biological events such as cell fate,

differentiation and proliferation, X-chromosome inactivation, stem cell identity and cancer [79,

80]. It has been shown that the PcG and TrxG complexes are bound to the same DNA sequences

which allows for switchable regulatory platforms [76].

1.2.1 PcG complexes

PcGs are essential in maintaining the expression status of genes imprinted during early

development and their dysfunction causes homeotic transformations[81]. There are five different

PcG complexes of which PRC1 and PRC2 are the best characterized [23].

PRC1 contains catalytic subunits RING1A and RING1B, which catalyze lysine 119

mono-ubiquitylation (K119) of histone H2A (H2AUb1). The PRC1 core complex contains the

chromodomain-containing protein Chromo Box Homolog 4 (CBX4), which aids SUMO E3

ligase to sumoylate many other proteins including DNA methyltransferases, such as DNMT3a

[82]. However the function of sumoylation in PcG-mediated gene silencing is not completely

clear.

The catalytic subunit of PRC2 that mediates di-methylation and tri-methylation of

H3K27, is formed by Enhancer of Zeste homolog 2 (EZH1/2), which binds to Suppressor of

zeste 12 (SUZ12) and one of Embryonic ectoderm development isoforms (EED 1-4) for full

activity [83]. The core of the PRC2 complex also contains cofactors such as Retinoblastoma-

associated protein, RbAp46/48 (also known as RBBP7/4), and JARIDs. Finally, histone

deacetylases, HDAC1 and HDAC2, transiently bind the core complex and are required for its

recruitment to target loci [83-87].

EZH2 mediates di-methylation and tri-methylation of H3K27, a modification recognized

by the chromodomain subunit of PRC1 complex, which then leads to chromatin structure

alteration and transcriptional repression [88, 89]. It has been shown that RNA polymerase II

Page 23: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

8

(RNApol II) pauses at some PRC2-rich regions and it is believed that short transcripts that are

bound to such paused RNApol II also recruit PRC2 [90, 91]. Recent studies have shown that the

function of PRC2 is not limited to gene regulation and it is also involved in other biological

process of cells such as cell cycle control and DNA damage [92-94].

PRC2 regulates thousands of genes including many cell type dependent transcriptional

factors [78, 95]. PRC2 components such as EZH2 are upregulated and act as oncogenes in

several types of cancers [96, 97]. For example, EZH2 has been used as a biomarker for breast

and prostate metastasis. Also, over expression of PRC2 induces silencing at the promoter of

tumor suppressor genes [97, 98]. Recent studies have identified mutation of EZH2 in SET-

domain of the protein which is associated with gain of function and induction in H3K27me3

repressive mark [99, 100]. Therefore, it is crucial, for the sake of improved understanding and

treatments of cancer, to decipher how PRC2 is regulated and distributed in cancer cells.

1.2.1.1 Mechanism of PRC2 recruitment

PRC2 is recruited to the specific DNA sequences, Polycomb Responsive Elements (PREs), in

Drosophila but only recently DNA elements have been described that recruit PRC2 to their

targets in vertebrates[101]. Importantly, PRC2 recruitment to target genes can be assisted by a

variety of other factors [86, 87]. For example, recently, non-coding RNAs (ncRNAs) have been

described as important PRC2 recruiters in vertebrates [85]. The involvement of ncRNAs was

first reported in the context of HOXD cluster repression. Here, the recruitment of PRC2 to its

regulatory elements is mediated by long ncRNA (lincRNA) HOTAIR which is expressed from

the HOXC cluster [102]. Moreover, ncRNAs are also involved in the X chromosome

inactivation (XCI) by recruiting EZH2 to initiate and spread silencing [102]. Recent studies

have been showing more evidence that lincRNAs are associated with PRC2 at the targeted genes

and knocking down these lincRNAs activates PRC2-repressed genes [103, 104].

In addition to ncRNAs and lincRNAs, there are several protein factors which are

involved in PRC2 recruitment to the target genes. These proteins include, the RNAi machinery

factors such as Argonauts, the transcription factor Snail family zinc finger 1 (SNAIL1) or the

Jumonji protein JARID2 [13, 105].

Page 24: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

9

Recent studies show that PRC2 is recruited to hypomethylated CpG rich regions [106-

108]. However the mechanism of the recruitment and to which extent DNA methylation repels

PRC2 complex recruitment still needs to be addressed.

1.2.1.2 PRC2-dependent epigenetic inheritance

Another important question concerning PRC2 activity is how PRC2-mediated signatures are

inherited during DNA replication when the histone structure and epigenetic marks have been

challenged. During S-phase, nucleosomes are disrupted ahead of the replication fork and then

reassembled to the newly synthesized DNA. Therefore a specific mechanism must exist to

preserve the parental PRC2-mediated silencing marks.

To date, two models have been proposed to explain the inheritance of the PRC2-mediated

epigenetic mark, H3K27me3. The first one is a “positive feedback” model in which the

preexisting H3K27me3 marks stimulate PRC2, thus methylating newly deposited histones [109,

110].

Recently, a Drosophila study has proposed another model. This model posits that

H3K27me3 inheritance does not rely on the actual mark but on the presence of the PRC2

complex [111]. Here, PRC2 remains bound to DNA during replication and the pre-existing

marks are in fact stripped off histones during replication. PRC2 components modify both

parental and daughter histones at the same place again in the G2 phase after DNA replication

[111].

1.2.2 TrxG

TrxG proteins are categorized into three classes based on their function. In vertebrates, the first

class includes the SET domain containing complexes that methylate Lys 4 of histone H3

(H3K4me3), a gene activating mark [79]. This class also includes Myeloid/Lymphoid Or

Mixed-Lineage Leukemia, MLL histone methyltransferases which mediate global gene

activation [76, 112].

In the second class of TrxG proteins are the sequence-specific DNA binding proteins that

contain some histone modifying enzymes and chromatin remodeling complexes. DNA binding

proteins localize to TrxG responsive elements (TREs), found in Drosophila, which often overlap

Page 25: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

10

with PREs [76, 78]. It has been suggested that CpG-rich sequences may perform this role and

recruit PcGs and TrxGs to DNA [106].

The third class contains the ATP-dependent chromatin remodeling complexes which

activate genes by inducing nucleosome eviction or nucleosome sliding or changing the chromatin

architecture (e.g., creating loops) all of which renders DNA target motifs more or less accessible

for transcription activators or repressors and RNA polymerase machinery [113]. There are

several types of ATP-dependent chromatin remodeling complexes and they regulate a large

number of genes in different cell types but they are also involved in other cellular process such

as cell cycle or cell signaling [33, 114, 115]. We will discuss these remodeling complexes in

more detail below.

1.2.2.1 ATP-dependent chromatin remodeling complexes

Cells use the ATP-dependent chromatin remolding complexes to adjust the DNA surrounding

histones to make it more accessible to transcriptional factors and RNA polymerase or providing

higher level of chromatin structure by looping which are associated with gene activation or

repression respectively. They are protein complexes conserved from yeast to mammals and the

complexes can be categorized into several subfamilies based on the presence of conserved

domains [46, 116].

One subfamily of ATPase chromatin remodelers includes the imitation switch (ISWI)

complexes, which contain different histone binding domains such as SANT and SLIDE (SANT-

Like ISWI) domains [117]. Next, the chromodomain helicase DNA-binding (CHD) subfamily

binds to acetylated histones [118]. The SWI/SNF chromatin remodeling complex is another

important subclass which was first identified in yeast in a screen aimed to identify genes that

regulate mating-type switching (SWI) and sucrose non-fermenting (SNF) phenotypes [119-121].

In mammals, there are two major SWI/SNF complexes: BAF (hSWI/SNFA) or PBAF

(hSWI/SNFB). These two complexes have 10 subunits in common, however, the catalytic

subunit of PBAF is Brahma-Related gene1 (BRG1) whereas BAF can associate with either

BRG1 or Brahma (BRM) [122]. Human BRG1 and BRM show approximately 74% identity on

the amino-acid level and although they both act as engines of the SWI/SNF complexes, they

seem to play different roles during differentiation and proliferation and cell signaling [123-125].

Page 26: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

11

BRG1 is essential for tumor suppression and plays important roles in differentiation, gene

regulation and cell cycle control [126]. For example, BRG1 is mutated, deleted or silenced in

several types of cancers such as non-small cell lung carcinoma, prostate cancer, breast cancer

and adrenal carcinoma [127]. BRG1 heterozygous mice are prone to transformation [123].

Importantly, our lab showed that BRG1 is required for the induction of a subset of Interferon !

(IFN!)- responsive genes [33, 115, 128]. Another pathway regulated by both BRG1 and BRM is

the Notch signaling pathway. Here, their activity is antagonized by PcG complexes [125, 129].

This is very interesting because, as described in Chapter 3, we observed the co-operation of

BRG1 and PRC2 in the regulation of genes downstream of the IFN! signaling pathway.

1.3 IFN!

Interferons are soluble proteins, which were initially described as factors secreted in response to

viral infection, designated to inhibit cell proliferation and viral replication [130]. There are two

types of IFNs, IFN #/" (type I) and IFN! (type II). Both types have antiviral and

antiproliferation activities but bind to different cell surface receptors and have different

biological effects [131, 132].

IFN! signaling is initiated by binding of the ligand to IFN! receptors (IFNGR1-

IFNGR2), which results in their oligomerization. This in turns leads to the phosphorylation of

receptor-associated Janus kinases, JAK1 and JAK2 [131, 132]. These kinases phosphorylate

Signal transducer and activator of transcription-1 (STAT1) [131, 133, 134]. Dimerized p-

STAT1 translocates to the nucleus and binds to regulatory elements of IFN! stimulated genes

(ISGs) [131, 133, 134]. ISGs respond to IFN! in most cell types [135, 136]. Unfortunately, very

little is known about the regulation of ISGs in cancer. As discussed below and in more detail in

Chapters 2 and 3, using the ISG Class II transactivator (CIITA), the master regulator of MHC

class II, as a model, our lab has uncovered several novel factors in ISG regulation in cancer cells

[137].

1.4 CIITA regulation

CIITA is the master regulator and a co-activator of MHC class II (MHC) induction [138].

CIITA is regulated by four alternative promotes (I-IV). Promoters (I-III) are used for the

constitutive form of CIITA induction which is observed in antigen presenting cells such as B

Page 27: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

12

lymphocytes and dendritic cells [138]. Promoter IV is used in the inducible form of CIITA in

other cell types and drives the IFN! response [139].

Immune cells target not only infected cells but can also detect transformed cells and

destroy them [131]. This process is called immunosurveillance [131]. CIITA is silent and not

responsive to IFN!, in tumors such as Uveal melanoma, adrenal carcinoma and colorectal

cancers [140, 141]. It is likely that silencing of CIITA is a strategy that cancer cells use to switch

off MHCII and escape immunosurveillance. Therefore, understanding the mechanism of CIITA

regulation should help improve drug design to treat cancers where MHCII is inactivated.

Our lab has used CIITA as a model to understand the regulation of ISGs but also to study

gene regulation in general and has made significant contributions in this field. Our previous

work linked SWI/SNF to regulation of ISGs in cancer cells and found that BRG1 is required for

CIITA to respond to INF! [33, 115, 137]. Our lab identified multiple STAT1/IRF1 enhancers

across the CIITA locus, spanning >100 kb far from transcription start site, which are critical for

CIITA induction in response to IFN! [137]. In addition, we showed that in the BRG1-deficient

adrenal carcinoma SW13 cells CIITA and a subset of ISGs are regulated by PRC2 repressive

complexes. We detected the PRC2-signature chromatin mark, H3K27me3 as well as the PRC2

components SUZ12 and EZH2 at multiple sites across the CIITA locus (unpublished data,

Chapter 3). We showed that re-introducing BRG1 to SW13 cells restored the IFN!

responsiveness of CIITA [33, 115]. Moreover, we showed that knocking down PRC2

components (EZH2 and SUZ12) rescued CIITA induction and reduced H3K27me3 across the

CIITA locus (unpublished data, Chapter 3).

Together, these findings raise the possibility that inactivation of SWI/SNF and/or

activation of PRC2-mediated repression might be mechanisms cancer cells use to silence MHCII

and escape immunosurveillance.

1.5 Identifying new factors in CIITA regulation

Using CIITA as a model of ISG regulation has several advantages. First, CIITA is a very

important ISG and a master regulator of MHC class II, and thus the knowledge of CIITA

regulation has broad clinical relevance. Further, as we have found, this gene is regulated by both

PRC2 and BRG1, members of PcG and TrxG family of proteins, respectively. Since multiple

Page 28: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

13

genes are regulated by these two factors, uncovering how they work in the context of CIITA

could likely be extended to many other PcG /TrxG regulated genes.

Z. Ni in our lab built a 194 kb BAC-CIITA luciferase (Luc) reporter containing all the

newly identified remote enhancers, and showed that the construct is, like the endogenous CIITA

locus, also BRG1-dependent. As described in Chapter 2, we took advantage of this reagent and

performed high throughput (HTP) siRNA screens to determine: A. whether we could rescue

CIITA responsiveness in BRG1-deficient cells by suppressing factors that repress this target

(Suppressor screen) and B. which additional regulators (activators or inhibitors) affect CIITA

regulation in the presence of BRG1 (Wild type screen). In Chapter 2, the optimization

procedures for the different steps of the siRNA screens are discussed and the identified hits are

reported. In Chapter 3 we focus on two hits identified by the Suppressor screen, both replication

fork proteins, and dissect the mechanism of these factors in the CIITA regulation.

1.5.1 Replication Fork proteins, epigenetic memory and CIITA regulation

As we will describe later, our screen for repressors of CIITA in BRG1-deficient cells uncovered

two candidates, CHAF1A (P150), the large subunit of CAF-1 histone chaperone complex and

PCNA, Proliferating Cell Nuclear Antigen, which is a cofactor of DNA polymerase delta. Both

identified hits are required for DNA replication and are also involved in replication-coupled gene

silencing.

The main function of CAF-1 is to assemble the histones H3/H4 into DNA at the

replication fork and the complex includes CHAF1A, CHAF1B and RbAp46/48 (RBBP4) [142].

Interestingly, RbAp46/48 is a component of several repressor complexes including PRC2 [84].

Recent studies have shown that CHAF1A is required for maintaining H3K9me3 epigenetic

modifications in heterochromatin regions after DNA replication (epigenetic memory) and was

identified in an siRNA screen aimed to identify factors required for silencing of a randomly

integrated vector in HeLa cells [143-145]. PCNA is a DNA polymerase accessory protein which

interacts with CHAF1A at the replication fork. There is growing evidence implicating both

PCNA and CHAF1A in gene silencing [146, 147]. We found that knock down of both of these

Page 29: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

14

factors enhanced responsiveness of CIITA and a subset of other PRC2-regulated ISGs as well as

the basal levels of non-ISGs in SW13 cells. We have now evidence indicating that CHAF1A

controls CIITA expression by proper transferring of PRC2- dependent repression mark,

H3K27me3, during S-phase which is discussed in Chapter3.

In the next section, published as a chapter in “SYSTEMS ANALYSIS OF CHROMATIN-

RELATED PROTEIN COMPLEXES IN CANCER”, we review in detail the current knowledge of

histone chaperones and epigenetic memory [148]. This section was written together with my

supervisor, Rod Bremner.

Page 30: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

15

1.5.2 Histone Chaperones, epigenetics and cancer

The first histone chaperone was isolated as a nuclear protein that prevents histone precipitation

and facilitates nucleosome formation [149]. Chaperones are typically acidic, ideal for binding to

basic, positively charged histones [150]. They bind histones in the cytoplasm, carry them into

the nucleus and facilitate nucleosome formation (Fig 1.1) [151, 152]. Chaperones were

originally thought of as simple carriers that prevent nonspecific DNA-histone interactions, but

are now known to regulate histone disassembly and reassembly, covalent histone and DNA

modifications, and the maintenance of epigenetic and chromatin states during replication,

transcription and DNA repair.

Classically, epigenetics is “the inheritance of variation (-genetics) beyond (epi-) changes

in the DNA sequence” [153], but the term is now used commonly to describe any modification

of a nucleosome, even in post-mitotic cells where “inheritance” is by definition a misnomer.

When DNA is replicated or repaired, cells “remember” which genes were silent or active and

nucleosomal positioning and covalent histone modifications are crucial for this process.

Chaperones redistribute parental histones and deposit new histones on replicated DNA and thus

they also have a profound role in epigenetic memory. Understanding this process is important

because epigenetic memory is perturbed in multiple human diseases, including cancer as we

review here for histone chaperones.

1.5.3 Nucleosome replication: Conservative vs. Semiconservative

Nucleosomes consist of a core (H3-H4)2 tetramer and two H2A-H2B dimers attached on

either side, with 146 bp of DNA wrapped around the entire octamer. Histones as well as DNA

are replicated, and must be incorporated into new nucleosomes and covalently modified to

maintain the chromatin state. To understand how histone chaperones operate it is important to

first understand nucleosome replication. For simplicity, we will limit our discussion on

replication of the (H3-H4)2 tetramer to the three non-centromeric mammalian somatic H3

Page 31: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

16

variants, H3.1, H3.2 and H3.3 that combine with an invariant H4 [154]. H3.1 and 3.2

show similar properties and we will refer predominantly to the more highly expressed H3.1

isoform.

Nucleosomes could, in theory, follow conservative or semi-conservative modes of

replication. In the former model, old (parental) histones are transferred intact to one of the two

replicated DNA strands, and new histones are used to generate a second nucleosome at the

matching position. In semi-conservative replication, the parental histone octamers are split, and

mix with new histones to create two hybrid nucleosomes. An elegant study suggests that in

HeLa cells H2A-H2B dimers are always replicated semi-conservatively (new mixed with old),

but H3.1-H4 tetramers undergo conservative replication [155]. Inducible Flag-tagged Histone

H3.1 plus growth in normal lysine (K0) media was used to label old nucleosomes, then Flag-

H3.1 expression was extinguished, cells were arrested at G2/M, released and grown in heavy

lysine isotope (K8) to label new histones through the next S-phase. Mononucleosomes with old

Flag-tagged H3.1 were purified, and Mass Spectrometry (MS) used to analyze gel purified

histones. Subtracting background label, only 2% of H3.1 or H4 in Flag-tagged (i.e. old)

nucleosomes contained K8 (i.e. new) label, and this did not increase much even after a second S-

phases. Thus, there is almost no splitting of (H3.1-H4)2 during S phase, indicating conservative

replication. In contrast, H2A and H2B in Flag-tagged nucleosomes were ~ 50% labeled with K8

after one S-phase, indicating semi-conservative replication. The latter is consistent with the fact

that H2A-H2B dimers are not in contact in the nucleosome, but reside either side of the tightly

bound pair of H3-H4 dimers [156].

The replication/deposition of (H3.3-H4)2 tetramers is different from that of (H3.1-H4)2

tetramers. Human H3.1 or H3.2 differ at only 5 or 4 amino acid positions from H3.3,

respectively, but whereas their expression is restricted to S-phase, H3.3 is expressed throughout

the cell cycle [157]. Also, while Asf1 (anti silencing function 1) and CAF1 are the chaperones

that deposit new H3.1-H4 dimers, HIRA (histone regulator A), DAXX (death associated protein

six) or DEK [158] chaperones deposit new H3.3-H4 dimers [159-163]. Consistent with separate

deposition pathways, H3.1/3.2-H4 dimers are not found with H3.3-H4 dimers in nucleosomes

[160]. Using the labeling system discussed above, Xu et al found that ~6% and ~20% of

nucleosomes with old Flag-tagged/K0 H3.3 contained new K8-labeled H3.3 after one or two S-

Page 32: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

17

phases, respectively [155]. This (H3.3-H4)2 tetramer splitting occurs during DNA synthesis but

not during replication-independent deposition, as it is greatly reduced when S-phase is blocked.

In summary, H3.1-H4 tetramers do not split but undergo conservative replication, some

H3.3-H4 tetramers split and undergo semi-conservative replication, and new H3.3-H4 complexes

added outside S-phase are deposited as tetramers and do not mix with H3.1-H4 dimers.

1.5.4 Chaperones regulate transport, modification, and deposition of new histones

Newly generated histones must avoid aggregation, move into the nucleus, accumulate

post-translational modifications, and form nucleosomes on DNA. Chaperones play crucial roles

in all these processes. A recent study combined protein purification, biochemical reconstitution

assays, and RNAi/genetics to deduce a comprehensive view of the early stages of histone

synthesis and transport into the mammalian or yeast nucleus (Fig 1.1) [164]. Here, we highlight

the mammalian process.

As H3.1 emerges from the ribosome its folding is assisted by HSC70 [164]. H3.1 is

monomethylated at this early stage on lysine 9 (K9me1), although the mechanism is unclear

[165]. The newly synthesized non-replicative histone H3.3 is typically dimethylated (K9me2) or

exhibits acetylation (K9ac and K14ac) [165].

From HSC70, H3 is passed to HSP90 which cooperates with the chaperone tNASP1

(“testicular Nuclear Autoantigenic Sperm Protein) to assemble H3.1-H4 dimers (Fig 1.1) [164].

Through splicing the NASP gene also encodes “somatic” NASP (sNASP) [166]. H3-H4 dimers

in the tNASP-HSP90 complex are passed onto sNASP, which also recruits the HAT1 (histone

acetyl transferase 1) holoenzyme, made up of HAT1 and the chaperone RbAp46 (RBBP7) [164].

RbAp46 and its fly homolog p55 bind histone H4 [167] and sNASP preferentially binds histone

H3 [168], providing a logical explanation as to how histone dimers are held by the sNASP-

RbAp46 dual chaperone complex. When H3-H4 dimers are passed to the sNASP-RbAp46

complex a dramatic reduction in H3K9me occurs by an unknown mechanism, and HAT1

Page 33: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

18

catalyzes H4 acetylation on K5 and K12 (Fig 1.1). sNASP can dimerize [169], and complexes

have been observed containing one or two H3-H4 dimers, but not tetramers [164] which, as

described later, form in the nucleus (Fig 1.1). In budding yeast (S. cerevisiae) the equivalent of

sNASP is Hif1, and the Hat1p/Hat2 holoenzyme performs acetylation, much like the mammalian

HAT1-Rbap46 complex. HAT1 or Hat1p driven K5ac and K12ac modifications appear to

enhance association with nuclear transporters and thus nuclear uptake [170, 171].

NASP is essential for murine development [166], and plays a key role in adjusting the

soluble reservoir of H3-H4. Reducing NASP leads to autophagy-mediated depletion of soluble

H3-H4, whereas reducing HAT1 (and thus histone acetylation) or Asf1 does not lower cytosolic

H3-H4 [172]. Thus, NASP provides a tunable cytoplasmic source of H3-H4.

After passing from the HSC70 to the tNASP complex and then to the

sNASP/Rbap46/HAT1 complex, the final cytosolic stage involves the transfer of H3-H4 dimers

to a complex containing the conserved chaperone ASF1 and Importin-4. There are two human

ASF1 genes, A and B, but only the B protein is found at this stage, although A likely takes over

when B is artificially removed [164]. Importin-4 is a karyopherin family member that mediates

transport through the nuclear pore. ASF1 acts as a sink for H3-H4 dimers since most non-

nucleosomal nuclear histone H3-H4 is found associated with this chaperone [173].

Once in the nucleus, H3.1-H4 dimers pass from ASF1 to the CAF1 (chromatin assembly

factor 1) complex. H3.3-H4 dimers are handled by HIRA for deposition on genic regions [159,

161], or DEK for an as yet uncharacterized target [158], and DAXX for deposition on

heterochromatin [162, 163, 174]. For simplicity, we will focus on the H3.1-H4/CAF1 interaction

below, but we will also discuss DEK and DAXX in the section on chaperones and cancer.

CAF1 was originally isolated as a human complex that promotes in vitro nucleosome

assembly on replicating SV40 viral DNA templates [142]. It is conserved from yeast to humans

and consists of three subunits termed Cac1-3 in yeast [175], p180, p105 and p55 (NURF) in

Drosophila [176, 177] , or p150 (CHAF1A), p60 (CHAF1B) and p48 in humans [142]. The latter

is also called Rbap48 (RBBP4), a close relative of Rbap46 (RBBP7) mentioned above (see

sNasp/Rbap46/HAT1 complex, (Fig 1.1)). Asf1 can bind directly to the mid-sized subunit of

CAF1 (p60/CHAF1B) [178, 179], while the small subunit (Rbap48) binds both H3 and H4 [180].

Page 34: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

19

For many years it was unclear whether newly synthesized histones H3 and H4 form

tetramers prior to deposition by CAF1, or only after deposition on DNA. Three recent studies all

suggest the former. The first found that mutations affecting neuronal fate in C. Elegans mapped

to a C-terminal region of histone H3 required for H3-H4 tetramerization [181]. C. Elegans has

24 histone H3 genes, but mutation of only one acted as a dominant negative to block

nucleosome formation. Depleting CAF1 or PCNA (required to recruit CAF1 – see below)

caused the same neuronal phenotype, whereas depleting Asf1 did not alter fate. These data

suggest that the phenotype results from an inability of CAF1 to assemble H3-H4 tetramers. The

second piece of evidence came from cross-linking studies which found that a single molecule of

yeast CAF1 binds H3-H4 tetramers [182]. And the third involved thermodynamic studies

showing that once yeast ASF1-H3-H4 binds to CAF1, ASF1 is dislodged and a second H3-H4

dimer is introduced to form the H3-H4 tetramer (Fig 1.1) [183].

The latter study also found that the affinity of CAF1 for unmodified H3-H4 dimers is ~2-

fold lower than that of ASF, raising the question of how the transfer could be thermodynamically

favorable. Notably, however, post-translational histone marks increase the affinity of histones

for downstream chaperones [184]. In the nucleus the yeast or mammalian acetyl transferases

Rtt109 or CBP/p300 catalyze H3K56 acetylation, respectively, which is stimulated considerably

by Asf1 or the yeast histone chaperone Vps75 [185-187], and this modification promotes

association of H3 with CAF1 or the yeast histone chaperone Rtt106 [188]. Yeast lacking Asf1 or

H3K56 acetylation have a reduced life span [189]. Another modification that influences binding

is PAK2-mediated phosphorylation of H4 Ser 47, which diverts H3.3-H4 dimers away from

CAF1 to HIRA [190]. Thus, chaperones facilitate histone modification and/or exploit these

events to guarantee the passage of histones along the chaperone chain in the right direction and

with the correct partners. Several other marks have been described on newly synthesized

histones, several of which affect chromatin assembly and sensitivity to DNA damaging agents,

but the underlying mechanisms are largely unclear [191].

Page 35: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

20

The final step in the journey of a new H3-H4 tetramer is deposition onto its highest

affinity partner, DNA. For the CAF1-(H3-4)2 complex, this is facilitated by interaction with

PCNA (proliferating cell nuclear antigen). PCNA forms a trimeric clamp that completely

encircles replicating DNA strands [192]. Its interaction with DNA involves water molecules,

allowing it to glide along the template and improve DNA polymerase processivity (the number

of nucleotides replicated without polymerase dissociation). The large subunit of CAF1

(p150/CHAF1A in humans) binds directly to PCNA [146]. As discussed below, CAF-1 is critical

for the inheritance of heterochromatin, and mutations in yeast PCNA that affect CAF1

recruitment disrupt heterochromatin-mediated silencing [193], and this interaction is also critical

for chromatin assembly after DNA damage [194]. Moreover, dominant negative CAF1 mutants

that do not bind PCNA or p60/CHAF1B disrupt chromatin deposition and lead to activation of

the DNA damage checkpoint [195].

In summary, chaperones guide H3-H4 dimers from their synthesis in the cytoplasm to

their destination on DNA through a thermodynamically favorable chain of binding reactions (Fig

1.1).

1.5.5 Chaperones recycle old histones during replication

Chaperones also disassemble nucleosomes as the replication fork passes, and then reassembles

them on both strands of replicated DNA. The first step in the disassembly of parental

nucleosomes is removal of H2A/H2B. FACT (facilitates chromatin transcription) is an

H2A/H2B chaperone and while most of the work on its function focuses on transcription, it is

also involved in altering chromatin structure during DNA replication [196, 197]. FACT is

recruited to the replication fork through an interaction with MCM4, one of six proteins (MCM2-

7) that make up the helicase that unwinds DNA [198]. Another chaperone that could be involved

Page 36: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

21

in removing H2A/H2B from DNA is Nap1 (nucleosome assembly protein 1) which operates in

collaboration with the ATP-dependent chromatin remodeling factor RSC in vitro [199]. Nap1 is

thought to be particularly important for removal of H2A/H2B during transcription [200],

reviewed in [201].

Once H2A/H2B is removed, the more stable H3/H4 can be displaced. H3/H4 dimers are

then assembled onto nascent DNA quickly, whereas histones H2A and H2B are added 2-10 min

after fork passage [202]. Asf1 plays a key role in coordinating the removal of H3/H4 dimers with

their re-deposition behind the fork [203]. Asf1 splits (H3-H4)2 histone tetramers into dimers and

an Asf1-H3/H4-MCM sandwich briefly tethers them to the helicase (Fig 1.1). Knockdown of

both Asf1 genes (Asf1a & b) prevents nucleosome removal, the helicase fails to unwind DNA

(reflected in reduced levels of single stranded (ss) DNA levels at the fork), and cells arrest in S-

phase. Inhibiting DNA polymerase causes an accumulation of parental histones on Asf1,

identifiable by covalent marks absent on newly synthesized histones (H3K9me3/ H4K16ac)

[203]. Asf1 is also required to bring new H3-H4 dimers to DNA (see above [204]), thus when

H3-H4 dimers are over-expressed, Asf1 becomes limiting, parental histones are not dislodged,

the helicase stalls, ssDNA levels drop, and cells arrest in S-phase [203]. This S-phase block can

be rescued by elevating Asf1 levels [203]. Therefore, Asf1 coordinates both recycling of parental

and introduction of new histone H3-H4 dimers.

The above data raise an interesting conceptual problem. As noted earlier, (H3.1-H4)2

tetramers exhibit conservative replication [155]. But Asf1, which plays a key role in tethering old

H3-H4 dimers to the replication fork [203], splits tetramers [150, 205]. Once old H3.1-H4

tetramers are split into dimers, how do they re-associate and remain separate from new H3.1-H4

dimers? Re-association of old dimers might be the only option given that new tetramers are

preassembled on CAF1 (see above). Whether H3.1-H4 dimers from an old nucleosome remain

closely associated to a pair of Asf1 molecules at the fork is unclear. Perhaps Asf1 traffics old

histones through CAF1 at the fork, although in vitro assays show that CAF-I cannot assemble

histones H3 and H4 purified from cellular chromatin onto DNA [142, 206]. Whether this is the

case at the replication fork in vivo, however, is unclear. Nap1 or its close relative Vps75, form

homodimers that adopt an earmuff structure and directly bind the two H3 proteins in an intact

(H3-H4)2 tetramer [207], but whether this feature is exploited during conservative replication of

Page 37: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

22

the core nucleosome is unknown. Alternatively, covalent modifications and/or associated

proteins (chaperones?) on old and/or new dimers preclude mixing during deposition.

There are two ASF genes in mammalian cells and while knockdown of both ASF1A and

B is required for acute arrest of cultured cells, removing ASF1B, but not A, blocks growth in

colony formation assays [208]. The reason for this difference is not fully resolved, but ASF1B

has other qualities that distinguish it from 1A, such as its down-regulation in quiescent or

senescent cells, and ASF1B deficiency causes unique effects on the transcriptome, and the

appearance of mitotic defects such as micronuclei and DNA bridges [208].

Page 38: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

23

Fig 1.1 Histone chaperone functions during DNA replication. In the cytoplasm (top) HSC70 chaperones newly translated H3, which is methylated by an unknown enzyme. Next, HSP90 and tNASP chaperone H3K9me1, and histone H4 joins the complex to generate H3-H4 dimers. H3-H4 is then transferred to a complex containing the chaperones sNASP and RBAP46, and the enzyme HAT1. H3 is demethylated a this stage by an unknown enzyme, and HAT1 acetylates H4 on K5 and K12. Next, the acetylated H3-H4 dimer is passed to ASF/importin for nuclear import, following which CBP/p300 acetylates H3 on K56. The latter facilitates transfer to the final chaperone in the chain, CAF1, which assembles H3-H4 dimers into (H3-H4) 2 tetramers. CAF1 is tethered to PCNA at the replication fork and deposits the (H3-H4)2 tetramer on DNA, following which H2A-H2B dimers are deposited, for example, by the dimeric chaperone complex FACT, which is tethered at the fork through an interaction with MCM4 in the helicase. At the fork the helicase unwinds DNA (towards the left), and behind it (to the right) polymerases (not shown for simplicity) synthesize the new leading or lagging strands of DNA. Ahead of the helicase, parental H2A-H2B dimers are displaced by FACT, and ASF1 then separates the remaining H3-H4 tetramer into dimers. The resultant H3-H4-Asf1 complex is tethered to the fork through an interaction of the histones with the helicase. H3-H4 is then redeposited on DNA, but it is unclear whether this occurs through another chaperone intermediate (e.g. CAF1 etc), or whether tetramers form directly on DNA after release from ASF1 (see text). The octamer is then completed by addition of two H2A-H2B dimers. Old and new histone H3-H4 tetramers do not mix (as shown), but old and new H2-H2B dimers can mix (here, for simplicity, no mixing is indicated).

Page 39: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

24

1.5.6 Chaperones and Epigenetic Memory: The Example of CAF1

Maintenance of nucleosomes during replication or repair is, by definition, a key aspect of

epigenetic memory. However, in addition to passing on old and depositing new histones,

chaperones play additional epigenetic roles. The full extent to which chaperones regulate this

process and the mechanisms therein are largely obscure. Most work has been performed on

CAF1 and ASF1, particularly on maintenance of heterochromatin. Below, we summarize the

data for CAF1 [209].

Heterochromatin is typically rich in repetitive DNA, such as centromeric satellite

sequences and telomeric repeats. These dense domains are gene poor, less accessible, coated

with repressive hypoacetylated histones and histone H3 trimethylated on lysine 9 (H3K9me3),

and replicated late in S-phase. H3K9me3 tethers heterochromatin protein 1 (HP1) which self-

associates to promote condensation [210]. Moreover, HP1 recruits the H3K9 methyl transferases

SUV39H1 and SUV39H2, which propagate this chromatin mark during replication (Fig 1.2)

[211, 212]. The DNA in these dense regions is easily visible upon staining with intercalating

fluorescent dyes such as 4$,6-diamidino-2-phenylindole (DAPI). DAPI-intense heterochromatin

spots remain even during their replication, so how is such densely packed DNA and chromatin

duplicated? As discussed below, CAF1 plays a major role in this process. Indeed, CAF1 is

crucial for heterochromatin maintenance in yeast, plant, fly, frog, mouse and human cells [213-

217].

Links between CAF1 and heterochromatin arose from work in budding yeast (S.

cerevisae) where, although dispensable for survival, its subunits are essential for efficient

silencing of marker genes proximal to telomeres [175, 218, 219]. Initially, it was thought that

silencing at mating type loci did not require CAF1, but more sensitive assays revealed that CAF1

is essential to maintain silencing, but not for re-establishment (e.g. following Sir protein

disruption) [220].

Heterochromatin in S. cerevisae relies on proteins like Rap2 and the Sir family of histone

deacetylases, but different factors are utilized in higher eukaryotes, such as the HP1 family.

Page 40: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

25

Remarkably, however, subsequent work provided that a direct link also exists between

heterochromatin maintenance and CAF1 in higher eukaryotes.

This connection came from the discovery that the N-terminus of murine Chaf1A/p150

binds directly to HP1 [219]. In human or murine cell lines, CAF1 colocalizes with HP1 in late

S-phase at sites of pericentric heterochromatin [221]. Labeling with thymidine analogues, such

as BrdU, and 3D imaging showed that heterochromatin is replicated at the surface of DAPI-

dense spots, and is then buried inside the domain after replication [222]. In addition to

trimethylation by Suv39h1/2, HP1 binding to the core of DAPI-rich heterochromatin also

requires an RNA component, but in contrast Chaf1A/p150 tethers an RNase and an Suv39h1/h2-

null resistant HP1 fraction to replicating DNA on the surface of heterochromatin domains in

mouse fibroblasts [222]. These data suggest that when DNA emerges at the surface of a

heterochromatic domain, HP1 is displaced and tethered at the replication fork by Chaf1a. Indeed,

while Chaf1a/p150 or Chaf1b/p60 knockdown reduces nucleosome deposition in S-phase DNA,

only the former causes cell cycle arrest in mid-S-phase, which is not associated with a DDR, but

rather an inability to replicate pericentric heterochromatin [222]. This RNAi-induced defect is

complemented by wild type Chaf1a, but not by mutants that do not bind HP1. Moreover, Chaf1a

knockdown does not arrest Suv39h1/h2 double null fibroblasts where HP1 is absent from DAPI-

rich heterochromatin [145]. Altogether, these data suggest that CAF1 displaces HP1 during

heterochromatin replication, and holds it at the replication fork ready for re-deposition (Fig 1.2).

CAF1-HP1 [222] exists separately from CAF1-H3.1-H4, thus a single CAF1 complex

deposit newly synthesized histones or handles old/new HP1 separately. CAF1-HP1 binds other

proteins critical for heterochromatin maintenance, including Methyl Binding Domain 1 (MBD1)

[223], which recruits the H3K9 methyltransferase SETDB [224]. While bound to CAF1,

SETDB1 stimulates mono-, but not di- or tri-, methylation of H3K9 [225]. Monomethylated K9

is an excellent substrate for di and tri methylation by Suv39h1 (Fig 1.2).

Page 41: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

26

In summary, CAF1 performs many functions relevant to the epigenetic inheritance of

heterochromatin (Fig 1.2): i. As at other loci it loads new histone H3.1-H4 tetramers onto

replicated DNA; ii. It displaces and re-deposits old parental HP1 at the surface of

heterochromatin domains, and brings in new HP1 to maintain heterochromatin on replicated

DNA; iii. CAF1-HP1-MDB1-SETDB1 mono-methylates H3K9 on new nucleosomes, allowing

Suv39h1/h2 - tethered to old nucleosomes – to convert H3K9me to H3K9me3, and the latter can

now receive old or new HP1 from CAF1.

As well as maintenance, CAF1 is also important for the de novo formation of murine

heterochromatin as Chaf1a null mouse embryos arrest at the 16 cell stage and fail to form DAPI-

rich spots [226]. In ES cells, where chromatin is more plastic than in differentiated cell types,

Chaf1a knockdown does not cause arrest, perhaps because HP1 is easier to displace in these cells

than fibroblasts. However, these depleted ES cells die after 4 days of knockdown [226], perhaps

because the failure to form heterochromatin disrupts chromosome segregation, although the latter

was not tested. Whether this survival function for CAF1 depends on interaction with HP1 has

not been tested.

The above model of Chaf1-HP1 mediated regulation of heterochromatin was established

primarily in mouse cells. The picture is less clear in other animal species/cell types. In human,

mouse, and chick cells removing Chaf1a/p150 or Chaf1b/p60 impairs nucleosome deposition in

S-phase [145, 195, 227-229]. In chick DT40 cells, like mouse fibroblasts, there is no DDR when

CAF1 is disrupted (indeed the response to UV or HU is dampened) [229]. However, whereas

only Chaf1a knockdown perturbs S-phase progression in mouse cells, knockout of Chaf1a or

Chaf1b delays S-phase in chick cells, and also causes extensive cell death by 48 hrs [145, 229].

Binding of chick Chaf1a to PCNA and Chaf1b is required for survival, but the interaction with

Page 42: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

27

HP1 is dispensable [229]. Whether Chaf1a HP1-binding mutants affect heterochromatin

replication in chicks is unknown. Unlike chick/mouse cells, CHAF1A small interfering RNA

(siCHAF1A) causes a DDR in the human cancer cell lines RTK, HeLa, and U2OS cells [195,

227], as does siCHAF1B in HeLa cells [228]. Viability was compromised in both the

U2OS/siCHAF1a and HeLa/siCHAF1B assays. Intact heterochromatin is important for proper

nucleation of spindles at mitosis, thus cell death in some of these scenarios might follow

disruption of this key process, but may also be the result of an S-phase DDR in some cases.

However, neither scenario applies in chicks as there is no DDR, and HP1 binding is dispensable

for survival. The extent to which the above variable responses to CAF subunit disruption reflect

differing chaperone redundancy, species/tissue specificity, and/or degree of neoplastic

transformation is unclear. Notably, an RNAi screen in HeLa cells identified CHAF1A as critical

to maintain silencing of an integrated GFP reporter gene [230]. Other heterochromatin

regulators were also identified, such as HP1 and SETDB1, consistent with the model discussed

above.

Page 43: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

28

Fig 1.2 Multiple roles for CAF1 at heterochromatin. Solid arrows indicate protein movements, dotted arrows indicate methylation events. Old and new nucleosomes, PCNA and DNA are shown as in Fig 1.1 i. CAF1 tethered to PCNA deposits new H3-H4 tetramers onto DNA (see Fig 1.1 for details). ii. A separate CAF1 complex tethers SETDB1 to the fork through an interaction with MBD1, and this enzyme monomethylates H3K9. This CAF1 complex also contains new HP1 to deposit on trimethylated H3K9. iii. SUV39H1, tethered to HP1, di and trimethylates H3K9. iv. New HP1, tethered to CAF1, is deposited on H3K9me3. v. SUV39H1 is transferred to newly deposited HP1. vii. Old (parental) HP1 is transferred to a third CAF1 complex at the fork. vii. Old HP1 is transferred (in this example) onto old, already trimethylated H3K9 (see Fig 1.1 for details on how old nucleosomes are transferred at the fork). viii. SUV39H1 is transferred to old HP1. It is likely that old and new HP1 is distributed randomly among old and new nucleosomes, although this has not been tested. The top right illustrates two complexes of CAF1 bound to one PCNA trimer, which is theoretically feasible, but whether this occurs, or whether CAF1-(H3-H4)2 and CAF1-MBD1-SETDB1-HP1 complexes bind sequentially (i.e. separately) is unknown. New and old SUV39H1/2 is not differentiated, but new enzyme is likely drawn from the nucleoplasm.

Page 44: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

29

1.5.7 Other potential roles for CAF1 in maintaining repressive chromatin states

Beyond its well established role in regulating HP1 bound heterochromatin, CAF1 may regulate

other repressive mechanisms. The Polycomb group (PcG) of proteins represses transcription at

multiple loci, including homeobox transcription factors required for specific developmental fates

[23]. Heterozygous mutations in the large Drosophila CAF1 subunit (p180, equivalent to

p150/CHAF1A in humans) enhance the effect of heterozygous mutations affecting the PcG

protein, Pc [216]. This genetic evidence suggests that CAF1 may facilitate the epigenetic effects

of PcG complexes. But how this operates, and the specific PcG complexes affected are unknown.

The CAF-1 subunit, RBBP4 (Rbap48) or p55 (Nurf55) in Drosophila, is also a subunit of

Polycomb Repressive Complex 2 (PRC2) *, but these functions are separate because structural

studies show that Nurf55 uses the same region to interact with histones H3 and H4 (as part of its

role in CAF1) and with SUZ12 (a component of PRC2) [180]. There is no biochemical evidence

that CAF-1 and PcG interact directly, but conceivably CAF-1 might recruit other proteins to the

fork that influence PcG activity. Apart from well known interactions with PCNA, ASF1,

histones, and HP1-MBD-SETDB1 (see above), CAF1 binds several other proteins [231], but

their role in epigenetic inheritance is unclear.

1.5.8 CAF1 and Cancer

In view of their central role in regulating nucleosome density, histone modifications, chromatin

structure, genome stability, and epigenetics it seems logical that cancer cells might manipulate

histone chaperones to promote mutagenesis and/or alter gene expression. The field is young, and

while most links are indirect, there are some striking examples of how chaperones influence

cancer progression. We review some key examples below.

Disrupting the CAF1 specific subunits p150 (CHAF1A) or p60 (CHAF1B), or their

equivalents in lower organisms, disrupts nucleosome frequency, and, in many cases, causes

Page 45: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

30

spontaneous DNA damage and/or increased susceptibility to DNA damage inducing agents [175,

195, 227]. Moreover, because CAF1 regulates heterochromatin, disrupting this function could

lead to large scale defects in chromosome alignment and segregation at mitosis. However,

inactivating mutations in CAF1 subunits have not been reported in cancer, likely because their

depletion is lethal in many circumstances [226, 228, 229]. Nevertheless, subtle sequence

variants, changes in post-translational modifications and/or altered levels of chaperones could

affect genome stability or gene expression. Indeed, excess CHAF1B/p60 correlates with poor

outcome in some cancers [232]. The genetic connection between CAF1 and Polycomb

phenotypes in Drosophila is also intriguing since excess Polycomb activity is common in human

tumors [23].

1.5.9 ASF1, HIRA and cancer

As discussed earlier, ASF1A and B are critical to buffer excess histones during replication stress

[233], and disrupting this process promotes a delayed DNA damage response [208]. In addition,

ASF1B has unique roles in long-term growth, gene regulation and chromosomal stability, and

elevated ASF1B expression is associated with high mitotic index, poor tumor grade and worse

outcomes in breast cancer [208]. Also, ASF1A is part of a network of factors critical to repress

the pro-apoptotic gene Fas in K-ras transformed cells [234]. ASF1 stimulates H3K56 acetylation

to promote transfer to the CAF1 complex and subsequent deposition on DNA (see above). This

histone modification promotes longevity in yeast [189], and is elevated together with ASF1 in

several cancer cell types [187]. H3K56ac is also important at sites of DNA repair [235].

Normally, this mark is rapidly removed upon H3 deposition, but is maintained at repair sites to

increase nucleosome “breathing” and facilitate repair complex access to DNA [236, 237]. The

connection between ASF1, CAF1 and H3K56ac provides a logical explanation for increased

DNA damage in the absence of one or more of these components [235].

The above data imply oncogenic roles for ASF1. Intriguingly however, ASF1A together

with the H3.3-H4 chaperone HIRA is also critical for Ras-induced senescence, an anti-cancer

response to DNA damage that forces permanent cell cycle exit [238]. Cancer cells overcome

Page 46: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

31

senescence by inactivating the RB and p53 tumor suppressor pathways, and it is presumably

beyond this point that the putative oncogenic roles of ASF1A discussed above come into effect.

The ability of ASF1 to promote senescence is intimately linked with the histone

chaperone HIRA. Normally, HIRA deposits H3.3 at genic or telomeric regions [161], but in

senescing cells it is required for the formation of Senescence Associated Heterochromatic Foci

(SAHF) that silence RB-E2F regulated cell cycle genes [239].

Early in senescing cells, PML bodies, also linked to senescence, contain HIRA and HP1,

and although ASF1A is not found in these structures, a HIRA-ASF1A interaction is critical for

SAHF formation [238]. Late in senescence the repressive H2A variant macroH2A (which also

has a separate role in silencing the X chromosome) is recruited to SAHF, which also requires the

HIRA-ASF1A interaction. ASF1B cannot bind HIRA and is not involved in senescence. Neither

HIRA nor ASF1A bind macroH2A, thus the chaperone for the latter in senescing cells is unclear,

although APLF (aprataxin and PNKP like factor) chaperones macroH2A after DNA damage

[240]. Whether HIRA-ASF1A dependent macroH2A deposition is linked to H3.3 and/or the

ability of ASF1 to dislodge pre-existing nucleosomes (see above) is unknown. In yeast, where

there is no macroH2A, interaction of the HIRA (Hir1 and Hir2) and Asf1 equivalents promotes

silencing at telomeres and repression of histone gene expression [241-245]. Consistent with its

pro-senescence function, low levels of ASF1A are associated with longevity in humans [246],

but as noted earlier, Asf1 is actually required for longevity in yeast [189].

1.5.10 DEK, DAXX, H3.3 and Cancer

Apart from HIRA, DEK and DAXX also chaperone H3.3, and strikingly both are mutated in a

variety of tumor types, providing direct evidence linking chaperones to cancer.

DEK is a highly abundant protein (almost as abundant as nucleosomes) with several roles

in regulating chromatin structure. The Drosophila ecdysone hormone binds the nuclear receptor

EcR and its ability to promote gene activation depends on DEK [247]. In the same study, DEK

was shown to promote nucleosome formation by chaperoning H3.3, which was dependent on

Page 47: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

32

CKII phosphorylation of DEK. Disrupting the latter blocked H3.3 binding, nucleosome

formation in vitro, and ecdysone mediated gene activation.

DEK also has roles in repressing transcription. Thus independent of CKII (and thus its

H3.3. chaperone function), DEK promotes heterochromatin through an RNA-dependent

interaction with HP1. Loss of DEK results in the concomitant depletion of HP1 and H3K9me3

from either constitutive pericentric heterochromatin or promoters silenced by the latter repressive

epigenetic mark [158]. Fruit fly DEK is in the same class as the H3K9 methyl transferase

Su(var)3-9, because its inactivation relieves position effect variegation (PEV), the repression

linked to translocation of a previously active gene to a location near heterochromatin [158].

Thus, both CAF1 and DEK have key roles in binding HP1 and regulating chromatin.

In line with a repressive function for DEK, it restricts access of complexes required for

transcription to chromatin templates in vitro, and it is dislodged by SET, a protein required for

Pol II-mediated transcription on such templates [248] . Intriguingly, SET, like DEK, is found as

a CAN/NUP214 fusion protein in leukemia [249].

DEK is linked to cancer in two ways. First translocations have been found in a subset of

AML that fuse its N-terminus to the C-terminus of the nucleoporin CAN (NUP214) [249]. This

translocation switches the location of CAN from the nuclear pore to the nucleoplasm [250]. The

DEK-CAN leukemic fusion protein cannot bind CKII, abolishing its histone chaperone activity,

arguing that defects in H3.3 deposition may be important for transformation [247], although the

critical targets are unclear. The effect of the translocation on HP1 or SET activity is also unclear.

Second, DEK over-expression is observed in various cancers, either through gain of its

chromosomal location 6p22 [251-254] or through induction by E2F, such as in cells expressing

the RB-inactivating human papilloma virus E7 oncoprotein [255, 256]. DEK over-expression

elevates H3K9me3 at silenced chromatin, which could be oncogenic in some contexts [158].

However, the effect of DEK over-expression on chaperoning H3.3 or the ability of SET to open

chromatin is unknown. DEK influences a variety of cancer hallmarks, such as survival,

senescence, DNA repair and invasiveness [251, 256-260], but exactly which of these biological

functions, if any, require its activating and/or repressive molecular chromatin roles remains

unclear. Complicating matters further, DEK has also been linked to other activities, such as

splicing and protein translation [261-263].

Page 48: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

33

DAXX was first identified as a FAS-binding protein and interacts with other factors that

regulate cell survival [264]. Three recent studies have shown that it is a histone H3.3 chaperone

[161-163]. Drane et al report that DAXX is more tightly associated with H3.3 than HIRA, and

that it promotes nucleosome deposition on plasmids in vitro, which was also demonstrated by

Lewis et al. Consistent with prior work [265, 266] , DAXX and the SWI/SNF-like chromatin

remodeling enzyme ATRX (#-thalassemia mental retardation X-linked syndrome) were found at

pericentric satellite repeats. These repetitive regions are transcribed, which Drane et al found

requires DAXX, H3.3 and ATRX. DAXX-mediated deposition of H3.3 on pericentric DNA is

replication independent and H3.3 co-localizes with DAXX and ATRX in PML bodies, which is

blocked if DAXX is missing. DAXX/ATRX-mediated recruitment of H3.3 to PML bodies and

pericentric heterochromatin is reminiscent of the requirement for HIRA to transport H3.3 to

PML bodies and senescence associated heterochromatin (see above).

Goldberg et al [161] focused on the genome-wide localization of H3.3, demonstrating

that it is enriched at known regulatory elements (active or poised, proximal or distal), across the

bodies of active genes, and at telomeres. HIRA was essential for enrichment at promoters and

gene bodies, but not at remote regulatory elements or telomeres. Like Drane et al they found

that H3.3 associates with DAXX/ATRX, and while ATRX was dispensable for H3.3 enrichment

at genic regions or regulatory elements it was essential at telomeres. Moreover, Lewis et al

found that DAXX, like ATRX, is essential for H3.3 deposition at telomeres [163]. Finally,

ATRX loss elevates transcription of the telomeric repeat-containing transcript TERRA [161].

Thus, paradoxically, DAXX/ATRX/H3.3 recruitment is linked to induction or down-regulation

of pericentric or telomeric transcription, respectively [161, 162]. The structure of DAXX bound

to a H3.3-H4 dimer has been solved and reveals specific interactions that explain why this

chaperone preferentially binds H3.3 rather than replicative H3 isoforms [174].

Recent deep sequencing efforts have exposed direct evidence for a link between

DAXX/ATRX and cancer. First, mutations in DAXX or ATRX were reported in almost half of

pancreatic neuroendocrine tumors [267]. Consistent with the key role of DAXX/ATRX in

depositing H3.3 at telomeres these mutations or loss of nuclear DAXX/ATRX are linked to

abnormal telomere structures, and analysis of other cancers with aberrant telomeres exposed

ATRX, although not DAXX, mutations in some brain tumors [268]. A subsequent study found

both ATRX and DAXX mutations in pediatric glioblastoma samples, but also a high frequency

Page 49: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

34

of H3.3 mutations that target Lysine 27 or Glycine 34 [269]. These variants were linked to

perturbed telomeres and altered transcription profiles, which is consistent with the H3.3

chaperone function discussed above, and with the link between lysine 27 methylation and

Polycomb-mediated repression. Finally, DAXX mutations have also been described in a subset

of AML patients [270].

1.5.11 Future directions

The term chaperone conjures up rather pedestrian images, yet these proteins have diverse

functions and their deregulation has drastic effects on cell homeostasis. The last decade has seen

dramatic advances in our understanding of their roles in replicating nucleosomes, histone

exchange, deposition of histone subtypes, epigenetic inheritance, transcription and DNA repair,

yet there remain many unanswered questions. How, for example, are old and new (H3-H4)2

tetramers kept apart at the replication fork? What precise mechanisms are used to replicate old

H2-H2B dimers or deposit new dimers? Why does CAF1 perturbation have such different

effects in distinct species/cell types on DNA damage or survival? Which functions of CAF1

(e.g. recruiting H3-H4, HP1, SETDB1 etc) are critical in these various contexts? What is the

molecular link connecting CAF1 to Polycomb function, and how is this function played out in

organisms other than Drosophila? The ties between histone chaperones, cancer and longevity

are also fascinating, but we only have a superficial understanding. What is the relevant

molecular effect in these biological contexts: DNA damage, gene expression, Polycomb

function, heterochromatin, and/or another role, and what are the key molecular players that

chaperon influence? What are the signals that switch HIRA from regulation of genic or

telomeric regions to silencing of cell cycle genes destined to be buried in SAHF? Or that control

the ability of ASF1 to promote senescence in collaboration with HIRA, versus silencing of pro-

apoptotic genes? And what are the precise functions of DEK, DEK fusions, and DAXX that are

so critical in driving cancer progression? These are only a few of the questions that will keep

researchers busy in the chaperone field for the next decade.

Page 50: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

35

1.6 Hypothesis

Previous work in our lab showed that in the BRG1-deficient SW13 adenocarcinoma cells, PRC2

blocks IFN!-induction of CIITA and a subset of other ISGs. We detected the PRC2 signature

epigenetic mark, H3K27me3, across the CIITA locus and at the promoters of other PRC2-

regulated ISGs. We hypothesized that there should be unknown components that collaborate

with PRC2 in repression of ISGs. The knowledge of what these factors are and how they

orchestrate the silencing of ISGs may improve our understanding of PRC2 mediated epigenetic

memory in general. To address this issue, we performed siRNA screens using a BAC-CIITA

vector containing all the remote elements crucial for CIITA regulation. Here we present an

unexpected novel link between CHAF1A and PRC2. We show that siCHAF1A rescues PRC2-

repressed genes in a cell cycle dependent manner. Interestingly, siCHAF1A does not lead to a

loss but a redistribution of H3K27me3 mark. Therefore, we believe that CHAF1A coordinates

repressive epigenetic memory by regulating the genomic positions at which PRC2 methylates

histone H3K27 during DNA synthesis.

Page 51: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

36

Chapter 2

2 RNAi Screens to Identify new components regulating IFN! responsive genes (ISGs)

Page 52: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

37

2.1 Abstract

Interferon-! (IFN!) signaling is a crucial pathway in the cells of the immune system and its

aberrant regulation is linked to many diseases including cancer. Therefore, identifying its

regulators is of much interest. The IFN! signaling pathway is initiated by the binding of a ligand

to the IFN! receptor which activates the signal transducer and activator of transcription (STAT1)

through the Janus (JAK1/JAK2) kinases. This in turn leads to the induction of IFN! stimulated

genes (ISGs). We have previously shown that Brahma-related gene 1 (BRG1), the ATPase that

drives the SWI/SNF chromatin remodeling complex, is essential for the expression of class II

transactivator (CIITA), an ISG and the master regulator of the class II major histocompatibility

complex (MHC II). For example, in the BRG1-deficient human adrenal carcinoma SW13 cell

line, the CIITA locus is unresponsive to IFN!. Surprisingly, however, a short CIITA reporter,

containing only the IFN!-responsive promoter IV (PIV) was BRG1-independent. These

observations led us to search for and identify remote elements, several kbs away from the

transcriptional start site that block IFN!-responsiveness of the CIITA locus in BRG1-deficient

cells. We also reported that BRG1 is normally recruited to these elements and drives the

chromatin conformational changes that counter the inhibition of CIITA expression. Our lab built

a 194 kb BAC-CIITA luciferase (Luc) reporter containing all the newly identified remote

regulators and showed that the construct is, like the endogenous CIITA locus, also BRG1-

dependent. Here, we took advantage of this tool and performed high throughput (HTP) siRNA

screens to determine: A. whether we could rescue CIITA responsiveness in BRG1-deficient cells

by suppressing factors that repress this target (Suppressor screen) and B. which additional

regulators (activators or inhibitors) affect CIITA regulation in the presence of BRG1 (Wild type

screen). In this chapter, the optimization procedures for the different steps of the siRNA screens

are discussed and the identified hits are reported.

Page 53: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

38

2.2 Introduction

IFN! is secreted by activated immune cells in response to the presence of pathogens and tumor

cells. This triggers protective defenses of the immune system aimed to eradicate pathogens or

tumors [131, 135],3. Since almost all types of cells respond to IFN!, exploring the mechanism by

which IFN! signaling pathway is regulated is of much interest [131, 271]. The IFN! signaling

pathway is initiated by binding of IFN! to its receptors which activates the JAK1 and JAK2

tyrosine kinases[272]. JAK1/JAK2 activation leads to phosphorylation of Signal Transducer and

Activator of Transcription (STAT1) [133, 135]. Once phosphorylated, STAT1 homodimerizes

and translocates to the nucleus and its incorporation with interferon regulatory factor 1 (IRF1)

contributes to the induction of Interferon gamma responsive genes (ISGs) including class II

activator (CIITA), a master regulator of MHC Class II [33, 133, 135, 273]. There are several

feedback mechanisms that control and inhibit the IFN! signaling pathway. For example, the

suppressors of cytokine signaling (SOCS) proteins, a family of SH2-containing proteins are

crucial negative regulators of cytokine signaling transduction [274, 275].

CIITA is a coactivator that induces the MHC Class II promoter[139]. It is constitutively

active in immune cells such as B lymphocytes, activated T lymphocytes and dendritic cells or is

induced by stimulation in most cell types [276]. CIITA is regulated by four promoters (PI-IV) in

a tissue specific manner [276]. PI and PIII are constitutively active in dendritic cells and B

cells[276]. PIV is activated by IFN! in a variety of non-immune cells [276]. Defects in CIITA

regulation are associated with bare lymphocyte syndrome (BLS), a sever immune disease, and

other human diseases, including cancer, multiple sclerosis, arthritis and myocardial infarction

[138, 277]. Studies show CIITA is often silenced in tumor cells [278, 279]. As mentioned

above, cancer cells can be recognized and eliminated by immune cells through interferon

secretion (immunosurveillance) [131]. CIITA silencing is thought to be a mechanism by which

cancer cells escape immunosurveillance, therefore, understanding the mechanism of CIITA

regulation can be exploited in cancer drug design.

Previous studies in our lab uncovered several cis-acting elements and trans-acting factors

essential for the IFN! responsiveness of CIITA. First, we reported that CIITA responsiveness to

IFN! requires Brahma related gene1 (BRG1), the catalytic subunit of the SWI/SNF family of

ATP-dependent chromatin remodeling complexes [33]. Next, our lab showed that although

Page 54: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

39

endogenous CIITA gene induction requires BRG1, induction of CIITA in short reporters, even

though properly chromatinized, is BRG1-independent[33, 115]. Chromatin immunoprecipitation

(ChIP) coupled with genomic tiling arrays (ChIP-chip) identified STAT1 and IRF1 binding sites,

as well as IFN!-induced histone modifications in a >100 Kb long stretch upstream and

downstream of the CIITA promoter [137]. ChIP-qPCR at the CIITA locus confirmed remote

events at -50, -16, -8, +40, and +59 kb, and low signal at intervening sites [137]. Next,

chromosome conformation capture (3C) showed that these functionally relevant hubs loop and

contact each other and also the transcription initiation site, and they are crucial for CIITA

activation[137]. Interestingly, we showed that remote regulatory sites are also involved in

regulating other IFN! responsive genes [137].

To study the function of CIITA regularity distal elements, Zuyao Ni in our lab built a 194

kb CIITA bacterial artificial chromosome (BAC) reporter containing all the remote elements (-

50, -16, -8, +40 and +59 ) regulating CIITA upstream of an IRES-Luc cassette [137]. BRG1-

deficient human adrenal carcinoma SW13 cells were transfected with the BAC vector and

SW13-BAC stable cell clones were selected in G418 [137] . Importantly, the selected clones

showed that IFN!-induced activation of Luc mimics the endogenous CIITA as it is also BRG1-

dependent[137]. Replacing the -50, -16, -8, +40 or +59 enhancers in the BAC blocked

responsiveness to IFN! [137]. These results show that BRG1-dependency at CIITA is conferred

by remote elements. In turn, this model suggests the existence of remote silencers that block

CIITA induction and BRG1 is required to overcome.

Next, to identify the silencers, which confer the BRG1 dependency of CIITA,

Mohammad Abou El Hassan in our lab examined silencing epigenetic marks across the CIITA

locus. He discovered H3K27me3 at remote sites (unpublished data). H3K27me3 modification is

mediated by the polycomb repressive complex 2 (PRC2)[280]. PRC2 contains 4 subunits: the

methyltransferase subunit EZH1/2, SUZ12, EED and histone binding proteins RBBP46/48.

PRC2 was first discovered in Drosophila where it represses Hox genes in multiple lineages, and

is linked to stem cell maintenance and cancer in mammals [281]. Interestingly, Brahma (the fly

homologue of BRG1) counters Polycomb activity [282, 283]. Further, Mohammad showed that

the PRC2 subunits EZH2 and SUZ12 are recruited to remote sites across the CIITA locus.

Introducing BRG1 to the system reduced (albeit modestly) the repressive epigenetic marks and

the recruitment of PRC2. Most importantly, knockdown of PRC2 subunits rescued

Page 55: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

40

responsiveness of CIITA and other BRG1-dependent ISGs. Taken together, these data

underscore the collaboration of BRG1 and PRC2 in the regulation of CIITA and suggest that

polycomb complexes regulate IFN! signaling in higher vertebrates.

To better understand the CIITA regulation, we decided to identify additional factors

involved. Importantly, in the recent years, siRNAs and development of the high-throughput

(HTP) genome-wide RNAi-based technology allowed for the siRNAs screen to serve as a

powerful and rapid alternative approach to study loss-of-function phenotypes especially for

characterizing novel pathway components [284-287].

RNAi silencing is a sequence-specific and post-transcriptional gene silencing. It initiates

when RNase III-like enzyme Dicer processes dsRNAs to small 21-23 basepair (bp) siRNAs.

These small siRNAs are then incorporated into the RNA-induced silencing complex (RISC)

[284] which is a multiprotein complex that incorporates siRNA or miRNA as a template to find a

complementary mRNA strand, it activates RNAse and cleaves the mRNA and decreases

production of the protein of interest by mRNA cleavage [284, 288, 289].

A previous siRNA screen to identify new components of JAK/STAT signaling pathway

in Drosophila revealed more than 100 functionally important proteins including signaling

factors, enzymes mediating post-translational protein modifications and transcription factors

[285, 290]. However, these studies used small reporters containing a small promoter of the gene

of interest. As our data shows, long-range elements far from transcription start site may have

crucial role in ISGs regulation. Therefore using small reporters as a read out for RNAi screen

might miss crucial regulatory factors.

In contrast, our BAC reporter incorporates essential remote regulatory elements of CIITA

transcription and mimics that of endogenous CIITA. We took advantage of this tool and

performed RNAi screens to identify new IFN! signaling regulators in the context of the CIITA

locus. We used a clone of SW13 cells, stably transfected with the BAC vector (SW13-CIITA-

BAC)[137]. In this way, we were able to perform two types of screens. A “Suppressor screen”

was performed in the absence of BRG1, which aimed to identify new factors involved in the

repression of CIITA. The second, Wild type (Wt) screen, was performed in the presence of

BRG1, by transducing SW13 cells with BRG1 adenovirus. In this latter screen we looked for

new IFN! signaling components, activators or inhibitors, normally associated with the CIITA

Page 56: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

41

regulation. Next, we selected 91 hits from each screen and performed secondary screens to

validate these hits using different siRNAs than the ones used in the primary screen.

We expect that the factors involved in the regulation of CIITA will likely be also used to

regulate other ISGs. Thus, our screens should allow us to deepen our understanding of ISG

regulation and hopefully provide new therapeutic targets for diseases caused by dysfunctions of

IFN! signaling.

Page 57: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

42

2.3 Material and Methods

2.3.1 Cell culture and adenovirus

Adenocarcinoma SW13 cells were grown in alpha-Mem media supplemented with 10% FBS.

Cells were treated with human IFN! (0.1 µg/ml; BioSource International). Virus was produced

and SW13 cells were transduced as previously described [115] . The amount of virus was

'titrated' so that BRG1 expression was equivalent to that in HeLa cells as described before [115].

Clone #38 of SW13 cells stably transected with BAC vector (SW13-CIITA-BAC38) was

maintained in alpha-MEM media supplemented with 10% FBS and 500 %g/ml of G418 as

described before [137].

2.3.2 HTP siRNA screening

For our screens we used the Dharmacon Human Genome siARRAY siRNA SMART pool library

based on polymerase based reaction (PCR) templates with an average length of 21 bp for the

screen. The entire library consists of 267 x 96 well plates in which 80 wells per plate (columns

3-12) each contain a pool of four siRNAs targeting one particular transcript (80 wells x 267

plates = 21,121 targets). Columns 1 and 2 are blank and are used for negative and positive

control samples. The library is divided into four sets of siRNAs, representing the kinome (10

plates: 779 targets), G protein coupled receptors (7 plates, 516 targets), other druggable targets

(76 plates, 6022 targets) and the rest of the genome (174 plates, 13,804 targets).

For our purposes we used 6801 siRNAs from the kinome and druggable libraries. 96

well plates were loaded with 2.5%M dsRNAs in 2 %l of siRNA buffer. The first row contained

non-targeting siRNAs as negative controls and the second row contained STAT1 siRNAs as

positive controls. For the robotic transfection, 10%l of serum and antibiotic free media was

added to each 96 plate well containing the siRNAs. The plates were incubated at room

temperature for 5 min. Next, 10 %l serum and antibiotic free media containing 1% dharmafect

lipid #1 was added to each well. The plates were incubated for 20 min at room temp. Next,

10,000 cells in 100%l serum containing media with %10 FBS were dispensed per well using and

automated liquid dispenser. In the WT screen, after 3 days, BRG1 adenovirus was added to the

plates without removing the media. At day 5, cells were lysed and luciferase activities were

Page 58: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

43

determined. For the secondary validation screen three 96 well plates were loaded with 91

Qiagen dsRNAs and 2 negative controls (PTBP1 and none targeting siRNA) and 4 positive

control for INF! pathway (JAK1, STAT1, IFNR1, SOCS1). Each plate contained 50nM of the

three different siRNAs targeting one particular gene. Cells were dispensed and transfected as

described for the primary screen. BRG1 adenovirus transfection and luciferase assay were

performed as described above.

2.3.3 BAC construction and reporter assays

Details of BAC construction and SW13-BAC38 were described before [137] . Luciferase assays

were performed using Promega kit as described before[115].

2.3.4 Statistical Methods for Hit Selection

Three methods were used to analyze HTP screen data, B-score, M-score and control base

normalization [291-293]. The B score uses the ratio of a raw value over a measure of variability

to smooth noise due to edge effects, missed well and plate to plate variation [291]. In the M-

score, the data were normalized using a robust form of the Z-score (i.e. Zero mean and unit

variance standardization)[292] whereby the median replaced the mean and the median absolute

deviation replaced the standard deviation. This normalization was termed “M-score”, and is

shown in Equation 1:

The M-score was applied on a plate-wise basis, such that the median and median absolute

deviation was derived for each individual plate. This corrected for variation in overall plate

intensity during the course of the experiment [292, 293]. Control base normalization calculates

the ratio of the luciferase activity in each well to the luciferase activity of negative siRNA loaded

in the first well of the same row.

(1)

Page 59: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

44

2.4 Results

2.4.1 The HTP siRNA screen optimization

To optimize the conditions for our HTP siRNA screens, I first wanted to ensure that in the

SW13-CIITA-BAC38 cell line, IFN! induces CIITA expression in the presence of BRG1, and

knocking down of the known CIITA activator STAT1 prevents induction.

Initially, we transfected SW13-CIITA-BAC38 cells with siSTAT1 for 3 days using four

different lipids, Dharmafect1, 2, 3 and 4. Since CIITA is not responsive to IFN! in the absence

of BRG1 in the BRG1-deficient SW13 cells, at day 3, we had to introduce BRG1 to the system

using adenovirus. The cells were transduced with the BRG1 adenovirus without removing the

transfection media and then IFN! was added. Cells were lysed, 24hrs later and luciferase

activity was assessed. I found that transfection of the siSTAT1 with Dharmafect 1, 2 and 4

inhibited Luc activities to similar degrees but Dharmafect 3 showed lower STAT1 knock down

efficiency based on Luc assay (Fig 2.4.1A). Thus, Dharmafect 1 was used in all subsequent

studies. Next, the amount of BRG1 adenovirus for optimal CIITA induction was determined.

Using 1%l of purified virus containing 4x106 PFU showed that more than 90% of cells were

transduced and expressed the GFP reporter (Fig 2.4.1B).

Next, I optimized the concentration of siRNA. I showed that 100 nM and 25 nM of

Dharmacon STAT1 siRNA had the same knock down efficiency (Fig 2.4.1C). In order to

minimize possible off target effects I selected the lower concentration (25 nM) of siRNAs for the

screens. I, then optimized the order of addition of siRNA, adenovirus and IFN!. I tried four

different scenarios (Fig 2.4.2). The strategy that showed maximum IFN!-induction in the

presence of negative control siRNA (siCtrl, which does not target any gene) and maximum

inhibition with siSTAT1 was as follows; Day 1: plate cells, Day 2: add siRNA Day 5: add 1%l

BRG1 virus without removing siRNA, Day 6: add IFN! without changing the media for 24hrs,

Day 7: Luc assay (Fig 2.4.2).

To test the automated HTP version of optimized protocol, half of a 96 well plate was

loaded with siSTAT1 and another half with the siCtrl. Next, the media containing Dharmafect 1

was added to the plate. After 20 min of incubation, SW13-CIITA-BAC38 cells were robotically

added to the plate. After 3 days, BRG1 virus was added to the plate without removing media

Page 60: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

45

containing the siRNA. After one day, IFN! was added to the plate and Luc activity was assessed

after 24hrs of IFN! treatment. The result demonstrated that the automated assay was robust, had

excellent signal to noise ratios and was thus appropriate for conducting siRNA screens (Fig

2.4.3).

During the screen optimization step, the hits were analyzed by the “B score”. This

method avoids positional effects by using samples as their own controls [291]. To test this

method in our system, we seeded cells in a 96 well plate, in which only well 7B received

siSTAT1 and the remainder received control siCtrl. Importantly, the top B score was assigned to

the 7B well despite edge effects and variability, confirming the B score as an appropriate method

for hit-identification (Fig 2.4.4).

Our optimized assay was then used to assess BAC-CIITA levels in the presence of BRG1

and IFN! in a pilot screen of 10 plates of the kinome Dharmacon library set. This library

included two obligatory activators of IFN! signaling, JAK1 and JAK2, and one feedback

inhibitor, SOCS1, all of which are known to affect CIITA expression [294, 295]. The first

column of each plate received the negative control siCtrl and the second column contained the

positive control, siSTAT1. The signal showed robust knockdown by siSTAT1, proving effective

transfection in multiple wells on every plate. Importantly, the B-score analysis highlighted all

the embedded positive controls that such as JAK1 and JAK2 that were known IFN! activators.

With a cut off of 3 fold above the standard deviation (SD), the B score identified 4/5 of the true

positives and no false positives (80% sensitivity, 100% specificity), while a cut off 2 fold above

SD, it found 5/5 positives and 4 more unknown positives (100% sensitivity). These data support

the utility of this approach in our hands. Therefore, we decided to use B score with 2SD as one

of the statistical methods to analyze the hits of the rest of the screen. The B scores for the pilot

screen are shown in Fig 2.4.5 A. The pilot screen hits contained 17 IFN! activators that

decreased Luc activity, and 38 inhibitors, which caused induction in Luc activity (Fig2.4.5 B).

Importantly, JAK1 and JAK2 were the top two activators, and SOCS1 was the fifth ranked

inhibitor. JAK3 and SOCS5, neither of which regulates IFN! signaling, had B scores close to

zero (not shown). Interestingly, several of the novel hits in our kinome screen are linked to IFN!

signaling or BRG1 function. For example, STK11 (LKB1) is one of the activator hits which is

known that interacts with BRG1 and is required for BRG1-dependent growth inhibition [296].

FRK is another hit in the pilot screen that is shown is involved in IFN! signaling in islet cells

Page 61: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

46

and MAP4K5 (GCKR) which shown that is activated by type I and II IFNs was also identified as

a hit in the pilot screen [297, 298].

These data illustrated the power of our screen to highlight signaling networks to identify

new components of IFN! or those that may cooperate with BRG1 to regulate CIITA. We next

performed HTP siRNA screens with the rest of the siRNA library.

Page 62: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

47

Fig 2.4.1 Optimizing siRNA transfection and viral transduction conditions. A. A clone of SW13-BAC38 cells (see text) was transfected with siSTAT1 or siControl using four different transfection reagents (Dharmacon#1, #2, #3, #4) . Next, cells were transduced with BRG1 adenovirus and stimulated with IFN!. Luc assay was performed 24hrs after the exposure to INF!. Similar results were obtained with each reagent, although Dharmafect #1, #2 or #4 worked the best. B. To optimize the amount of BRG1 adenovirus, SW13-BAC-38 cells were transduced with the virus for 24 hrs followed by IFN! treatment for 6 hrs. We found that 1µl of the virus containing 4x106 plaque-forming units (PFU) infected more than 90% of the cells and the highest level of Luc activity occurred after 6hrs of IFN! treatment. For the screen we selected the amount of the virus that provided maximum BAC-CIITA induction (green arrow). C. To optimize the amount of siRNA, we transfected SW13-BAC38 cells with 100nM and 25nM of Dharmacon siCtrl and siSTAT1. Both concentration had the same knock down efficiency. We used 25nM of siRNA for the screen.

Page 63: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

48

Fig 2.4.2 Optimizing the timing and order of the screen steps. SW13-BAC38 cells were seeded on 96 well plates and 1 day later subjected to four different protocols as indicated. Optimal IFN!-induced Luc activity and inhibition with siSTAT1 but not siCtrl was achieved with protocol #3.

Page 64: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

49

Fig 2.4.3 Automated HTP version of the optimized protocol. SW13-BAC38 cells were robotically seeded in a 96 well plate, half of which contained control or STAT1 siRNA as indicated. The plate was incubated for 3 days and then was transduced with BRG1 virus and treated with IFN! as shown in protocol #3 in Fig 2.3.2.

Page 65: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

50

Fig 2.4.4 Testing the “B score” method of hit analysis in the optimized protocol. We performed our optimized robotic protocol (Fig 2.3) for a 96 well plate in which 95 wells received siCtrl, and well 7B received siSTAT1. Then we used “B score” method to analyze the result. Note the edge effect in column 12, and variability among position elsewhere, yet the B score ranked STAT1 siRNAs highest.

Page 66: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

51

2.4.5 A. Plot of B scores from the pilot Wt screen. SW13-BAC38 were transfected with Dharamcon Kinome library (10 plates) in the present of BRG1 as shown in Fig2.3.4. The hits were identified by the B score method and 2 threshold lines were used. The blue line represents 3 SD from the mean and the yellow line represents 2 SD from the mean. B. Activators and inhibitors of CIITA induction from the pilot Kinome screen. Hits are ranked according to the B score. Blue color represents > 3 SD; yellow color represents > 2 SD from the mean. Canonical IFN! regulators are highlighted in pink.

Page 67: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

52

2.4.2 The primary WT and Suppressor screens

For our siRNA screen we used the Dharmacon kinome and druggable libraries (779+6022=6801

targets). Each of the 6801 dsRNA pools was aliquoted into 96-well plates. Negative control

siCtrl, was aliquoted in the first column of each plate and the positive control siSTAT1 was

loaded on the second column of each plate (Fig 2.4.6). Cells were incubated with siRNAs for 3

days. Next, cells were incubated for another day with BRG1 adenovirus (WT screen) or without

(Suppressor screen) followed by IFN! treatment for 24 hrs. Luciferase activity was then

measured and results assessed by 3 methods to identify hits.

In addition to the B score that we tested in pilot screen, we also used M score and

control-based normalization as explained in the methods section, with the goal of identifying

priority hits that were highly ranked in all three methods. The top 1% of hits obtained with each

of the three methods were selected and hits common to all three methods were ranked #1, those

common in 2 methods were ranked #2, while hits selected by only one method were ranked #3

(Fig 2.4.6A). We ranked 152 activators and 145 inhibitors from the primary WT screen and 114

inhibitor hits from Suppressor screen (Apx1.1-3). Scatter graphs of Wt and Suppressor hits

analyzed by the B-Score method are shown in Fig 2.4.6 B-C. CHAF1A and PCNA, the focus of

further analyses in Chapter 3, were prominent hits in the Suppressor screen.

Page 68: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

53

Page 69: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

54

Fig 2.4.6 Schematic diagram of the primary siRNA screen. Each of the 6801 dsRNA pools of Dharmacon siRNAs were aliquoted into 96-well plates. SiCtrl and siSTAT1 were loaded to the first and second columns of each plate, respectively. Next, SW13-BAC38 cells were transfected for Wt screen and Supp screens and luciferase activity was measured. The top 1% of hits obtained with each three analysis methods (B score and M score and Control Base) were selected. Further, the hits common to all three methods were ranked #1, those common in B score and M score were ranked #2a, B score and Control Base normalization ranked #2b and common hits in M score and Control Base normalization were ranked #2c. Hits selected by only one method were ranked #3 (3a for B score, 3b for M score and 3c for Control Base normalization). In this way in the primary Wt screen we ranked in total 152 activators, 145 inhibitors and in Suppressor screen we ranked 114 hits, see Apx1.1 -3. B. Wt screen activators and inhibitor hits identified by B-Score. Blue dots above the yellow line are hits based on B-Score method where the cutoff is 2.5 SD above mean. C. Inhibitor hits from the Suppressor screen identified by the B-Score. Hits above the red line which include CHAF1A and PCNA are more than that 9SD above the mean.

Page 70: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

55

2.4.3 The secondary WT and Suppressor screens for hits validation

To validate the hits from the primary screens, we performed secondary screens. For these

screens we selected 91 candidates from each WT or Suppressor primary screens. These hits

were the ones identified by all three or at least two statistical methods (M score, B score and

Control Base normalization), and some of the hits that were only identified by one of the

methods. The secondary screens were performed three times using three different Qiagen library

siRNAs for each hit. Importantly, these siRNAs are different from the Dharmacon library

siRNAs. The assays were also performed in 96 well plates like the primary screens. Two wells

of each plate were loaded with negative controls, PTBP1 siRNA, which was not a hit in the

primary screen selection process (shown yellow in Fig 2.4.7), “Qiagene All-star negative

siRNA”, and positive control siRNAs for IFN! signaling, IFNR1, JAK1, STAT1, SMARC4,

SOCS1 (shown orange in Fig 2.4.7). For the secondary screens SW13-BAC38 cells were

transfected with 50 nM of 3 separate Qiagene siRNAs, targeting 3 different sites of mRNA so

that there was one siRNA per gene per plate as shown in Fig 2.3.7, for 3 days. The cells were

then incubated for another day in the presence (secondary WT screen) or absence (secondary

Suppressor screen) of BRG1 adenovirus. After this IFN! was added to the cells and luciferase

activity was measured. Data was analyzed by the M score method. To exclude genes affecting

cell proliferation or survival, the alamar blue assay which stains dead cells, was run prior to

collection of lysates for the Luc assay.

By the M score method of normalization, of the 91 activator and inhibitor hits selected

for the secondary Wt screen, 22 hits (26%) were verified, and in the secondary Suppressor

screen, 11 hits out 91 were verified by at least one siRNA (~10%). Except KPNB1, which was a

hit verified by the secondary Wt screen, none of the other validated hits affected cell

proliferation. The hits were classified, based on PubMed and Genecards, based on their

predicted functions (Apx1.4-6).

Page 71: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

56

Figure 2.4.7 Schematic diagram of secondary validation screen. 92 hits (both activators and inhibitors) from the primary Wt screen and 92 hits from the primary Suppressor screen (i.e., inhibitors) were selected for the secondary screen validation. SW13-BAC38 cells were transfected by 50 nM of 3 separate siRNAs for 3 days and were incubated for another day in the presence (secondary Wt screen) or absence (secondary Supp screen) of Ad-BRG1. After 24 hrs IFN! was added to the cells and luciferase activity was measured. To exclude genes affecting cell proliferation or survival the alamar blue assay was run prior to collection of lysates for the Luc assay. The validated hits are listed in Apx1.4-6

Page 72: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

57

2.4.4 Tertiary Validation on Endogenous CIITA

In the next step, I tested whether the siRNAs that rescued responsiveness of the BAC-

CIITA reporter in both primary and secondary screens also altered the responsiveness on

endogenous CIITA locus in SW13-deficient SW13 cells. Real time PCR analysis validated the

siRNAs against the histone chaperones CHAF1A and PCNA (for results see Chapter 3).

However, other hits were not validated at the endogenous level (data not shown).

Page 73: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

58

2.5 Discussion

IFN! signaling pathway plays essential roles in the proper development and function of the

immune system [139]. Its deregulation is associated with several human diseases including

cancer [131]. IFN!-mediated signaling leads to the activation of ISGs including CIITA, the

crucial co-activator of the MHC Class II locus. This gene is silent or mutated in many cancers

[138, 277]. Likely, the loss of CIITA and MHC Class II is used by tumor cells to escape from

the body’s immune surveillance. Therefore better understanding of IFN! signaling components

and CIITA regulation will help us not only to fill in some of the gaps in this field but will also

lead to an improved drug design.

Previous studies have showed that the regulation of CIITA is highly complex, for

example there are four promoters and long-range enhancers, which loop and contact the

promoters upon the IFN! response [115, 276]. We also showed that the IFN! responsiveness of

CIITA requires BRG1 [33, 137]. Here, I performed siRNA screens to identify unknown

repressors that confer BRG1 dependency of CIITA and to discover additional components of

IFN! signaling that are regulating the CIITA responsiveness to IFN!.

In contrast to the previous siRNA screens to find new components of JAK/STAT

signaling, which used short reporters [285, 290], we used a BAC vector containing distal

enhancers crucial for CIITA induction. The BAC vector behaves liked the endogenous CIITA

locus, meaning that CIITA-BAC is silent in the absence of BRG1 and is responsive to IFN! in

the presence of BRG1 [137]. This allowed us to conveniently perform 2 screens at the same

time. In the first screen, in the presence of BRG1 (WT screen), in which we aimed to identify

novel IFN! activators and inhibitors, we identified 152 potential activators and 145 potential

inhibitors. In the absence of BRG1 (Suppressor screen) we identified 114 potential silencers that

blocked CIITA induction.

As the first step towards validating these hits, we performed a secondary screen for some

of the hits identified from both primary screens. From each primary screen we selected 91 hits to

perform secondary screens. The secondary screens validated 10% and 26% of hits from

Suppressor and WT screens, respectively. Interestingly among the validated hits in the WT

screen, there are some well-known IFN! regulators such as IFN! receptor1 (IFNGR1), JAK1 and

JAK2 and SOCS1, which attests to the ability of our system to detect true positives. I did not

Page 74: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

59

follow up on Wt screen hits at the endogenous level of CIITA. However, further assessment of

hits from the BAC-CIITA Suppressor screen using the endogenous CIITA locus revealed that

while siRNAs against PCNA and CHAF1A rescued IFN! -responsiveness, other siRNAs did not.

These observations suggest that while some important aspects of CIITA regulation are

maintained in our reporter lines others have been disrupted. The hits we observed with the

reporter but not the endogenous locus may be false positives or, alternatively, there may be

additional levels of repression exerted upon the endogenous locus that are missing in the

reporter. Therefore, in our screens using this reporter, it would be better to have had more strict

criteria for selecting hits from primary screen. For example, in the B-Score graph showing hits

from Suppressor screen CHAF1A and PCNA are ranked 5th and 7th (Fig 2.4.6C). Thus,

assessment of the top ten hits would have been sufficient to expose the low frequency of true

positives. With hindsight, this would have been preferable to our actual approach of validating

the top 1% of hits with the Luc reporter, and then carrying apparently validated hits forward to

the endogenous gene assay..

Our original goal was to perform a genome-wide screen, but we paused after screening

~25% of the genome to validate which, in view of our low endogenous validation rate, proved to

be a wise decision. In light of our findings, it would be better to pursue genome wide screens

using a read-out of endogenous gene activity, such as the protein product of an ISG, or a reporter

integrated into the endogenous locus. (e.g. Luc or GFP fused in frame to the N- or C-terminus of

an ISG).

Overall, our screens characterized novel and interesting regulators never before

associated with the IFN! network. Considerable additional work is required to define which of

the hits in our WT screen affect endogenous ISG regulation, as well as to define whether any of

the Suppressor Screen hits that did not affect endogenous CIITA actually play a role in

suppressing ISG expression in the absence of BRG1. In the next chapter, we provide extensive

data linking two of the suppressor screen hits to the mechanism of epigenetic regulation by

PRC2. Moreover, our data reveal that this link is of general significance, beyond the regulation

of ISGs. This discovery radically changes our understanding of how epigenetic memory is

maintained.

Page 75: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

60

Chapter 3

3 CHAF1A Regulates the Distribution of PRC2-mediated epigenetic mark, H3K27me3, During Replication

All figures were generated by Mina Rafiei except Figure 3.4.4B,C,D; and 3.4.8C; and 3.4.9; and

3.4.10 B,C which were generated by Tom Leung. Also some unpublished data mentioned in the

text was generated by Mohamed Abou El Hassan (manuscript in preparation).

Page 76: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

61

3.1 Abstract

CIITA (Class II Major Histocompatibility Complex, Transactivator) is a crucial interferon

gamma (IFN!) responsive gene (ISG) and a master regulator of MHC class II. Defects in CIITA

regulation lead to severe immune diseases and are linked to other types of diseases including

cancer. In unpublished work, our lab showed that in SW13 adenocarcinoma cells, which is

deficient for BRG1, a subunit of the SWI/SNF chromatin remodeling apparatus, Polycomb

Repressive Complex -2 (PRC2) blocks IFN!-induction of CIITA and a subset of ISGs. We also

detected the PRC2-mediated epigenetic mark, H3K27me3, across the CIITA locus and at the

promoters of other PRC2-regulated ISGs. Re-introducing BRG1 reduces the H3K27me3 mark

permitting access to transcription factors and RNA polymerase, which leads to CIITA induction.

These data reveal antagonism between PRC2 and BRG1 in the context of ISG regulation in

cancer cells. To find other factors involved in this process, we performed an siRNA screen using

a BAC-CIITA vector containing all the remote elements crucial for CIITA regulation (Chapter 2).

Here, we focus on one of the hits found in this screen, Chromatin assembly factor 1, CHAF1A,

part of the CAF-1 nucleosome deposition complex, a replication fork protein and histone

chaperone. We showed that siCHAF1A restores IFN!-responsiveness at the endogenous CIITA

locus. The expression of other PRC2-repressed genes (ISGs and non-ISGs) was also rescued by

siCHAF1A and in a DNA replication-dependent manner. Importantly, removing CHAF1A

reduced the H3K27me3 repressive mark across the CIITA locus and at the promoters of other

PRC2-repressed genes. Surprisingly, siCHAF1A did not affect bulk levels of H3K27me3, but

caused a striking redistribution of the mark. Thus, CHAF1A coordinates repressive epigenetic

memory by regulating the genomic positions at which PRC2 methylates histone H3K27 during

DNA synthesis.

Page 77: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

62

3.2 Introduction

CIITA (Class II Major Histocompatibility Complex, Transactivator) is an IFN! responsive gene

(ISG) which is required for the expression and induction of MHC class II [133, 299]. CIITA and

MHC class II genes are constitutively expressed in immune cells such as dendritic cells and B

cells but are inducible in most cells in response to IFN! [115, 131, 135, 136]. IFN! is secreted

by activated T and NK cells to inhibit proliferation, virus infection and tumorigenesis [131, 135].

IFN! signaling is initiated by binding of IFN! to its receptors, activating the Janus kinases JAK1

and JAK2 which then phosphorylate Signal Transducer and Transactivator-1 (STAT1).

Phosphorylated STAT1 forms homodimers that are transferred to the nucleus where they

collaborate with Interferon Regulatory Factor1 (IRF1) to induce ISGs including CIITA [132, 300,

301]. Previously, we showed that in spite of normal IFN! signaling, CIITA is silenced in

adenocarcinoma SW13 cells, which are deficient in BRG1, a subunit of the SWI/SNF chromatin

remodeling apparatus, and re-introducing BRG1 rescues CIITA responsive [115, 127, 137, 278].

Since activated immune cells are able to distinguish and destroy cancer cells

(immunosurveillance), it is thought that cancer cells might silence CIITA to inactivate MHC

class II and thus escape immunosurveillance [131, 302]. Identifying CIITA regulators will

improve our understanding of IFN! signaling and facilitate the development of strategies to

battle the diseases associated with CIITA defects. Towards this goal Mohamed Abou El Hassan

in our lab detected Polycomb Repressive Complex 2 (PRC2) components and the PRC2-

mediated repressive epigenetic mark, trimethylated lysine 27 of Histone H3 (H3K27me3) across

the CIITA locus (unpublished data).

Polycomb group proteins (PcG) are chromatin associated complexes that repress

thousands of genes involved in development and differentiation in plants and mammals [303-

306]. Recent findings have linked PcG proteins to cell cycle control, chromosome X-

inactivation, cell fate decision, stem cell differentiation and cancer [307-311]. The core of PRC2

complex in mammals, consists of the methyl transferase Enhancer of Zeste Homolog 2 (EZH2),

or its paralog EZH1, which catalyze H3K27me3, Suppressor of Zeste 12 (SUZ12), and one of

the Embryonic Ectoderm Development (EED) isoforms [280, 312]. There are some other co-

factors such as histone binding proteins, PCL, RBBP4 (RbAp48) and RBBP7 (RbAp46) and

JARID, a JmjC domain containing demethylase, which are not part of the core complex but help

the recruitment and enzymatic activity of PRC2[23, 280]. Also, recent data suggests that certain

Page 78: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

63

histone and DNA-modifying enzymes (such as DNMT1) and long non-coding RNAs may guide

PRC2 to its gene [23]. Despite the wealth of information on PRC2, some fundamental issues

such as the mechanism as to how the H3K27me3 mark regulates gene silencing, how PRC2-

mediated epigenetic silencing is targeted to the appropriate loci, or how it is remembered during

replication still need to be addressed.

We observed that re-introducing BRG1 into SW13 cells modestly reduced H3K27me3

across CIITA, and facilitated induction of an active H3K4me mark at an upstream CIITA

enhancer (unpublished data). Moreover, BRG1 rescued IFN! responsiveness of this locus[137].

These observations indicate an antagonistic relationship between BRG1 and PRC2 at ISGs.

To gain more insight into the mechanism of PRC2-dependent repression of ISGs, we

performed an siRNA screen using a BAC-CIITA vector containing all the remote elements

crucial for CIITA regulation (Chapter 2). Among the hits identified by the screen was CHAF1A,

a subunit of CAF-1 histone chaperone (HC) [142]. The complex contains three subunits,

CHAF1A (P150), CHAF1B (P60) and RbAp48 (RBBP4) [142, 313]. To date, it is the only

known HC involved in the progression of replication and de novo incorporation of newly

synthesized H3/H4 after DNA replication. CAF-1 is recruited to the replication forks through a

direct interaction between its large subunit, CHAF1A, and Proliferating Cell Nuclear Antigen

(PCNA), the processivity factor for the DNA polymerases [146], which, importantly, was also a

hit in our screen.

Recently, multiple lines of evidence suggest that CHAF1A has a crucial role in

maintaining heterochromatin epigenetic marks (epigenetic memory) in yeast, Drosophila, plants

and mammals[146, 175, 218, 220, 314]. CHAF1A interacts directly with Heterochromatic

Protein (HP1a) and Methyl-CpG-binding domain protein 1 (MBD1) which recruits histone

deacetylases (HDACs) to their binding site [145, 223-225]. Moreover, SET domain Bifurcated 1

(SETDB1), a histone H3K9 methyltransferase forms a complex with both CHAF1A and HP1

during S- phase [224]. In this chapter we addressed whether CHAF1A is also required for

maintaining H3K27me3 during replication, both at ISGs and other PRC2 repressed targets.

Page 79: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

64

3.3 Material and methods

3.3.1 Cell culture and siRNA transfection

Human adenocarcinoma SW13 cells were grown in #-minimal essential media (#-MEM)

supplemented with 10% fetal bovine serum (FBS). Cells were treated with human IFN! (0.1

µg/ml; BioSource International) [137]. Fifty nM of siRNA were transfected for 3 days using

DharmaFECT-1 (Dharmacon) reagent according to the manufacturer’s instructions. Cells were

split at day 3, fresh media added, and incubated for 24 hrs. On day 4, cells were treated with

IFN! for 6 hrs and were lysate collected for mRNA extraction or Western blotting. SiRNAs

from Dharmacon and Qiagene used in this study are listed in Apx2.

3.3.2 RNA extraction and reverse transcription (RT)

RNA was isolated using TRIzol reagent (Invitrogen). A 2 %g aliqot of total RNA was diluted in

20%l of RNase free water, heated to 90º for 5 minutes, then combined with 30 %l of first strand

master mix [10%g of random Pd(N)6 primers, 72 U RNase inhibitor (Roche# 799-017), 1X first

strand buffer (Invitrogen#18064-014), 1mM dNTPs, 10mM dithiothreitol (DTT), 50U of

Superscript II reverse transcriptase (Invitrogen #18064-014)] and incubated for 1h at 37ºC

followed by 90ºC for 10 min. Next, 200 %l of water was added to the cDNAs to a total volume

250 %l. 4 %l of first strand cDNA was amplified using primers listed in Apx3 by RT-PCR. Copy

number of each cDNA was calculated from Qt values and was normalized to the level of HPRT

of the same sample.

3.3.3 Western Blotting

Cell lysate was prepared from a 100mm plate in 50mM Tris-HCL pH7.4, 150mM KCl, 15mM

NaCl, 30mM MgCl2, 10mM EGTA, 0.5 % NP40 and protease inhibitors. A 100 %g aliquot of

total cell lysate was sonicated and boiled with sample buffer (2% SDS, 100 mM dithiothreitol,

60 mM Tris [pH 6.8], 0.01% bromophenol blue) and separated on an SDS-7.5 % polyacrylamide

gel and transferred to a nitrocellulose membrane. Intensity of protein bands were quantified by

Odyssey. Antibodies used to detect the proteins are listed in Apx4.

Page 80: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

65

3.3.4 Chromatin immunoprecipitation (ChIP).

6 x 106 Cells per 100mm plate were cross-linked with 1% formaldehyde at room temperature for

10 minutes, washed two times with cold PBS, collected and centrifuged for 5 minutes at 1200

rpm. Cells were resuspended in 1.0 ml lysis buffer (1% SDS, 10mM EDTA, 50 mM Tris-HCL

PH 8) and protease inhibitors (1X aprotinin, leupeptin and pepstatin) from Sigma, were added to

the cell lysate. The lysates were incubated on ice for 10 minutes sonicated to an average size of

500bp using the sonicator (Viba Cell, Sonics and Material Inc, Danbury, USA). Next, chromatin

was pre-cleared with 25 %l of pre-cleared Staph A (Staphylococcal A cells, Calbiochem) for 15

min on a rocker at 4ºC. For immunoprecipitation, 100 %l of pre-cleared chromatin was used with

2%g antibody and incubated at 4ºC overnight. Next, samples were centrifuged and transferred to

new tubes and incubated with 10 %l of Staph A per IP for 15 min rotating at room temperature.

The samples were centrifuged for 5 min and the pellet were washed 2 times with ChIP Dialysis

Buffer (2mM EDTA, 50mM Tris-HCL pH 8, 0.2% sarkosyl), and 4 times with IP Wash Buffer

(100mM Tris-HCl pH 9, 500mMLiCl. 1% NP-40, 1% deoxycholic acid). The samples were then

eluted 2 times using 150 %l of ChIP Elution Buffer (50 mM NaHCO3, 1% SDS). Then, samples

were heated at 65ºC overnight to reverse the cross-links. After reverse cross-linking, DNA

fragments were purified with QIAEXII Gel Extraction Kit (Qiagen, Ca). Next, DNA was eluted

with 10mM Tris-HCl pH 8 at 42ºC for 10 min. The amount of DNA fragments precipitated was

detected by qPCR using selected primers.

3.3.5 Quantitative Real-Time PCR (qPCR)

4%l of each samples were mixed with SYBR Green PCR master mix (Applied Biosystem)

containing each primer and qPCR was performed in duplicate using Applied Biosystem PRISM

7900HT. The program consisted of 40 cycles of 95ºC for 15 seconds and 55ºC for 30 seconds.

A final cycle (95ºC, 15 seconds, 60ºC for 10 sec) generated a dissociation curve to confirm a

single product. A standard curve based on genomic DNA (1ng of DNA=300 copies) was

generated to quantify the copy number of each sample. In all cases, the low background signal

obtained with no antibody signal was subtracted. Details of primers used for qPCR assays are in

Apx3.

Page 81: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

66

3.3.6 Plasmid construction

CHAF1A CDNA was amplified from CHAF1A cDNA clone from Open Biosystems using the

following primers: attB1, 5’-GGGG ACA AGT TTG TAC AAA AAA GCA GGC TCA GCC

GCC ACC ATG CTG GAG GAG CTG GAG TGC-3’) and attB2, 5’-GGG G AC CAC TTT

GTA CAA GAA AGC TGG GTA - GGA TGC ACC CAG TGG GCT CGC-3’). The

amplified product was extracted from the Agarose gel using Qiagen Gel extraction kit and was

cloned into pLenti6/UBC/V5-DEST vector (Invitrogen) by LR reaction using Gateway strategy

as described and provided by Invitrogen.

3.3.7 Immunostaining

2x105 cells were seeded on coverslip in a 6 well plate and incubated overnight. Next cells were

pre-extracted with nuclear extraction buffer (50mM Tris, pH7.2, 300 mM NaCl, 1mM DTT,

0.5% (w/v) before fixing with 4% PFA for 10 min. Next, cells were incubated in cold BSA-PBT

for 1hr for blocking. Then primary antibody were added to the cells for 1hr washed 3 times and

secondary antibody were add for 1hr fallowed by washing for three times with PBT. Coverslips

were mounted with Vector Lab Vector Shield and viewed by Zeiss laser confocal microscope.

3.3.8 Cell cycle block

2x106 cells were per plate in 10cm plates ad incubated overnight. Next cells were transfected

with siCtrl or siCHAF1A for 2 days and media was refreshed with or without Aphidicolin on day

3. Next half of the plates were incubated with or without IFN! for 6 hours. IFN! treated plates

were harvested 6 hours later and stored -20. Another half of the plates were washed with PBS

for 3 times and then refreshed with normal media and incubated for 24hrs followed by treatment

with IFN! for 6hrs. Cell lysate and cDNA were prepared from each sample as described above

for Western blotting and RT-PCR analysis.

Page 82: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

67

3.4 Results

3.4.1 CHAF1A and PCNA repress CIITA induction

We showed before that CIITA is silent and not responsive to IFN! in SW13 BRG1-defecient

adrenal carcinoma cells[33]. Unpublished data in our lab showed that PRC2 components are

recruited to the promoter and distal sites at the CIITA locus and keep the gene silent. To identify

other components involved in CIITA silencing, we performed a Suppressor screen (i.e. to find

siRNAs that suppress defective ISG- responsiveness in BRG1 deficient cells) as described in

Chapter 2. For this screen, a part of the Dharmacon Human Genome siARRAY siRNA SMART

pool was used and CIITA-BAC reporter stably integrated into the genome of SW13 cells was

used as a molecular read-out. We selected two hits, CHAF1A and PCNA, from the Suppressor

screen for additional validation and functional studies. These genes are functionally related and

involved in H3/H4 deposition during replication.

First, we investigated the roles of CHAF1A and PCNA in endogenous CIITA induction.

To knock down CHAF1A and PCNA, three different siRNAs were used from Qiagene siRNA

library (siCHAF1A#2, siCHAF1A#3, and siCHAF1A #4) and one siRNA from Dharmacon

library (siCHAF1A#1). The western blot confirmed that all 4 siRNAs targeting either CHAF1A

or PCNA reduced the protein level of targeted genes by more than 50% (Fig 3.4.1A). The

mRNA level of knock down of CHAF1A and PCNA was confirmed by RT-PCR; more than 50%

reduction in mRNA level was observed (Fig 3.4.1B). Importantly, knocking down either

CHAF1A or PCNA using any of the siRNAs rescued the CIITA response to IFN! by

approximately seven fold or four fold relative to siCtrl, respectively, without changing CIITA

basal level (Fig3.4.1C). For the rest of this chapter, we focused on CHAF1A to investigate its

role in mediating PRC2 silencing of IFN! regulated genes.

Multiple siRNAs against CHAF1A rescued IFN! induction of CIITA in BRG1-deficient

SW13 cells (Fig 3.4.1A), indicating that this result was not due to an off-target knockdown. To

further validate this conclusion a stable cell line was generated overexpressing CHAF1A ~10-

fold (Fig 3.4.1D). When these cells were challenged with siCHAF1A#1 CHAF1A protein was

reduced but to levels equivalent to that in untreated or siCtrl-treated SW13 cells (Fig 3.4.1D).

When these cells were exposed to IFN!, CIITA remained silent, proving that the critical target of

siCHAF#1 in SW13 cells is CHAF1A (Fig 3.4.1E). These results demonstrate that

Page 83: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

68

overexpression of CHAF1A rescues the phenotypes observed in siCHAF1A treated cells,

confirming its involvement in CIITA silencing.

Page 84: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

69

Page 85: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

70

Fig 3.4.1 CHAF1A and PCNA repress endogenous CIITA induction. A. All four siRNAs targeting CHAF1A or PCNA reduce the protein expression. SW13 cells were transfected with 4 different siRNAs targeting CHAF1A (siCHAF1A #1, #2, #3, #4) or PCNA (siPCNA #1, #2, #3, #4) for 4 days. Western blots were performed with cell lysates collected after 6 hr treatment with IFN!. Upper panels show Western blots, lower panels graph quantification of protein levels. B. RT-qPCR was used to analyze RNA from cells in (A), confirming successful knockdown. C. Removing CHAF1A or PCNA rescues IFN! responsiveness of CIITA in BRG1-deficient cells. RT-qPCR was used to analyze CIITA levels in RNA from cells in (A). Results are plotted relative to the siCtrl transfected cells. D. CHAF1A over-expression in SW13 stable cells. SW13 cells were stably transduced with CHAF1A lentivirus (SW13-Lenti-CHAF1A), these cells or parental SW13 cells were transfected for four days with siCtrl (lanes1, 2) or siCHAF1A (lanes 3, 4) and lysates were collected after 6 hrs of IFN! treatment. Western Blotting showed 10 fold higher CHAF1A protein in SW13-Lenti-CHAF1A cells compared to parental SW13 cells, and siCHAF1A reduced the protein in the stable line to the level similar to that seen in parental SW13 cells. E. CHAF1A over-expression in siCHAF1A-treated cells reinstates CIITA repression. RT-qPCR analysis was performed on RNA from cells treated as in (D). Results are average of three experiments +/- SD.

Page 86: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

71

3.4.2 CIITA rescue by CHAF1A KD is BRM independent

BRG1 is the catalytic ATPase subunit of the SWI/SNF chromatin remodeling complex. In

humans, SWI/SNF associates either with BRG1 or BRM which share more than 70% amino acid

sequence identity[315, 316]. In some BRG1-mutant cell lines, including SW13 cells, BRM is

intact but silenced [317], raising the possibility that siCHAF1A might rescue CIITA

responsiveness by inducing BRM. Indeed, real time PCR showed an induction of BRM mRNA

in siCHAF1A treated SW13 cells (Fig 3.4.2B), although this did not result in detectable levels of

BRM protein as assessed by Western blotting (Fig 3.4.2A). To test whether CIITA rescue by

siCHAF1A was indeed independent of BRM, we performed double knock down using

siCHAF1A and siBRM. SiBRM reduced BRM mRNA as shown in (Fig 3.4.2B), but did not

impair rescue of CIITA to response to IFN! where CHAF1A was knocked-down (Fig 3.4.2C).

These results show that siCHAF1A rescues CIITA induction is not via induction of BRM.

Page 87: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

72

Fig 3.4.2 CIITA rescue by siCHAF1A is BRM-independent. A. Undetectable levels of BRM protein in siCHAF1A treated cells. SW13 cells were transfected with siCtrl, siCHAF1A or siCHAF1A + siBRM for 4 days and exposed to IFN! for 6 hrs. Western Blotting analysis showed successful CHAF1A depletion, but no detectable BRM protein. HeLa cell lysate was loaded as a control for BRM detection. B, C RT-qPCR analysis of RNA from cells treated as in (A) showed some induction of BRM RNA, which was down-regulated after exposure to siBRM (B), but with no effect on CIITA responsiveness (C). Results are average of three experiments +/- SD.

Page 88: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

73

3.4.3 The effect of siCHAF1A on known regulators of IFN! signaling

The binding of IFN! to its receptors activates a signaling cascade which leads to phosphorylation

of STAT1 (generating p-STAT1), and induction of IRF1, two major transcription factors for ISG

induction. It is possible that the effect of CHAF1A KD to rescue CIITA responsiveness could be

caused by the effect of CHAF1A on IFN! signaling regulators such as STAT1 and IRF1(Fig

3.4.3A) or p-STAT1(Fig 3.4.3B) . To address this question, we assessed the protein level of

STAT1, IRF1 and IFN! activated form of STAT1, p-STAT1, in CHAF1A-knock down cells.

Western blotting analysis showed there may be a small change in both p-STAT1 and IRF1, but

since this does not affect the levels of other BRG1/PRC2 independent ISGs (Fig 3.4.6A), we can

propose that it is unlikely to be the explanation for rescue of BRG1/PRC2 regulated genes.

Page 89: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

74

Fig 3.4.3 The effect of siCHAF1A on the level of IRF1, p-STAT1 or STAT1 proteins. SW13 cells were transfected with siCHAF1A or siCtrl for 4 days followed by IFN! treatment for for 6hrs. Western Blot analysis was assessed for STAT1, IRF1, p-STAT1.

Page 90: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

75

3.4.4 Effects of siCHAF1A on cell survival and DNA damage

Several biological effects of CHAF1A depletion have been reported before [194, 318]. In

mammals, but not in yeast, knock down or mutation of CHAF1A impairs nucleosomal assembly

during replication, and causes S-phase arrest which can lead to a DNA damage and apoptosis

[195, 228, 318] . Other studies also showed that CHAF1A is required in deposition of H3 to the

DNA damaged induced sites [319, 320]. To test the effect of siCHAF1A on cell survival, we

transfected SW13 cells with two different siRNAs and investigated cell viability at the time of

transfection and up to 4 days after transfection by trypan blue which selectively stains dead cells.

SiCHAF1A had little or no effect on cell death at day 4 (Fig 3.4.4.A). Immunostaining for

cleaved caspase 3, a marker of apoptotic cells, also detected no difference between siCHAF1A

and siCtrl treated cells (Fig 3.4.4.B). Moreover, we also assessed cell growth and carefully

examined four different sub-phases of S-phase identifiable by replication fork patterning (S1-

S4). We observed that while siCHAF1A slightly lowered the growth of SW13 cells, the

progression through S-phase was unaffected (Fig 3.4.4.C). Thus, siCHAF1A had marginal or no

effects on the growth and survival of SW13 cells. As discussed later in this chapter, inhibiting

the cell cycle by aphidicolin, that also induces DNA damage (Fig 3.4.4D) did not rescue IFN!

responsiveness of CIITA, and actually blocked rescue by siCHAF1A (Fig 3.4.10), arguing that

the effects of siCHAF1A on CIITA are not an indirect consequence of cell cycle inhibition or

DNA damage.

In line with prior reports [318] Immunostaining showed that siCHAF1A induced !H2Ax,

a marker of DNA damage (Fig 3.4.4 D). However, while treatment of SW13 cells with 2 %M of

the potent DNA damaging agent Methyl Methane-Sulfonate (MMS) also induced !H2Ax (Fig

3.4.6 D), it did not rescue responsiveness of CIITA to IFN! (Fig 3.4.6 E). In conclusion, the

effect of siCHAF1A on CIITA induction is not an indirect consequence of DNA damage.

Page 91: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

76

Page 92: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

77

Fig 3.4.4 Effects of siCHAF1A on cell growth, apoptosis and DNA damage. A. Cell growth is slightly effected by siCHAF1A. SW13 cells were transfected with siCHAF1A#1 and siCHAF1A#2 for 4 days. Cell number was measured by trypan blue staining and plotted. B. siCHAF1A does not induce apoptosis. SW13 cells were treated with siCHAF1A or siCtrl for 4 days then left untreated or exposed to IFN! for 24 hrs, or with etoposide as a positive control for apoptosis. Cells were stained with DAPI (blue) to mark nuclei and cleaved caspase 3 (red) to detect apoptosis. C. S-phase distribution is not affected by siCHAF1A. SW13 cells were treated as in B, incubated with EdU for 15min before pre-extraction and fixation and then stained for CHAF1A and PCNA and the characteristic pattern of replication fork distribution used to quantify the fraction of cells in each the four identifiable S phase stages (S1-S4). Cells in 10 randomly selected fields were quantified. D. siCHAF1A KD induces DNA damage. SW13 cells were treated as in B or were exposed to Aphidicolin and 2%M Methyl Methane-Sulfonate (MMS), both DNA damaging drugs, for 24hrs, then stained for DAPI (blue) and !H2Ax (green), a DNA damage marker. E. DNA damage does not rescue CIITA responsiveness. SW13 cells were treated 2 %M MMS for 24 hrs before IFN! treatment for 6hrs. RT-qPCR was used to measure CIITA mRNA levels. Results are average of three experiments +/- SD.

Page 93: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

78

3.4.5 Depleting other components linked to histone deposition, DNA replication or gene silencing does not rescue CIITA

We also used RNAi to deplete a series of other factors involved in DNA replication, DNA repair,

and gene silencing, none of which rescued IFN!-induction of CIITA (Fig 3.4.5A). 1. First Non-

coding RNAs have been also implicated in the recruitment of PRC2 and gene silencing in

heterochromatin [321, 322]. Argonaut proteins which are part of the RISC complex recruit non-

coding RNAs to DNA which facilitates PRC2-dependent gene repression. To test whether

Argonauts are involved in CIITA silencing, we knocked down different isoforms of Argonaut

proteins (Ago1 and Ago2). 2. There is evidence implicating DNA methylation in the regulation

of PRC2 and H3K27me3 distribution. Thus we knocked-down the DNA methyltransferase,

DNMT1. 3. We assessed various proteins that interact with CHAF1A to regulate H3K9me3

epigenetic marks: MBD1, EHMT2 (G9A), SETDB1, and HP1 (all 3 isoforms #, ", !) [223, 225,

323]. 4. Since CHAF1A functions during replication we examined other components of DNA

replication such DNA polymerase epsilon (POLE) and delta (POLD). 5. HIRA is another H3

histone chaperone involved in the replication-independent H3 deposition [324] 6. Further, since

CHAF1A is also involved in DNA repair we looked at RAD9A, a cell cycle checkpoint protein

required for cell cycle arrest and DNA damage repair in response to DNA damage [318]. Real

Time PCR showed more than 50% knock down of all targeted genes (Fig 3.4.5A). However,

none of these siRNAs mimicked the effect of siCHAF1A (Fig 3.4.5B).

We cannot firmly conclude that none of these proteins regulate CIITA responsiveness

because a) Westerns would be required to assess knockdown efficiency, and b) Several of the

proteins may function redundantly with related factors. Nevertheless, all these data stand in stark

contrast to the clear effect of siCHAF1A, underscoring the importance of CHAF1A in repressing

CIITA responsiveness in BRG1-deficient cells.

Human CAF-1 histone chaperone complex contains three subunits, CHAF1A (P150),

CHAF1B (P60) and RbAp48 (RBBP4) [182, 313]. CHAF1A and CHAF1B interact and are

required for replication dependent nucleosome assembly in human cells [313]. As described

above, CHAF1A, the largest subunit of CAF-1, is involved in CIITA silencing. To investigate the

specificity of CHAF1A subunit of CAF-1 complex in CIITA regulation we knocked down

Page 94: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

79

CHAF1B and RbAp48. Real Time PCR analysis showed that > 50% reduction of CHAF1B or

RbAp48 mRNA (Fig 3.4.5B) did not affect CIITA transcription (Fig 3.4.5A).

Page 95: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

80

Fig 3.4.5. Other CAF-1 components, DNA replication factors, alternative H3 histone chaperon, and known CHAF1A interacting proteins involved in gene silencing appear to be dispensable for CIITA silencing. A. SW13 cells were transfected with DNA replication factors siPOLD, siPOLE, a DNA repair factor, RAD9A , a H3 histone chaperone HIRA, gene silencing factors, siAGO1/2, DNMT1, HP1(#,",!), a histone lysine 9 methyltransferase, EHMT2 and CHAF1A interacting silencing factors, SETDB1 and MBD1 but none of them rescued IFN!-induction of CIITA. B. siRNAs targeting selected genes reduced the mRNA level of targeted genes more than 50%. SW13 cells were transfected with siRNAs for each genes for 4 days and were then collected after a 6 hr IFN! treatment. The percentage of remaining mRNA of targeted genes were measured by RT-qPCR and showed more than 50% knock down for the targeted genes. Results are average of three experiments +/- SD.

Page 96: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

81

3.4.6 CHAF1A represses multiple PRC2-regulated ISGs

Mohamed Abou El Hassan in our lab found that in addition to CIITA, multiple BRG1-dependent

ISGs are also repressed by PRC2 (unpublished data). Thus, we wondered whether CHAF1A

also collaborates with PRC2 in the repression of these other genes. To address this question, we

selected: IFI2, GBP1, GBP3, IFITM3 and examined the expression of these genes after

CHAF1A KD (Fig 3.4.6). We used IRF1, a BRG1/PRC2-independent ISG which is responsive

in SW13 cells, as a control. As an additional control, we used PITX2, a silenced non-ISG.

Importantly, siCHAF1A rescued all of the BRG1/PRC2-regulated ISGs, but had no effect on

IRF1 and PITX2 (Fig 3.4.6). This indicates that CHAF1A is most probably only involved in the

regulation of ISGs regulated by both BRG1 and PRC2.

Page 97: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

82

Fig 3.4.6. CHAF1A KD rescues the induction of multiple PRC2/BRG1dependent ISGs in response to IFN!. A. CHAF1A KD rescues the expression of multiple BRG1/PRC2-dependent ISGs. SW13 cells were transfected with siCHAF1A or siCtrl for 4 days. Cells were then treated with IFN! for 6 hrs. The mRNA level of the indicated BRG1 dependent ISGs, the BRG1 independent ISG IRF1, or the silent non-ISG PITX2 were measured by RT-PCR Results are average of three experiments +/- SD.

Page 98: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

83

3.4.7 CHAF1A is required for PRC2-mediated chromatin modification

CHAF1A is involved in replication-dependent chromatin assembly [313, 325]. CHAF1A also

maintains the heterochromatin epigenetic modification H3K9m3 at heterochromatin during

replication through interactions with the histone H3K9-methyltransferase SetDB1,

heterochromatin protein 1 (HP1), ensure proper transfer of the heterochromatic epigenetic

modifications during DNA replication [145, 223, 225, 326]. However, CHAF1A has not been

connected previously to PRC2 and H3K27me3 deposition. As mentioned above, previous work

in our lab showed that PRC2 represses CIITA and components of the PRC2 complex, such as

EZH2 or SUZ12, in addition to the PRC2-mediated silencing mark, H3K27me3, were detected

across the CIITA locus (unpublished data). Thus, it is possible that CHAF1A may be involved in

maintaining the epigenetic repressive mark H3K27me3 at PRC2-repressed genes. To investigate

this possibility we used ChIP to compare H3K27me3 levels across CIITA in siCtrl or siCHAF1A

treated SW13 cells. Also, since CHAF1A is involved in H3-H4 tetramer deposition, we tested

whether histone H3 deposition is disrupted by siCHAF1A. SW13 cells were transfected with

two different CHAF1A siRNAs (#1 or #2) or siCtrl for four days. Western analysis showed that

each siRNA reduced CHAF1A protein level by > 50% compared to siCtrl (Fig 3.4.7A). The

ChIP for H3 did not indicate any change across the CIITA locus in siCtrl or siCHAF1A-treated

SW13 cells (Fig 3.4.7 B). However, the H3K27me3 mark was markedly reduced across the

CIITA locus after CHAF1A knock down (Fig 3.4.7C).

Next we investigated whether siCHAF1A reduces the H3K27me3 mark at the promoters

of other PRC2-dependent ISGs. Significant levels of H3K27me3 were detected at the GBP3 and

GBP4 promoters, and siCHAF1A reduced the mark by ~50% (Fig 3.4.7D). IRF1, a

PRC2/BRG1 independent ISG served as a negative control (Fig 3.4.7E). These data suggest that

CHAF1A is required for PRC2 dependent H3K27 methylation at PRC2/BRG1 dependent ISGs,

providing a mechanism to explain why siCHAF1A rescues IFNg responsiveness at such loci.

Page 99: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

84

Fig 3.4.7. CHAF1A KD reduces H3K27me3, a PRC2 mediated histone modification at PRC2/BRG1 regulated ISGs. A. Both CHAF1A siRNAs (siCHAF1A#1 and siCHAF1A#2 reduced CHAF1A protein level by more than 80%. SW13 cells were transfected with 2 different siCHAF1A, either with siCHAF1A#1 or siCHAF1A#2 or siCtrl for 4 days. Cell lysates of siCHAF1A and siCtrl cells were used for Western Blotting analysis. B. ChIP showed no disruption in H3 deposition in CHAF1A KD across the CIITA locus. Chromatin lysates of SW13 cells, either transfected with siCHAF1A#1 and siCHAF1A#2 or siCtrl, were used for ChIP, using H3 antibody, followed by RT-qPCR. RT-qPCR results examining the remote elements across CIITA showed no changes in H3 deposition in CHAF1A KD compared to the control cells. C. SW13 cells transfected with either siCHAF1A#1 or siCHAF1A#2 showed significant reduction in H3K27me3 compared to the siCtrl-treated cells across CIITA. D. CHAF1A is required for the deposition of H3K27me3 at the promoters of PRC2/BRG1 dependent ISGs. SW13 cells were transfected with siCHAF1A#1 or siCtrl for 4 days. ChIP experiment was performed using an H3K27me3 antibody for siCHAF1A and siCtrl chromatin lysate. ChIP-qPCR analysis confirmed a reduction of PRC2 chromatin mark, H3K27me3 at the promoters of BRG1/PRC2regulated ISGs. Results are average of three experiments +/- SD. Asterisks indicate a significant reduction in H3K27me3 mark (p<0.05).

Page 100: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

85

3.4.8 General role for CHAF1A in promoting PRC2-mediated gene silencing

We next asked whether CHAF1A represses PRC2-regulated genes that are not ISGs. We first

examined the expression of a known PRC2 non-ISG target gene, E-cadherin [13, 327, 328].

Interestingly, siCHAF1A elevated the basal level of E-cadherin (Fig 3.4.8A) and ChIP analysis

revealed reduced levels of H3K27me3 at the promoter (Fig 3.4.8B).

Mohamed Abou El Hassan in our lab identified multiple PRC2 repressed non-ISGs

through RNA-seq analysis following siSUZ12 treatment of SW13 cells (unpublished data). In

preliminary analysis (n = 1) of six of these loci, we found that siCHAF1A also induced the basal

levels of these targets (Fig 3.4.8C). Altogether, these data suggest that the collaboration

between CHAF1A and PRC2 in gene silencing may be of broad relevance.

Page 101: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

86

Fig 3.4.8 Requirement of CHAF1A in PRC2-mediated gene silencing is a general effect. A. siCHAF1A induces the basal levels of E-Cadherin, a BRG1/PRC2-regulated gene. SW13 cells were transfected with siCHAF1A or siCtrl for 4 days and E-Cadherin levels assessed by RT-qPCR. B. siCHAF1A reduces H3K27me3 levels at the E-Cadherin promoter. H3K27me3 ChIP analysis was performed on chromatin from the cells treated as in (A). . Asterisks indicate a significant reduction in H3K27me3 mark. (p<0.05 C. siCHAF1A rescues PRC2 regulated non-ISGs. SW13 cells were transfected with siCHAF1A and siCtrl for 4 days and mRNA levels of selected PRC2 regulated genes were assessed by RT-qPCR . CDH 2 was used as a negative control (n=1))

Page 102: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

87

3.4.9 CHAF1A-mediated repression requires S-phase

CHAF1A might promote PRC2 function during DNA replication, ensuring that newly deposited

nucleosomes acquire the H3K27me3 mark. Alternatively, CHAF1A might regulate PRC2

activity independent of its role as a replication fork protein. To examine these alternate

possibilities, we asked whether the effects of siCHAF1A on BRG1/PRC2 regulated ISGs and

non-ISGs required DNA replication. In this experiment, SW13 cells were either allowed to

divide or arrested at G1/S by aphidicolin before transfection with siCtrl or siCHAF1A. On day 4

of the experiment RNA was collected to assess gene expression, and in a parallel set of plates

aphidicolin was washed off to release cells into the cell cycle, following which RNA levels were

assessed on day 6. Cell cycle arrest was confirmed by flow cytometry (data not shown).

Western blotting analysis showed that knock down was efficient in dividing, arrested, and

arrested-then-released cells (data not shown). Strikingly, however, CIITA, a PRC2/BRG1

dependent ISGs, and E-cadherin, a PRC2 dependent non-ISG remained silent in arrested cells,

and gene induction was only observed when cells were allowed to divide from day 4 to day 6

(Fig 3.4.9 A,B). The PRC2/BRG1 independent genes IRF1 and TBP were used as negative

controls (Fig 3.4.9C,D). These data indicate that CHAF1A-mediated repression of PRC2-

regulated genes requires active cell division and likely occurs through a replication-dependent

epigenetic mechanism.

The large error bar of E-cadherin expression at day four, Fig 3.4.9A and CIITA expression at

day 6 of Fig 3.4.9B may be the result of two factors: either the knock down is not efficient

enough, or the cells have not gone through enough cell cycle to show the efficient reduction of

H3K27me3. Indeed, I observed that if cells are allowed to divide longer better gene induction is

detected, with less variability.

Page 103: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

88

Fig 3.4.9 The rescue of PRC2-regulated genes after CHAF1A KD requires cell cycle progression. SW13 cells were treated with or without aphidicolin (AP) overnight to arrest S phase before transfecting with siCHAF1A or siCtrl for 4 days. Cells were collected and mRNA was extracted to perform RT-qPCR. GBP2, IFITM3 and CIITA are BRG1-dependent ISGs that are rescued by siCHAF1A in the absence of AP, rescue was reversed in the presence of AP. IRF1, a BRG1-independent ISG which is not induced by siCHAF1A and TBP, a housekeeping gene, were used as a negative controls. Results are the average of two independent experiments +/-SD.

Page 104: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

89

3.4.10 SiCHAF1A causes a dramatic redistribution of H3K27me3 without changing PRC2 localization

We showed that siCHAF1A reduces the PRC2-mediated histone H3K27me3 mark at the

regulatory elements of PRC2 target genes (Fig 3.4.7C). One possibility to explain this result

would be that siCHAF1A reduces the protein level of PRC2 components or total level of

H3K27me3. Interestingly, Western blot analysis did not show any change in the total level of

H3K27me3 or PRC2 components (Fig 3.4.10A). One way to explain this result is that,

siCHAF1A reduces the chromatin bound H3K27me3 and not the total protein. However,

fractionation of lysates revealed no change in the chromatin bound H3K27me3 in siCHAF1A

treated cells (data not shown). These results show there must be another mechanism through

which siCHAF1A reduces the PRC2-mediated histone marks at specific targets.

One possibility is that the effect of siCHAF1A on PRC2 targets is to affect the

recruitment of PRC2 to replication forks and/or the activity of PRC2 at forks, thus affecting the

distribution of H3K27me3. Immunostaining suggested that siCHAF1A did not affect the

recruitment of PRC2 to replication forks (Fig 3.4.10B). Thus, CHAF1A is not physically

involved in the recruitment or maintenance of PRC2 at sites of replication. However,

Immunostaining revealed a striking redistribution H3K27me3 to the nuclear periphery (Fig

3.4.10 C). Thus, while CHAF1A does not affect bulk levels of H3K27me3 (Fig3.4.10A), it is

critical for the appropriate distribution of H3K27me3, suggesting that it may regulate PRC2

activity at the fork (Fig 3.4.10 C).

Page 105: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

90

Page 106: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

91

Fig 3.4.10 CHAF1A KD does not change PRC2 protein level and localization but it alters H3K27me3 distribution. A. CHAF1A KD has no effect on PRC2 components protein levels or relevant chromatin modification. Western Blot was performed after 4 days of transfecting SW13 cells with siCHAF1A or siCtrl. The protein levels of H3K27me3, H3, EZH2, SUZI12 were not affected by CHAF1A KD. Results are average of three experiments +/- SD. B. siCHAF1A does not block PRC2 recruitment to replication forks. SW13 cells were transfected with siCHAF1A or siCtrl for 4 days before being reseeded onto coverslips for immunostaining for EZH to mark PRC2 location or PCNA to mark replication forks. Co-localization at the fork was unaffected in the absence of CHAF1A. C. CHAF1A KD causes a dramatic redistribution of H3K27me3. Cells from (A) were collected at day four and seeded onto cover slips. They were stained the next day with a H3K27me3 antibody. Gray scale images shown were overlaid images of five 0.4 um thick z-stack images. Surface plot of H3K27me3 of cells in the gray scale images are shown on the left.

Page 107: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

92

3.5 Discussion

3.5.1 A novel direct link between CHAF1 and PRC2-mediated gene repression during replication

We discovered an unexpected and interesting link between CHAF1A, the large subunit of CAF-1

histone chaperone and PRC2-mediated gene regulation in the human adrenal carcinoma SW13

cell line. Depleting CHAF1A rescued PRC2-dependent repression of both ISGs and non-ISGs in

these cells. Interestingly this rescue is replication-dependent as it does not occur when the cell

cycle is blocked. Since CHAF1A deposits H3 during replication, CHAF1A is most likely

needed to transfer the PRC2-mediated silencing mark during this time.

Generally, in mammals there are two major H3 epigenetic silencing marks which define

the silent status of genes: SUV39-mediated H3K9me3 and the PRC2-mediated H3K27me3.

During replication the nucleosomes are disassembled in front of replication forks and then

reassembled, now including newly synthesized histones. It is important that the epigenetic

modification of the parental histones is copied to the new ones.

Very little is known about the mechanism of how this occurs, however deregulation of this step

has been linked to several diseases including cancer. Recent data reinforce the idea that

CHAF1A is likely a key player in this process [329, 330].

Most studies of CHAF1A and epigenetic memory focus on the effect of CHAF1A on

H3K9me3 silencing mark. For example, CHAF1A maintains of this mark at telomeres and yeast

mating type loci [146, 147, 177, 216, 331]. Also, CHAF1A has been identified as a

transcriptional repressor in the screen for silencing factors in human cells and the 5$ truncated

form of this protein impairs the maintenance of transcriptional gene silencing [143, 147, 326].

The mechanism through which CHAF1A maintains the H3K9me3 mark involves the

heterochromatic protein HP1#, MBD1 and, SETDB1, a histone H3K9 methyltransferase during

S- phase [145, 223-225].

In contrast to H3K9me3, the mechanism of H3K27me3 maintenance is not well

understood. Here we show that CHAF1A also maintains this PRC2-mediated epigenetic mark.

Page 108: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

93

Interestingly, the only other occasion when a link between CHAF1A and PRC2 has been

suggested is a study showing that a Drosophila CHAF1A (P180) mutant disrupts PRC2-

mediated silencing in developmentally important genes [216]. However, this study did not

address the functional significance of this observation. Therefore, our study shows for the first

time the functional link between CHAF1A and PRC2-dependent gene silencing in mammalian

cells.

3.5.2 CHAF1A is required for printing the H3K27me3 mark at the proper targets

To dissect the mechanism through which CHAF1A maintains the H3K27me3 mark, we first

wanted to know if siCHAF1A reduces the protein levels of PRC2 components. However this

was not the case. Moreover, removing CHAF1A did not affect the bulk levels of H3K27me3 on

chromatin. Importantly, we found that siCHAF1A KD resulted in redistribution of the

H3K27me3 mark from a central to peripheral nuclear location. This striking observation

suggests that CHAF1A is required for the printing of H3K27me3 by PRC2 at the appropriate

targets. To find out how CHAF1A is involved in maintaining H3K27me3 during S-phase, we

need to understand how PRC2 is recruited to its targets and what affects its activity.

There are two proposed models for PcGs recruitment and function. The first one is the

“instructive model” which is based on direct interaction of PRC2 with sequence specific TFs or

non-coding RNAs [332-335]. The major challenge for this model is that it does not explain how

PRC2 binds multiple diverse non-coding RNAs or TFs, which is required to define gene

expression patterns during development [336]. However, recently an alternative “responsive”

model for PRC2 function has been proposed [336]. This “responsive” model is derived from the

preferential binding and activity of PcG at hypomethylated CpG islands (CGIs) in vertebrates

[337, 338]. In this model, both PcG and TrxG complexes constantly sample CGIs-containing

chromatin sites. The chromatin conformation created by CGIs, in combination with “silencing”

or “anti-silencing” factors, favors the recruitment of either PcG or TrxG [336]. For example,

H3K4me3, H3K36me3, RNA polymerase II or H3K27ac, block PRC2 activity or recruitment

while they promote TrxG activity or recruitment and poise genes for activation [76, 336, 339].

Page 109: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

94

In the absence of these “anti-silencing” factors, PRC2 is recruited to CGIs and through

positive feedback processes accumulates H3K27me3 mark at the PRC2 targets. For example,

H3K27me3 recruits the PRC1 complex which would compact chromatin, and it has been shown

that PRC2 prefers to methylate H3K27 when the chromatin is compact [340]. Finally binding of

PRC2 complexes to their targets and accumulation of H3K27me3 transfers the status of the

targeted sites from “sample” to “established” state [336, 340, 341].

Based on the “responsive” model for PRC2 recruitment and function, it is possible that CHAF1A

regulates some or all of the chromatin features that regulate PRC2 recruitment and/or activity.

For example, since DNA methylation inhibits PRC2 (see above) it is possible that a CHAF1A-

containing complex at the replication fork ensures that CGIs which are PRC2 targets remain

unmethylated. In agreement with this notion, it has been shown that the DNA methylase

DNMT1 interacts with PCNA [146, 147, 194, 342]. DNMT1 is required for the maintenance of

DNA methylation during replication [58, 343]. This is intriguing, because our siRNA

Suppressor screen aimed to identify novel factors associated with the silencing of CIITA, aside

from CHAF1A also turned up PCNA. Several studies showed that CHAF1A and PCNA interact

at the replication fork and that PCNA is required for epigenetic inheritance during replication

[146, 147, 194]. Therefore, we hypothesize that PCNA may also be involved in the appropriate

targeting of H3K27me3 by CHAF1A during replication through DNA methylation.

We observed that in the absence of CHAF1A, H3K27me3 is re-distributed from a primarily

central (euchromatin) to a peripheral (heterochromatin) nuclear location. Other work in the lab

using electron spectroscopic tomography revealed that siCHAF1A does not affect the location of

heterochromatin itself (E. Fussner), arguing that the chromatin mark has not moved to the

chromatin subtype. In the absence of CHAF1A it is possible that another histone chaperone is

recruited to the replication fork to substitute for CHAF1A. However this replacement complex

might not faithfully transfer the pattern of DNA methylation or chromatin modifications from

parental DNA to the daughter strands during replication. Perhaps during early S-phase, when

euchromatic regions lacking 5mC are replicated, the replacement histone chaperone recruits

DNMT1, or one of the other members of this family (e.g. DNMT3), which aberrantly methylates

DNA sequences, including previously hypomethylated CGIs which are normally targeted by

PRC2. This aberrant DNA methylation would block PRC2 activity, and after two cell cycles

would result in a dramatic increase in 5mC, concomitant inhibition of PRC2 and loss of

Page 110: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

95

H3K27me3. Equally, the absence of CHAF1A during replication of heterochromatin could lead

to a deficit in 5mC maintenance, which could permit the spread of H3K27me3 mark into these

regions. Since we observe accumulation of H3K27me3 at the nuclear periphery (i.e. at

heterochromatin), it is possible that during replication the absence of CHAF1A-driven DNA

methylation leads to redistribution of H3K27me3. That is, the H3K27me3 mark is gained where

the DNA methylation is lost [108]. This is consistent with recently published studies indicating

that the pattern of H3K27me3 distribution correlates with hypomethylated regions [108].

Indeed, when DNA methylation is absent, e.g. following DNMT1 deletion, depletion or drug-

mediated inhibition, H3K27me3 redistributes such that it is depleted at formerly enriched areas

and elevated at formerly H3K27me3 poor regions [108]. This may be akin to the phenomenon

we observe in siCHA1A treated cells.

Interestingly, another piece of evidence implicating DNA methylation in the mechanism of

CHAF1A action comes from the recent preliminary data obtained by Tom Leung in our lab. He

found that siCHAF1A causes redistribution of DNA methylation in the opposite way of

H3K27me3 (from peripheral to central part of nuclei). Notably, total levels of 5mC were

unchanged, implying that there is no loss of either H3K27me3 or 5mC, but an inversion in their

localization. While it is established that 5mC repels PRC2/H3K27me3, it remains unclear

whether the reverse is also true. These data require extensive future genomic analysis of the

redistribution events, including kinetic studies to define when they occur relative to each other,

and parallel assessment of the location of the enzyme complexes involved.

Another possibility is that the epigenetic modification such as H3K4me3, H3K36me3 and

H3K27ac that are known to block PRC2 function [336] are aberrantly accumulated to PRC2

target sites by an unknown histone chaperone which functions in the absence of CHAF1A. To

test if this is the case, we can investigate whether any of these modifications have been increased

at PRC2 target sites in siCHAF1A treated cells. Finally, we note that CHAF1A can bind to

sumoylating enzymes [344], and PRC2 activity is modified by sumoylation [345], thus it will be

of interest to determine whether this and/or other post transcriptional modifications of PRC2 are

affected by CHAF1A depletion.

In summary, our data indicate that CHAF1A regulates the precise pattern of H3K27me3

distribution. The striking redistribution of this mark when CHAF1 is absent may be connected

Page 111: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

96

to an inverse effect on the localization of 5mC, although considerable work is required to test

this or alternate hypotheses.

Page 112: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

97

4 Discussion and Future Directions

Page 113: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

98

4.1 Discussion

4.1.1 CHAF1A maintains H3K27me3 during replication

The expression status of many genes is decided during development and for a large number of

them is maintained by the collaboration of PcG and TrxG groups of proteins [23]. These two

groups of proteins have antagonistic effects and mediate the epigenetic marks H3K27me3 and

H3K4me3 which correlate with gene repression and gene activation respectively. There is a

good understanding of how PcGs or TrxGs regulate gene expression and how they are recruited

to the targeted genes. However, the mechanism explaining how the epigenetic marks are

maintained during replication is lacking. In this study we describe a novel link between

CHAF1A, the large subunit of the CAF-1 histone chaperone complex, and the PRC2-mediated

deposition of the repressive epigenetic modification, H3K27me3.

During DNA replication, histones are disassembled in front of the replication fork and

reassembled behind it. It is not entirely clear how the epigenetic information on histones is

faithfully transferred to the daughter protein, and thus how the expression status of any given

gene is kept silent or active during replication.

There is already some evidence implicating the role of CHAF1A in the maintenance of

H3K9me3 in the heterochromatin regions. It has been proposed that CHAF1A, together with

PCNA, recruit histone methylases such as SUV39 and SETDB1, and DNA methylases to

maintain epigenetic memory [225] . However, how the H3K27me3 silencing epigenetic

modifications are transferred during replication, is not clear. Here we provide evidence that

indeed CHAF1A is a key player in this process.

Previously, our lab showed that in the BRG1-deficient adrenal carcinoma SW13 cells,

CIITA, the master regulator of MHC-Class II, is repressed by PRC2 and H3K27me3 was

detected cross the CIITA locus (M Abou El Hassan, unpublished data). We performed an RNAi

screen to uncover additional factors which together with PRC2 participate in the silencing of

CIITA. We validated two interesting hits which interact with each other and are both involved in

DNA replication, CHAF1A and PCNA. We observed that the KD of either of these genes

rescued the response of CIITA to IFN! in SW13 cells. In agreement, we found that siCHAF1A

reduces the H3K27me3 repressive marks across the CIITA locus. Further, we showed that other

Page 114: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

99

PRC2-regulated ISGs were also responsive to IFN! after CHAF1A depletion, and the

H3K27me3 mark was reduced at the promoters of these genes. Interestingly, we showed that

this effect is not limited to ISGs and that other PRC2-regulated non-ISGs, such as E-cadherin,

were also induced after siCHAF1A and the H3K27me3 mark was reduced.

Importantly, we observed that the rescue of the tested genes after CHAF1A KD is cell

cycle dependent. Since CHAF1A is known to deposit the histone H3 to the newly synthesized

DNA during replication, we believe that CHAF1A is likely a crucial factor maintaining the

H3K27 trimethylation on newly synthesized histones.

4.1.2 CHAF1A may guide the H3K27me3 deposition indirectly

How does CHAF1A guide the deposition of the PRC2 mark H3K27me3? First, CHAF1A may

instruct PRC2 indirectly by means of DNA methylation. Indeed, it has been shown in

mammalian cells that the status of DNA methylation on the CpG islands inversely regulates the

establishment of H3K27me3 by PRC2 [51, 52, 106, 108]. The hypo-methylated CpG islands

through an unknown mechanism promote H3K27 methylation [108]. Preliminary data in our lab

supports this notion as T Leung observed by Immunostaining that H3K27me3 and 5mC show

opposite patterns of distribution in the nucleus, and that siCHAF1A inverts these patterns

(unpublished data). How could this work? Several candidates might be involved in this process.

For example, TET1 is a DNA demethylase required for the recruitment of PRC2 to its targets in

mouse embryonic stem cells [346]. Interestingly, TET1 binds both PCNA and EZH2 [62].

Further, links between PRC2 and the DNMT family members such as DNMT1 and DNMT3a

have been recently described [58, 347]. DNMT1 and PCNA also interact at replication fork [58,

61]. Since CHAF1A also interacts with PCNA, cross-talk among CHAF1A, PCNA and DNA

methylases at the replication fork seems likely. Thus, we envisage that in response to

siCHAF1A, the loss of DNA methylation at the nuclear periphery (heterochromatin ) would

result in the spread of PRC2 marks into these sites, and the concomitant dilution of H3K27me3

at canonical PRC2-targeted regions, central parts of nuclei (euchromatin), resulting in elevated

expression/responsiveness of previously repressed loci; indeed, this is exactly what is observed

Page 115: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

100

when DNA methylation is inhibited in mouse embryonic fibroblasts due to expression of a

hypomorphic DNMT1 allele, knockdown of DNMT1, or treatment with 5-aza-cytidine, a DNMT

inhibitor, and induction of PRC2 targets was also observed in murine embryos expressing an

unstable DNMT1 allele [108].

Aberrant DNA and histone methylation has been reported in many types of cancer. There

are CHAF1A variants listed in the Catalogue of Somatic Mutations in Cancer (COSMIC)

database (http://cancer.sanger.ac.uk/cancergenome/projects/cosmic/), although the frequency is

low in each cancer and the functional relevance, if any, is unclear. It is possible that the activity

of CHAF1A may be compromised in cancer cells in ways other than direct mutation. It is

intriguing to speculate that the aberrant distribution of DNA methylation observed in many

cancers may be the consequence of the lack of CHAF1A activity, although this remains to be

tested.

Another possibility is that CHAF1A is required for PRC2 recruitment to the proper sites.

There are two proposed models for PcGs recruitment. In the first model the “instructive model”

PcG directly interacts with sequence specific TFs or non-coding RNAs [332-335]. For example

it has been shown that non-coding RNAs are required for the recruitment PRC2 in PRC2-

mediated X-inactivation [348, 349]. Therefore, identifying the factors that recruit non-coding

RNAs to their targets may provide additional clues about CHAF1A activity in the epigenetic

memory maintenance.

The second model which recently has been proposed is “responsive” [336]. This

“responsive” model is based on observation that PcG preferentially binds at hypomethylated

CpG islands (CGIs) in vertebrates [337, 338]. This model focuses on the effect of chromatin

conformation and modification created by combination of CGIs and histone modification on PcG

and TrxG complexes recruitment. In this model both PcG and TrxG sample CGIs-containing

chromatin sites. If the chromatin conformation contains “anti-silencing factors” such as

H3K4me3, H3K36me3, RNA polymerase II or H3K27ac, this blocks PRC2 activity or

recruitment while promoting TrxG activity or recruitment, poising genes for activation [76, 336,

339]. In contrast, in the absence of “anti-silencing” factors, PRC1 and PRC2 are recruited to the

chromatin and mediate histone modifications which eventually induce gene silencing [336].

Therefore, it is possible that in the absence of CHAF1A the chromatin conformation is changed

Page 116: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

101

by aberrant DNA methylation or recruitment of these “anti-silencing” factors which block PRC2

recruitment.

4.1.3 CHAF1A may activate PRC2

Conceivably, CHAF1A may activate PRC2. It has been recently reported that PRC2 remains

associated with DNA during replication [111]. Thus, it is possible that during DNA replication,

CHAF1A activates PRC2. Intriguingly, there is already evidence in the literature supporting this

hypothesis.

It has been shown that PRC2 components, SUZ12 and EZH2 can be SUMOylated, which

promotes PRC2 activation [345]. As mentioned above, CHAF1A recruits SUMO2/3 enzymes

which SUMOylate proteins at replication forks [344]. Thus, it is possible that SUMO2/3

recruited by CHAF1A, activate PRC2. According to this model, CHAF1A would be required for

the direct enzymatic activation of PRC2.

Taken together, it is possible that CHAF1A guides the PRC2 activity not in one way but

by the combination of direct and indirect interactions. Further, many other factors are likely to

be involved. In the next section we describe the approaches that will help to elucidate the

mechanism of CHAF1A action and fill in some of the gaps in our understanding of how this

factor orchestrates the epigenetic memory maintenance.

Page 117: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

102

4.2 Future directions

There are still many gaps in our understanding of the role of CHAF1A in PRC2-mediated gene

repression and the maintenance of the repressive PRC2 signature mark, H3K27me3, during DNA

replication. Here we discuss some of the high-priority experiments designed to address these

issues.

4.2.1 The global effect of depleting CHAF1A on PRC2 regulated genes and redistribution of H3K27me3

So far, we only focused on the regulation of CIITA and a few other PRC2-regulated genes. We

would like to further investigate the importance of CHAF1A in gene regulation of PRC2 targets,

in general.

To investigate the PRC2 gene targets in SW13 cells, our lab has previously performed

RNA sequencing study (RNA-seq) following depletion of the PRC2 core component SUZ12 (M.

About El Hassan, unpublished data). Next, we would like to perform RNA-seq in siCHAF1A

treated cells. Comparing the results of these two RNA-seq experiments will reveal how many

genes are targeted by both CHAF1A and PRC2. A high percentage of target overlap will

strengthen the position of CHAF1A in the regulation of PRC2-target genes. On the other hand a

low percentage of overlap will indicate that only select genes require the co-operation of

CHAF1A and PRC2.

By immunofluorescence (IF) staining we showed that siCHAF1A does not affect global

localization of PRC2 (T Leung, unpublished data). However since the resolution of IF is limited,

we will perform chromatin immunoprecipitation (ChIP) with massively parallel sequencing

(ChIP-Seq) for PRC2 components (e.g., SUZ12 and EZH2) and H3K27me3, either in the

presence or absence of CHAF1A to ascertain whether the recruitment of PRC2 is affected by the

loss of CHAF1A.

4.2.2 DNA methylation and histone modification

We showed that siCHAF1A causes the redistribution of H3K27me3 in SW13 cells. Further, we

also have preliminary data showing that the redistribution of DNA methylation in siCHAF1A

Page 118: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

103

treated cells occurs in the opposite way of H3K27me (T. Leung, unpublished data). To further

study this phenomenon, we will deplete CHAF1A and perform ChIP-Seq for H3K27me3 to

identify the global targets of this mark. Also, we will perform methylated DNA

immunoprecipitation (MeDIP-seq) to identify the DNA methylated regions following

siCHAF1A. The data from these two experiments will clarify two issues: 1. whether there is an

overlap between the reduction of H3K27me3 at the regulatory elements of the genes regulated by

both CHAF1A and PRC2. 2. Whether and to what extent there is a correlation between loss and

gain of DNA methylation and H3K27me3 across the genome.

If we show that indeed there is a correlation between the loss of DNA methylation and

gain of H3K27me3, and vice versa, this would strengthen our hypothesis that CHAF1A has a

global role in coordinating the distribution of these epigenetic marks. The next step, then, will

be to uncover the mechanism linking the observed DNA methylation and histone methylation

redistribution.

Another possibility is that the epigenetic modification such as H3K4me3, H3K36me3 and

H3K27ac that are known to block PRC2 function [336] are aberrantly accumulated to PRC2

target sites by an unknown histone chaperone which functions in the absence of CHAF1A. To

test if this is the case, we will perform IF and investigate whether any of these histone

modifications have been accumulated at PRC2 target sites in siCHAF1A treated cells.

4.2.3 Identifying the components involved in the CHAF1A-dependent maintenance of H3K27me3

We showed that siCHAF1A leads to H3K27me3 redistribution in a DNA replication manner and

T. Leung in our lab has preliminary data suggesting that DNA methylation is also redistributed in

the opposite way. The next step will be to identify the protein complexes or components

involved.

It has been shown that the DNA methylase DNMT1 and the DNA demethylase TET1

interact with PCNA [58, 62, 343]. Since CHAF1A binds PCNA at the replication forks, it is

likely that either these two enzymes or other members of their families collaborate with

CHAF1A in the epigenetic mark maintenance. To test whether this is the case, we will

immunostain for DNMT1, TET1 and other members of these protein families to examine how

Page 119: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

104

their staining pattern is affected by CHAF1A KD. If the staining pattern is affected, this would

argue that CHAF1A affects their recruitment to the fork. However, it may be possible that

CHAF1A is required not for the localization of the enzymes but for instructing the enzymes

where to lay down the epigenetic mark.

We also observed that CHAF1A KD rescues E-cadherin, which is also silenced in SW13

cells by PRC2. E-cadherin is a crucial factor in Epithelial-mesenchymal transitions (EMT), and

is repressed in several tumor cells, such as breast cancer [327, 350, 351]. Since it has been

shown that repression of E-cadherin is linked to metastasis, many studies attempted to identify

its epigenetic regulators. For example, the transcription factor Snail recruits various epigenetic

modifiers to E-cadherin promoters such as LSD1/HDAC [13]. Other histone modifying enzymes

such as G9a/SUV39H are also found at the E-cadherin promoter and in turn recruit DNMTs [13].

Thus, it would be important to investigate the distribution of these factors after CHAF1A KD,

first by immunostaining and then at specific loci using ChIP. This may indicate whether any of

these factors co-operate with CHAF1A.

Last but not least, recent studies showed that ZF-CxxC domain-containing proteins, such

as CFP1 (CxxC finger protein 1), MLL (mixed lineage leukaemia protein), KDM (lysine

demethylase) 2A and KDM2B (Histone-H3-Lysine-36 Demethylase B) bind hypomethylated

DNA and recruit PRC2 to the CpG islands [352]. It will be important, using immunostaining

and ChIP, to determine whether these proteins interact with CHAF1 and are involved in its

activities.

4.2.4 Structure-Function analyses of CHAF1A

CHAF1A contains several domains that are required for its different functions. For example the

N-terminal domain binds PCNA and HP1#, proteins required for gene silencing and maintaining

heterochromatin regions [144, 145, 194]. The C-terminal domain binds CHAF1B, another sub-

unit of the CAF-I complex, and this is required for H3 deposition during replication[325, 353].

CHAF1A N- and C-terminal deletion mutants would be a useful starting point to define

which regions of CHAF1A are essential for the regulation of H3K27me3 distribution.

Expressing CHAF1A vectors in siCHAF1A treated cells coupled with RT-PCR analysis of PRC2

target loci (e.g. E cadherin or CIITA) would reveal which region of the protein is critical to

Page 120: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

105

promote PRC2 action. These assays would utilize a siCHAF1A that targets the 3" UTR of the

mRNA, missing in the engineered vectors, so that only the endogenous gene would be depleted.

Fine mapping would be used to define point mutations in a domain that disrupts the repressive

function. IP for the wild type and mutated forms of CHAF1A followed by Mass spectrometry

(Mass Spec) would be used to identify the factors that are missing in the mutated complex.

Additional functional studies will be needed to test the significance of these novel factors at the

replication forks. For example, it would be important to use RNAi or dominant negative

strategies to define whether interfering with the function of any of these CHAF1A integrators

alleviates repression of PRC2 target genes or affects DNA methylation.

4.2.5 Using aniPOND to screen for additional factors co-operating with CHAF1A at the replication forks

Isolation of Protein on Nascent DNA (iPOND) is a technique, which is used to purify the

replication fork components for analysis by Western blotting or Mass Spec [354, 355]. Our lab

has recently developed a variation of this technique: accelerated native iPOND (aniPOND),

which improves sensitivity [356]. It will thus be of considerable interest to use aniPOND to

purify replication forks in siCtrl or siCHAF1A treated cells and assess the protein components by

Mass Spec. The factors missing in the protein complexes after siCHAF1A may be the critical

factors that influence PRC2 function and/or regulate DNA methylation. Additional functional

studies will then be need to validate whether and how these factors are required to maintain

PRC2-mediated marks.

4.3 Concluding remarks

Although the mechanism of DNA replication has been thoroughly studied, how the epigenetic

status of chromatin is inherited during this process is not completely understood. DNA

replication occurs rapidly, which makes it difficult to study the individual steps of nucleosome

duplication. H3K27me3 maintains the status of repressed genes and this is decided early in

development. It is very crucial to ensure that during replication the information dictated by

PRC2 is transferred faithfully to the newly synthesized DNA. Here, I provided strong evidence

that CHAF1A is a key factor in this process in mammalian cells. I showed that this protein is

Page 121: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

106

required for the distribution of H3K27me3 to their proper targets. We also have preliminary

evidence that this process may be linked to DNA methylation.

Since, aberrant DNA and histone methylation are seen in many diseases, including

cancer, dissecting the mechanism and identifying the factors required for the proper maintenance

of these epigenetic marks is crucial. This knowledge will both deepen our understanding of this

process and may lead to improved therapies.

Page 122: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

107

References

1. Yu, L.W. and Y.H. Gui, [Post-transcriptional protein modification of Gata4]. Zhongguo Dang Dai Er Ke Za Zhi, 2012. 14(10): p. 800-3.

2. Chu, S., Transcriptional regulation by post-transcriptional modification--role of phosphorylation in Sp1 transcriptional activity. Gene, 2012. 508(1): p. 1-8.

3. Lande-Diner, L. and H. Cedar, Silence of the genes--mechanisms of long-term repression. Nat Rev Genet, 2005. 6(8): p. 648-54.

4. Lund, A.H. and M. van Lohuizen, Polycomb complexes and silencing mechanisms. Curr Opin Cell Biol, 2004. 16(3): p. 239-46.

5. Schoenherr, C.J. and D.J. Anderson, The neuron-restrictive silencer factor (NRSF): a coordinate repressor of multiple neuron-specific genes. Science, 1995. 267(5202): p. 1360-3.

6. Varelas, X., et al., The Crumbs complex couples cell density sensing to Hippo-dependent control of the TGF-beta-SMAD pathway. Dev Cell, 2010. 19(6): p. 831-44.

7. Armstrong, J.A., J.J. Bieker, and B.M. Emerson, A SWI/SNF-related chromatin remodeling complex, E-RC1, is required for tissue-specific transcriptional regulation by EKLF in vitro. Cell, 1998. 95(1): p. 93-104.

8. Breiling, A., et al., General transcription factors bind promoters repressed by Polycomb group proteins. Nature, 2001. 412(6847): p. 651-5.

9. Istrail, S. and E.H. Davidson, Logic functions of the genomic cis-regulatory code. Proc Natl Acad Sci U S A, 2005. 102(14): p. 4954-9.

10. Maston, G.A., S.K. Evans, and M.R. Green, Transcriptional regulatory elements in the human genome. Annu Rev Genomics Hum Genet, 2006. 7: p. 29-59.

11. Londono, A., L. Riego-Ruiz, and G.R. Arguello-Astorga, DNA-binding specificity determinants of replication proteins encoded by eukaryotic ssDNA viruses are adjacent to widely separated RCR conserved motifs. Arch Virol, 2010. 155(7): p. 1033-46.

12. Thackray, V.G., Fox tales: Regulation of gonadotropin gene expression by forkhead transcription factors. Mol Cell Endocrinol, 2013.

13. Herranz, N., et al., Polycomb complex 2 is required for E-cadherin repression by the Snail1 transcription factor. Mol Cell Biol, 2008. 28(15): p. 4772-81.

14. Struhl, K., Helix-turn-helix, zinc-finger, and leucine-zipper motifs for eukaryotic transcriptional regulatory proteins. Trends Biochem Sci, 1989. 14(4): p. 137-40.

Page 123: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

108

15. Pearson, J.C., D. Lemons, and W. McGinnis, Modulating Hox gene functions during animal body patterning. Nat Rev Genet, 2005. 6(12): p. 893-904.

16. Gao, Y., et al., SMAD7 antagonizes key TGFbeta superfamily signaling in mouse granulosa cells in vitro. Reproduction, 2013. 146(1): p. 1-11.

17. Watanabe, M. and M. Whitman, The role of transcription factors involved in TGFbeta superfamily signaling during development. Cell Mol Biol (Noisy-le-grand), 1999. 45(5): p. 537-43.

18. Wrana, J.L., Signaling by the TGFbeta Superfamily. Cold Spring Harb Perspect Biol, 2013. 5(10).

19. Brivanlou, A.H. and J.E. Darnell, Jr., Signal transduction and the control of gene expression. Science, 2002. 295(5556): p. 813-8.

20. Wu, C.Y., et al., Epigenetic reprogramming and post-transcriptional regulation during the epithelial-mesenchymal transition. Trends Genet, 2012. 28(9): p. 454-63.

21. Peinado, H., D. Olmeda, and A. Cano, Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer, 2007. 7(6): p. 415-28.

22. Yang, M.H. and K.J. Wu, TWIST activation by hypoxia inducible factor-1 (HIF-1): implications in metastasis and development. Cell Cycle, 2008. 7(14): p. 2090-6.

23. Lanzuolo, C. and V. Orlando, Memories from the polycomb group proteins. Annu Rev Genet, 2012. 46: p. 561-89.

24. Peinado, H., et al., Snail mediates E-cadherin repression by the recruitment of the Sin3A/histone deacetylase 1 (HDAC1)/HDAC2 complex. Mol Cell Biol, 2004. 24(1): p. 306-19.

25. Chiou, S.K. and E. White, p300 binding by E1A cosegregates with p53 induction but is dispensable for apoptosis. J Virol, 1997. 71(5): p. 3515-25.

26. Crish, J.F. and R.L. Eckert, Synergistic activation of human involucrin gene expression by Fra-1 and p300--evidence for the presence of a multiprotein complex. J Invest Dermatol, 2008. 128(3): p. 530-41.

27. Eckner, R., p300 and CBP as transcriptional regulators and targets of oncogenic events. Biol Chem, 1996. 377(11): p. 685-8.

28. Felzien, L.K., et al., Specificity of cyclin E-Cdk2, TFIIB, and E1A interactions with a common domain of the p300 coactivator. Mol Cell Biol, 1999. 19(6): p. 4241-6.

29. Gingras, S., et al., p300/CBP is required for transcriptional induction by interleukin-4 and interacts with Stat6. Nucleic Acids Res, 1999. 27(13): p. 2722-9.

Page 124: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

109

30. Huang, X., et al., Coronary development is regulated by ATP-dependent SWI/SNF chromatin remodeling component BAF180. Dev Biol, 2008. 319(2): p. 258-66.

31. Katsani, K.R., T. Mahmoudi, and C.P. Verrijzer, Selective gene regulation by SWI/SNF-related chromatin remodeling factors. Curr Top Microbiol Immunol, 2003. 274: p. 113-41.

32. Martin, D.M., Chromatin remodeling in development and disease: focus on CHD7. PLoS Genet, 2010. 6(7): p. e1001010.

33. Pattenden, S.G., et al., Interferon-gamma-induced chromatin remodeling at the CIITA locus is BRG1 dependent. Embo J, 2002. 21(8): p. 1978-1986.

34. Peterson, C.L. and J.W. Tamkun, The SWI-SNF complex: a chromatin remodeling machine? Trends Biochem Sci, 1995. 20(4): p. 143-6.

35. Wu, J.I., Diverse functions of ATP-dependent chromatin remodeling complexes in development and cancer. Acta Biochim Biophys Sin (Shanghai), 2012. 44(1): p. 54-69.

36. Kornberg, R.D. and Y. Lorch, Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome. Cell, 1999. 98(3): p. 285-94.

37. Strauss, F. and A. Prunell, Nucleosome spacing in rat liver chromatin. A study with exonuclease III. Nucleic Acids Res, 1982. 10(7): p. 2275-93.

38. Prunell, A. and R.D. Kornberg, Variable center to center distance of nucleosomes in chromatin. J Mol Biol, 1982. 154(3): p. 515-23.

39. Kornberg, R.D. and Y. Lorch, Chromatin structure and transcription. Annu Rev Cell Biol, 1992. 8: p. 563-87.

40. Kornberg, R.D., Chromatin structure: a repeating unit of histones and DNA. Science, 1974. 184(4139): p. 868-71.

41. Kornberg, R.D. and J.O. Thomas, Chromatin structure; oligomers of the histones. Science, 1974. 184(4139): p. 865-8.

42. Soppe, W.J., et al., DNA methylation controls histone H3 lysine 9 methylation and heterochromatin assembly in Arabidopsis. EMBO J, 2002. 21(23): p. 6549-59.

43. Fransz, P.F. and J.H. de Jong, Chromatin dynamics in plants. Curr Opin Plant Biol, 2002. 5(6): p. 560-7.

44. Fransz, P., et al., Interphase chromosomes in Arabidopsis are organized as well defined chromocenters from which euchromatin loops emanate. Proc Natl Acad Sci U S A, 2002. 99(22): p. 14584-9.

45. STTARR imaging facility UHN resources/services http://www.sttarr.ca/index.php?option=com_content&task=view&id=50&Itemid=73.

Page 125: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

110

46. Narlikar, G.J., H.Y. Fan, and R.E. Kingston, Cooperation between complexes that regulate chromatin structure and transcription. Cell, 2002. 108(4): p. 475-87.

47. Vignali, M., et al., ATP-dependent chromatin-remodeling complexes. Mol Cell Biol, 2000. 20(6): p. 1899-910.

48. Peterson, C.L. and M.A. Laniel, Histones and histone modifications. Curr Biol, 2004. 14(14): p. R546-51.

49. Fischle, W., Y. Wang, and C.D. Allis, Histone and chromatin cross-talk. Curr Opin Cell Biol, 2003. 15(2): p. 172-83.

50. Mohan, K.N. and J.R. Chaillet, Cell and molecular biology of DNA methyltransferase 1. Int Rev Cell Mol Biol, 2013. 306: p. 1-42.

51. Bird, A., The dinucleotide CG as a genomic signalling module. J Mol Biol, 2011. 409(1): p. 47-53.

52. Lister, R., et al., Human DNA methylomes at base resolution show widespread epigenomic differences. Nature, 2009. 462(7271): p. 315-22.

53. Illingworth, R., et al., A novel CpG island set identifies tissue-specific methylation at developmental gene loci. PLoS Biol, 2008. 6(1): p. e22.

54. Baylin, S.B., S.A. Belinsky, and J.G. Herman, Aberrant methylation of gene promoters in cancer---concepts, misconcepts, and promise. J Natl Cancer Inst, 2000. 92(18): p. 1460-1.

55. Robertson, K.D., DNA methylation, methyltransferases, and cancer. Oncogene, 2001. 20(24): p. 3139-55.

56. Lorincz, M.C., D. Schubeler, and M. Groudine, Methylation-mediated proviral silencing is associated with MeCP2 recruitment and localized histone H3 deacetylation. Mol Cell Biol, 2001. 21(23): p. 7913-22.

57. Khorasanizadeh, S., The nucleosome: from genomic organization to genomic regulation. Cell, 2004. 116(2): p. 259-72.

58. Cartron, P.F., et al., HDAC1-mSin3a-NCOR1, Dnmt3b-HDAC1-Egr1 and Dnmt1-PCNA-UHRF1-G9a regulate the NY-ESO1 gene expression. Mol Oncol, 2013. 7(3): p. 452-63.

59. Robertson, K.D., et al., The human DNA methyltransferases (DNMTs) 1, 3a and 3b: coordinate mRNA expression in normal tissues and overexpression in tumors. Nucleic Acids Res, 1999. 27(11): p. 2291-8.

60. Leonhardt, H., et al., A targeting sequence directs DNA methyltransferase to sites of DNA replication in mammalian nuclei. Cell, 1992. 71(5): p. 865-73.

Page 126: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

111

61. Schermelleh, L., et al., Dynamics of Dnmt1 interaction with the replication machinery and its role in postreplicative maintenance of DNA methylation. Nucleic Acids Res, 2007. 35(13): p. 4301-12.

62. Cartron, P.F., et al., Identification of TET1 Partners That Control Its DNA-Demethylating Function. Genes Cancer, 2013. 4(5-6): p. 235-41.

63. Litt, M.D., et al., Correlation between histone lysine methylation and developmental changes at the chicken beta-globin locus. Science, 2001. 293(5539): p. 2453-5.

64. Ruthenburg, A.J., C.D. Allis, and J. Wysocka, Methylation of lysine 4 on histone H3: intricacy of writing and reading a single epigenetic mark. Mol Cell, 2007. 25(1): p. 15-30.

65. Klose, R.J. and Y. Zhang, Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol, 2007. 8(4): p. 307-18.

66. Garcia, B.A., et al., Organismal differences in post-translational modifications in histones H3 and H4. J Biol Chem, 2007. 282(10): p. 7641-55.

67. Wu, H., et al., Structural biology of human H3K9 methyltransferases. PLoS One, 2010. 5(1): p. e8570.

68. Zhang, Y. and D. Reinberg, Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev, 2001. 15(18): p. 2343-60.

69. Peters, A.H., et al., Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell, 2001. 107(3): p. 323-37.

70. Lachner, M., et al., Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature, 2001. 410(6824): p. 116-20.

71. Jacobs, S.A. and S. Khorasanizadeh, Structure of HP1 chromodomain bound to a lysine 9-methylated histone H3 tail. Science, 2002. 295(5562): p. 2080-3.

72. Fischle, W., et al., Regulation of HP1-chromatin binding by histone H3 methylation and phosphorylation. Nature, 2005. 438(7071): p. 1116-22.

73. Nielsen, P.R., et al., Structure of the HP1 chromodomain bound to histone H3 methylated at lysine 9. Nature, 2002. 416(6876): p. 103-7.

74. Akam, M., The molecular basis for metameric pattern in the Drosophila embryo. Development, 1987. 101(1): p. 1-22.

75. Ingham, P.W., trithorax and the regulation of homeotic gene expression in Drosophila: a historical perspective. Int J Dev Biol, 1998. 42(3): p. 423-9.

Page 127: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

112

76. Schuettengruber, B., et al., Trithorax group proteins: switching genes on and keeping them active. Nat Rev Mol Cell Biol, 2011. 12(12): p. 799-814.

77. Lewis, E.B., A gene complex controlling segmentation in Drosophila. Nature, 1978. 276(5688): p. 565-70.

78. Schuettengruber, B. and G. Cavalli, Recruitment of polycomb group complexes and their role in the dynamic regulation of cell fate choice. Development, 2009. 136(21): p. 3531-42.

79. Schuettengruber, B., et al., Genome regulation by polycomb and trithorax proteins. Cell, 2007. 128(4): p. 735-45.

80. Pullirsch, D., et al., The Trithorax group protein Ash2l and Saf-A are recruited to the inactive X chromosome at the onset of stable X inactivation. Development, 2010. 137(6): p. 935-43.

81. Kennison, J.A., The Polycomb and trithorax group proteins of Drosophila: trans-regulators of homeotic gene function. Annu Rev Genet, 1995. 29: p. 289-303.

82. Li, B., et al., Polycomb protein Cbx4 promotes SUMO modification of de novo DNA methyltransferase Dnmt3a. Biochem J, 2007. 405(2): p. 369-78.

83. Pasini, D., et al., Suz12 is essential for mouse development and for EZH2 histone methyltransferase activity. Embo J, 2004. 23(20): p. 4061-71.

84. Kuzmichev, A., et al., Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein. Genes Dev, 2002. 16(22): p. 2893-905.

85. Morey, L. and K. Helin, Polycomb group protein-mediated repression of transcription. Trends Biochem Sci, 2010. 35(6): p. 323-32.

86. Li, G., et al., Jarid2 and PRC2, partners in regulating gene expression. Genes Dev, 2010. 24(4): p. 368-80.

87. Pasini, D., et al., JARID2 regulates binding of the Polycomb repressive complex 2 to target genes in ES cells. Nature, 2010. 464(7286): p. 306-10.

88. Czermin, B., et al., Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites. Cell, 2002. 111(2): p. 185-96.

89. Lanzuolo, C., F. Lo Sardo, and V. Orlando, Concerted epigenetic signatures inheritance at PcG targets through replication. Cell Cycle, 2012. 11(7): p. 1296-300.

90. Stock, J.K., et al., Ring1-mediated ubiquitination of H2A restrains poised RNA polymerase II at bivalent genes in mouse ES cells. Nat Cell Biol, 2007. 9(12): p. 1428-35.

Page 128: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

113

91. Kanhere, A., et al., Short RNAs are transcribed from repressed polycomb target genes and interact with polycomb repressive complex-2. Mol Cell, 2010. 38(5): p. 675-88.

92. Peng, J.C., et al., Jarid2/Jumonji coordinates control of PRC2 enzymatic activity and target gene occupancy in pluripotent cells. Cell, 2009. 139(7): p. 1290-302.

93. Rajasekhar, V.K. and M. Begemann, Concise review: roles of polycomb group proteins in development and disease: a stem cell perspective. Stem Cells, 2007. 25(10): p. 2498-510.

94. Rouleau, M., et al., PARP-3 associates with polycomb group bodies and with components of the DNA damage repair machinery. J Cell Biochem, 2007. 100(2): p. 385-401.

95. Siebold, A.P., et al., Polycomb Repressive Complex 2 and Trithorax modulate Drosophila longevity and stress resistance. Proc Natl Acad Sci U S A, 2010. 107(1): p. 169-74.

96. Varambally, S., et al., The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature, 2002. 419(6907): p. 624-9.

97. Kleer, C.G., et al., EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A, 2003. 100(20): p. 11606-11.

98. Sato, T., et al., PRC2 overexpression and PRC2-target gene repression relating to poorer prognosis in small cell lung cancer. Sci Rep, 2013. 3: p. 1911.

99. Chase, A. and N.C. Cross, Aberrations of EZH2 in cancer. Clin Cancer Res, 2011. 17(9): p. 2613-8.

100. Ryan, R.J., et al., EZH2 codon 641 mutations are common in BCL2-rearranged germinal center B cell lymphomas. PLoS One, 2011. 6(12): p. e28585.

101. Sing, A., et al., A vertebrate Polycomb response element governs segmentation of the posterior hindbrain. Cell, 2009. 138(5): p. 885-97.

102. Rinn, J.L., et al., Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell, 2007. 129(7): p. 1311-23.

103. Guttman, M., et al., Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature, 2009. 458(7235): p. 223-7.

104. Khalil, A.M., et al., Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci U S A, 2009. 106(28): p. 11667-72.

105. Kim, D.H., et al., Argonaute-1 directs siRNA-mediated transcriptional gene silencing in human cells. Nat Struct Mol Biol, 2006. 13(9): p. 793-7.

Page 129: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

114

106. Mendenhall, E.M., et al., GC-rich sequence elements recruit PRC2 in mammalian ES cells. PLoS Genet, 2010. 6(12): p. e1001244.

107. Lynch, M.D., et al., An interspecies analysis reveals a key role for unmethylated CpG dinucleotides in vertebrate Polycomb complex recruitment. EMBO J, 2012. 31(2): p. 317-29.

108. Reddington, J.P., et al., Redistribution of H3K27me3 upon DNA hypomethylation results in de-repression of Polycomb target genes. Genome Biol, 2013. 14(3): p. R25.

109. Corpet, A. and G. Almouzni, Making copies of chromatin: the challenge of nucleosomal organization and epigenetic information. Trends Cell Biol, 2009. 19(1): p. 29-41.

110. Hansen, K.H., et al., A model for transmission of the H3K27me3 epigenetic mark. Nat Cell Biol, 2008. 10(11): p. 1291-300.

111. Petruk, S., et al., TrxG and PcG proteins but not methylated histones remain associated with DNA through replication. Cell, 2012. 150(5): p. 922-33.

112. Wu, M., et al., Molecular regulation of H3K4 trimethylation by Wdr82, a component of human Set1/COMPASS. Mol Cell Biol, 2008. 28(24): p. 7337-44.

113. Winston, F. and M. Carlson, Yeast SNF/SWI transcriptional activators and the SPT/SIN chromatin connection. Trends Genet, 1992. 8(11): p. 387-91.

114. Ko, M., et al., Chromatin remodeling, development and disease. Mutat Res, 2008. 647(1-2): p. 59-67.

115. Ni, Z., et al., Apical role for BRG1 in cytokine-induced promoter assembly. Proc Natl Acad Sci U S A, 2005. 102(41): p. 14611-6.

116. Flaus, A. and T. Owen-Hughes, Mechanisms for nucleosome mobilization. Biopolymers, 2003. 68(4): p. 563-78.

117. Ho, L. and G.R. Crabtree, Chromatin remodelling during development. Nature, 2010. 463(7280): p. 474-84.

118. Gaspar-Maia, A., et al., Chd1 regulates open chromatin and pluripotency of embryonic stem cells. Nature, 2009. 460(7257): p. 863-8.

119. Abrams, E., L. Neigeborn, and M. Carlson, Molecular analysis of SNF2 and SNF5, genes required for expression of glucose-repressible genes in Saccharomyces cerevisiae. Mol Cell Biol, 1986. 6(11): p. 3643-51.

120. Carlson, M. and B.C. Laurent, The SNF/SWI family of global transcriptional activators. Curr Opin Cell Biol, 1994. 6(3): p. 396-402.

121. Neigeborn, L. and M. Carlson, Genes affecting the regulation of SUC2 gene expression by glucose repression in Saccharomyces cerevisiae. Genetics, 1984. 108(4): p. 845-58.

Page 130: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

115

122. Kadam, S. and B.M. Emerson, Transcriptional specificity of human SWI/SNF BRG1 and BRM chromatin remodeling complexes. Mol Cell, 2003. 11(2): p. 377-89.

123. Bultman, S., et al., A Brg1 null mutation in the mouse reveals functional differences among mammalian SWI/SNF complexes. Mol Cell, 2000. 6(6): p. 1287-95.

124. Khavari, P.A., et al., BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature, 1993. 366(6451): p. 170-4.

125. Kadam, S. and B.M. Emerson, Transcriptional specificity of human SWI/SNF BRG1 and BRM chromatin remodeling complexes. Mol Cell, 2003. 11(2): p. 377-89.

126. Cohen, S.M., et al., BRG1 co-localizes with DNA replication factors and is required for efficient replication fork progression. Nucleic Acids Res, 2010. 38(20): p. 6906-19.

127. Wong, A.K., et al., BRG1, a component of the SWI-SNF complex, is mutated in multiple human tumor cell lines. Cancer Res, 2000. 60(21): p. 6171-7.

128. Ni, Z. and R. Bremner, Brahma-related gene 1-dependent STAT3 recruitment at IL-6-inducible genes. J Immunol, 2007. 178(1): p. 345-51.

129. Armstrong, J.A., et al., Genetic screens for enhancers of brahma reveal functional interactions between the BRM chromatin-remodeling complex and the delta-notch signal transduction pathway in Drosophila. Genetics, 2005. 170(4): p. 1761-74.

130. Strehlow, I. and T. Decker, Transcriptional induction of IFN-gamma-responsive genes is modulated by DNA surrounding the interferon stimulation response element. Nucleic Acids Res, 1992. 20(15): p. 3865-72.

131. Dunn, G.P., C.M. Koebel, and R.D. Schreiber, Interferons, immunity and cancer immunoediting. Nat Rev Immunol, 2006. 6(11): p. 836-48.

132. Pestka, S., C.D. Krause, and M.R. Walter, Interferons, interferon-like cytokines, and their receptors. Immunol Rev, 2004. 202: p. 8-32.

133. Boehm, U., et al., Cellular responses to interferon-gamma. Annu Rev Immunol, 1997. 15: p. 749-95.

134. Celada, A., M.J. Klemsz, and R.A. Maki, Interferon-gamma activates multiple pathways to regulate the expression of the genes for major histocompatibility class II I-A beta, tumor necrosis factor and complement component C3 in mouse macrophages. Eur. J. Immunol., 1989. 19: p. 1103-9.

135. Maher, S.G., et al., Interferon: cellular executioner or white knight? Curr Med Chem, 2007. 14(12): p. 1279-89.

136. Ullrich, E., et al., Therapy-induced tumor immunosurveillance involves IFN-producing killer dendritic cells. Cancer Res, 2007. 67(3): p. 851-3.

Page 131: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

116

137. Ni, Z., et al., The chromatin-remodeling enzyme BRG1 coordinates CIITA induction through many interdependent distal enhancers. Nat Immunol, 2008. 9(7): p. 785-93.

138. Wright, K.L. and J.P. Ting, Epigenetic regulation of MHC-II and CIITA genes. Trends Immunol, 2006. 27(9): p. 405-12.

139. Muhlethaler-Mottet, A., et al., Expression of MHC class II molecules in different cellular and functional compartments is controlled by differential usage of multiple promoters of the transactivator CIITA. Embo J, 1997. 16(10): p. 2851-60.

140. Holling, T.M., et al., A Role for EZH2 in Silencing of IFN-{gamma} Inducible MHC2TA Transcription in Uveal Melanoma. J Immunol, 2007. 179(8): p. 5317-25.

141. Satoh, A., et al., Epigenetic inactivation of class II transactivator (CIITA) is associated with the absence of interferon-gamma-induced HLA-DR expression in colorectal and gastric cancer cells. Oncogene, 2004. 23(55): p. 8876-86.

142. Smith, S. and B. Stillman, Purification and characterization of CAF-I, a human cell factor required for chromatin assembly during DNA replication in vitro. Cell, 1989. 58(1): p. 15-25.

143. Poleshko, A., et al., Identification of a functional network of human epigenetic silencing factors. J Biol Chem. 285(1): p. 422-33.

144. Huang, H., et al., Drosophila CAF-1 regulates HP1-mediated epigenetic silencing and pericentric heterochromatin stability. J Cell Sci, 2010. 123(Pt 16): p. 2853-61.

145. Quivy, J.P., et al., The HP1-p150/CAF-1 interaction is required for pericentric heterochromatin replication and S-phase progression in mouse cells. Nat Struct Mol Biol, 2008. 15(9): p. 972-9.

146. Shibahara, K. and B. Stillman, Replication-dependent marking of DNA by PCNA facilitates CAF-1-coupled inheritance of chromatin. Cell, 1999. 96(4): p. 575-85.

147. Rolef Ben-Shahar, T., et al., Two fundamentally distinct PCNA interaction peptides contribute to chromatin assembly factor 1 function. Mol Cell Biol, 2009. 29(24): p. 6353-65.

148. Rafiei, M. and R. Bremner, Histone Chaperones, Epigenetics, and Cancer. Ststems Analysis of Chromatin-Related protein Complexes in Cancer, ed. A. Emili, J. Greenblatt, and S. Wodak. 2013: Springer.

149. Laskey, R.A., et al., Nucleosomes are assembled by an acidic protein which binds histones and transfers them to DNA. Nature, 1978. 275(5679): p. 416-20.

150. English, C.M., et al., Structural basis for the histone chaperone activity of Asf1. Cell, 2006. 127(3): p. 495-508.

Page 132: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

117

151. Akey, C.W. and K. Luger, Histone chaperones and nucleosome assembly. Curr Opin Struct Biol, 2003. 13(1): p. 6-14.

152. Loyola, A. and G. Almouzni, Histone chaperones, a supporting role in the limelight. Biochim Biophys Acta, 2004. 1677(1-3): p. 3-11.

153. Bonasio, R., S. Tu, and D. Reinberg, Molecular signals of epigenetic states. Science, 2010. 330(6004): p. 612-6.

154. Hake, S.B. and C.D. Allis, Histone H3 variants and their potential role in indexing mammalian genomes: the "H3 barcode hypothesis". Proc Natl Acad Sci U S A, 2006. 103(17): p. 6428-35.

155. Xu, M., et al., Partitioning of histone H3-H4 tetramers during DNA replication-dependent chromatin assembly. Science, 2010. 328(5974): p. 94-8.

156. Luger, K., et al., Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature, 1997. 389(6648): p. 251-60.

157. Wu, R.S., S. Tsai, and W.M. Bonner, Patterns of histone variant synthesis can distinguish G0 from G1 cells. Cell, 1982. 31(2 Pt 1): p. 367-74.

158. Kappes, F., et al., The DEK oncoprotein is a Su(var) that is essential to heterochromatin integrity. Genes Dev, 2011. 25(7): p. 673-8.

159. Ray-Gallet, D., et al., HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol Cell, 2002. 9(5): p. 1091-100.

160. Graham, P.J., Developments in occupational health and safety qualifications. Ann Occup Hyg, 1992. 36(6): p. 663-8.

161. Goldberg, A.D., et al., Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell, 2010. 140(5): p. 678-91.

162. Drane, P., et al., The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev, 2010. 24(12): p. 1253-65.

163. Lewis, P.W., et al., Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci U S A, 2011. 107(32): p. 14075-80.

164. Campos, E.I., et al., The program for processing newly synthesized histones H3.1 and H4. Nat Struct Mol Biol, 2010. 17(11): p. 1343-51.

165. Loyola, A., et al., PTMs on H3 variants before chromatin assembly potentiate their final epigenetic state. Mol Cell, 2006. 24(2): p. 309-16.

Page 133: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

118

166. Richardson, R.T., et al., Characterization of the NASP promoter in 3T3 fibroblasts and mouse spermatogenic cells. Gene, 2006. 371(1): p. 52-8.

167. Song, J.J., J.D. Garlick, and R.E. Kingston, Structural basis of histone H4 recognition by p55. Genes Dev, 2008. 22(10): p. 1313-8.

168. Wang, H., S.T. Walsh, and M.R. Parthun, Expanded binding specificity of the human histone chaperone NASP. Nucleic Acids Res, 2008. 36(18): p. 5763-72.

169. Finn, R.M., et al., sNASP, a histone H1-specific eukaryotic chaperone dimer that facilitates chromatin assembly. Biophys J, 2008. 95(3): p. 1314-25.

170. Ejlassi-Lassallette, A., et al., H4 replication-dependent diacetylation and Hat1 promote S-phase chromatin assembly in vivo. Mol Biol Cell, 2011. 22(2): p. 245-55.

171. Zhang, H., et al., Human histone acetyltransferase 1 protein preferentially acetylates H4 histone molecules in H3.1-H4 over H3.3-H4. J Biol Chem, 2012. 287(9): p. 6573-81.

172. Cook, A.J., et al., A specific function for the histone chaperone NASP to fine-tune a reservoir of soluble H3-H4 in the histone supply chain. Mol Cell, 2011. 44(6): p. 918-27.

173. Tagami, H., et al., Histone H3.1 and H3.3 complexes mediate nucleosome assembly pathways dependent or independent of DNA synthesis. Cell, 2004. 116(1): p. 51-61.

174. Elsasser, S.J., et al., DAXX envelops a histone H3.3-H4 dimer for H3.3-specific recognition. Nature, 2012. 491(7425): p. 560-5.

175. Kaufman, P.D., R. Kobayashi, and B. Stillman, Ultraviolet radiation sensitivity and reduction of telomeric silencing in Saccharomyces cerevisiae cells lacking chromatin assembly factor-I. Genes Dev, 1997. 11(3): p. 345-57.

176. Bulger, M., et al., Assembly of regularly spaced nucleosome arrays by Drosophila chromatin assembly factor 1 and a 56-kDa histone-binding protein. Proc Natl Acad Sci U S A, 1995. 92(25): p. 11726-30.

177. Kamakaka, R.T., et al., Postreplicative chromatin assembly by Drosophila and human chromatin assembly factor 1. Mol Cell Biol, 1996. 16(3): p. 810-7.

178. Tyler, J.K., et al., Interaction between the Drosophila CAF-1 and ASF1 chromatin assembly factors. Mol Cell Biol, 2001. 21(19): p. 6574-84.

179. Malay, A.D., et al., Crystal structures of fission yeast histone chaperone Asf1 complexed with the Hip1 B-domain or the Cac2 C terminus. J Biol Chem, 2008. 283(20): p. 14022-31.

180. Nowak, A.J., et al., Chromatin-modifying complex component Nurf55/p55 associates with histones H3 and H4 and polycomb repressive complex 2 subunit Su(z)12 through partially overlapping binding sites. J Biol Chem, 2011. 286(26): p. 23388-96.

Page 134: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

119

181. Nakano, S., B. Stillman, and H.R. Horvitz, Replication-coupled chromatin assembly generates a neuronal bilateral asymmetry in C. elegans. Cell, 2011. 147(7): p. 1525-36.

182. Winkler, D.D., et al., Yeast CAF-1 assembles histone (H3-H4)2 tetramers prior to DNA deposition. Nucleic Acids Res, 2012. 40(20): p. 10139-49.

183. Liu, W.H., et al., CAF-1-induced oligomerization of histones H3/H4 and mutually exclusive interactions with Asf1 guide H3/H4 transitions among histone chaperones and DNA. Nucleic Acids Res, 2011. 40(22): p. 11229-39.

184. Liu, W.H. and M.E. Churchill, Histone transfer among chaperones. Biochem Soc Trans, 2012. 40(2): p. 357-63.

185. Tsubota, T., et al., Histone H3-K56 acetylation is catalyzed by histone chaperone-dependent complexes. Mol Cell, 2007. 25(5): p. 703-12.

186. Han, J., et al., The Rtt109-Vps75 histone acetyltransferase complex acetylates non-nucleosomal histone H3. J Biol Chem, 2007. 282(19): p. 14158-64.

187. Das, C., et al., CBP/p300-mediated acetylation of histone H3 on lysine 56. Nature, 2009. 459(7243): p. 113-7.

188. Li, Q., et al., Acetylation of histone H3 lysine 56 regulates replication-coupled nucleosome assembly. Cell, 2008. 134(2): p. 244-55.

189. Feser, J., et al., Elevated histone expression promotes life span extension. Mol Cell, 2010. 39(5): p. 724-35.

190. Kang, B., et al., Phosphorylation of H4 Ser 47 promotes HIRA-mediated nucleosome assembly. Genes Dev, 2011. 25(13): p. 1359-64.

191. Burgess, R.J. and Z. Zhang, Histone chaperones in nucleosome assembly and human disease. Nat Struct Mol Biol, 2013. 20(1): p. 14-22.

192. Lopez de Saro, F.J., Regulation of interactions with sliding clamps during DNA replication and repair. Curr Genomics, 2009. 10(3): p. 206-15.

193. Zhang, Z., K. Shibahara, and B. Stillman, PCNA connects DNA replication to epigenetic inheritance in yeast. Nature, 2000. 408(6809): p. 221-5.

194. Moggs, J.G., et al., A CAF-1-PCNA-mediated chromatin assembly pathway triggered by sensing DNA damage. Mol Cell Biol, 2000. 20(4): p. 1206-18.

195. Ye, X., et al., Defective S phase chromatin assembly causes DNA damage, activation of the S phase checkpoint, and S phase arrest. Mol Cell, 2003. 11(2): p. 341-51.

196. Ransom, M., et al., FACT and the proteasome promote promoter chromatin disassembly and transcriptional initiation. J Biol Chem, 2009. 284(35): p. 23461-71.

Page 135: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

120

197. Winkler, D.D. and K. Luger, The histone chaperone FACT: structural insights and mechanisms for nucleosome reorganization. J Biol Chem, 2011. 286(21): p. 18369-74.

198. Gambus, A., et al., GINS maintains association of Cdc45 with MCM in replisome progression complexes at eukaryotic DNA replication forks. Nat Cell Biol, 2006. 8(4): p. 358-66.

199. Lorch, Y., B. Maier-Davis, and R.D. Kornberg, Chromatin remodeling by nucleosome disassembly in vitro. Proc Natl Acad Sci U S A, 2006. 103(9): p. 3090-3.

200. Park, Y.J., et al., Nucleosome assembly protein 1 exchanges histone H2A-H2B dimers and assists nucleosome sliding. J Biol Chem, 2005. 280(3): p. 1817-25.

201. Park, Y.J. and K. Luger, Structure and function of nucleosome assembly proteins. Biochem Cell Biol, 2006. 84(4): p. 549-58.

202. Worcel, A., S. Han, and M.L. Wong, Assembly of newly replicated chromatin. Cell, 1978. 15(3): p. 969-77.

203. Groth, A., et al., Regulation of replication fork progression through histone supply and demand. Science, 2007. 318(5858): p. 1928-31.

204. Tyler, J.K., et al., The RCAF complex mediates chromatin assembly during DNA replication and repair. Nature, 1999. 402(6761): p. 555-60.

205. Natsume, R., et al., Structure and function of the histone chaperone CIA/ASF1 complexed with histones H3 and H4. Nature, 2007. 446(7133): p. 338-41.

206. Smith, S. and B. Stillman, Stepwise assembly of chromatin during DNA replication in vitro. Embo J, 1991. 10(4): p. 971-80.

207. Bowman, A., et al., The histone chaperones Nap1 and Vps75 bind histones H3 and H4 in a tetrameric conformation. Mol Cell, 2011. 41(4): p. 398-408.

208. Corpet, A., et al., Asf1b, the necessary Asf1 isoform for proliferation, is predictive of outcome in breast cancer. Embo J, 2011. 30(3): p. 480-93.

209. Mousson, F., F. Ochsenbein, and C. Mann, The histone chaperone Asf1 at the crossroads of chromatin and DNA checkpoint pathways. Chromosoma, 2007. 116(2): p. 79-93.

210. Zeng, W., A.R. Ball, Jr., and K. Yokomori, HP1: heterochromatin binding proteins working the genome. Epigenetics, 2010. 5(4): p. 287-92.

211. Schultz, D.C., et al., SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev, 2002. 16(8): p. 919-32.

212. Schotta, G., et al., Central role of Drosophila SU(VAR)3-9 in histone H3-K9 methylation and heterochromatic gene silencing. Embo J, 2002. 21(5): p. 1121-31.

Page 136: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

121

213. Quivy, J.P., P. Grandi, and G. Almouzni, Dimerization of the largest subunit of chromatin assembly factor 1: importance in vitro and during Xenopus early development. Embo J, 2001. 20(8): p. 2015-27.

214. Ono, T., et al., Chromatin assembly factor 1 ensures the stable maintenance of silent chromatin states in Arabidopsis. Genes Cells, 2006. 11(2): p. 153-62.

215. Schonrock, N., et al., Functional genomic analysis of CAF-1 mutants in Arabidopsis thaliana. J Biol Chem, 2006. 281(14): p. 9560-8.

216. Song, Y., et al., CAF-1 is essential for Drosophila development and involved in the maintenance of epigenetic memory. Dev Biol, 2007. 311(1): p. 213-22.

217. Dohke, K., et al., Fission yeast chromatin assembly factor 1 assists in the replication-coupled maintenance of heterochromatin. Genes Cells, 2008. 13(10): p. 1027-43.

218. Enomoto, S., et al., RLF2, a subunit of yeast chromatin assembly factor-I, is required for telomeric chromatin function in vivo. Genes Dev, 1997. 11(3): p. 358-70.

219. Monson, E.K., D. de Bruin, and V.A. Zakian, The yeast Cac1 protein is required for the stable inheritance of transcriptionally repressed chromatin at telomeres. Proc Natl Acad Sci U S A, 1997. 94(24): p. 13081-6.

220. Enomoto, S. and J. Berman, Chromatin assembly factor I contributes to the maintenance, but not the re-establishment, of silencing at the yeast silent mating loci. Genes Dev, 1998. 12(2): p. 219-32.

221. Taddei, A., et al., Duplication and maintenance of heterochromatin domains. J Cell Biol, 1999. 147(6): p. 1153-66.

222. Quivy, J.P., et al., A CAF-1 dependent pool of HP1 during heterochromatin duplication. EMBO J, 2004. 23(17): p. 3516-26.

223. Reese, B.E., et al., The methyl-CpG binding protein MBD1 interacts with the p150 subunit of chromatin assembly factor 1. Mol Cell Biol, 2003. 23(9): p. 3226-36.

224. Sarraf, S.A. and I. Stancheva, Methyl-CpG binding protein MBD1 couples histone H3 methylation at lysine 9 by SETDB1 to DNA replication and chromatin assembly. Mol Cell, 2004. 15(4): p. 595-605.

225. Loyola, A., et al., The HP1alpha-CAF1-SetDB1-containing complex provides H3K9me1 for Suv39-mediated K9me3 in pericentric heterochromatin. EMBO Rep, 2009. 10(7): p. 769-75.

226. Houlard, M., et al., CAF-1 is essential for heterochromatin organization in pluripotent embryonic cells. PLoS Genet, 2006. 2(11): p. e181.

Page 137: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

122

227. Hoek, M. and B. Stillman, Chromatin assembly factor 1 is essential and couples chromatin assembly to DNA replication in vivo. Proc Natl Acad Sci U S A, 2003. 100(21): p. 12183-8.

228. Nabatiyan, A. and T. Krude, Silencing of chromatin assembly factor 1 in human cells leads to cell death and loss of chromatin assembly during DNA synthesis. Mol Cell Biol, 2004. 24(7): p. 2853-62.

229. Takami, Y., et al., Essential role of chromatin assembly factor-1-mediated rapid nucleosome assembly for DNA replication and cell division in vertebrate cells. Mol Biol Cell, 2007. 18(1): p. 129-41.

230. Poleshko, A., et al., Identification of a functional network of human epigenetic silencing factors. J Biol Chem, 2010. 285(1): p. 422-33.

231. Hoek, M., M.P. Myers, and B. Stillman, An analysis of CAF-1-interacting proteins reveals dynamic and direct interactions with the KU complex and 14-3-3 proteins. J Biol Chem, 2011. 286(12): p. 10876-87.

232. Polo, S.E., et al., Clinical significance and prognostic value of chromatin assembly factor-1 overexpression in human solid tumours. Histopathology, 2010. 57(5): p. 716-24.

233. Groth, A., et al., Human Asf1 regulates the flow of S phase histones during replicational stress. Mol Cell, 2005. 17(2): p. 301-11.

234. Gazin, C., et al., An elaborate pathway required for Ras-mediated epigenetic silencing. Nature, 2007. 449(7165): p. 1073-7.

235. Avvakumov, N., A. Nourani, and J. Cote, Histone chaperones: modulators of chromatin marks. Mol Cell, 2011. 41(5): p. 502-14.

236. Masumoto, H., et al., A role for cell-cycle-regulated histone H3 lysine 56 acetylation in the DNA damage response. Nature, 2005. 436(7048): p. 294-8.

237. Neumann, H., et al., A method for genetically installing site-specific acetylation in recombinant histones defines the effects of H3 K56 acetylation. Mol Cell, 2009. 36(1): p. 153-63.

238. Zhang, R., et al., Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev Cell, 2005. 8(1): p. 19-30.

239. Rai, T.S. and P.D. Adams, Lessons from senescence: Chromatin maintenance in non-proliferating cells. Biochim Biophys Acta, 2012. 1819(3-4): p. 322-31.

240. Mehrotra, P.V., et al., DNA repair factor APLF is a histone chaperone. Mol Cell, 2011. 41(1): p. 46-55.

Page 138: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

123

241. Spector, M.S., et al., Hir1p and Hir2p function as transcriptional corepressors to regulate histone gene transcription in the Saccharomyces cerevisiae cell cycle. Mol Cell Biol, 1997. 17(2): p. 545-52.

242. Sharp, J.A., et al., Yeast histone deposition protein Asf1p requires Hir proteins and PCNA for heterochromatic silencing. Curr Biol, 2001. 11(7): p. 463-73.

243. Sutton, A., et al., Yeast ASF1 protein is required for cell cycle regulation of histone gene transcription. Genetics, 2001. 158(2): p. 587-96.

244. Daganzo, S.M., et al., Structure and function of the conserved core of histone deposition protein Asf1. Curr Biol, 2003. 13(24): p. 2148-58.

245. Fillingham, J., et al., Two-color cell array screen reveals interdependent roles for histone chaperones and a chromatin boundary regulator in histone gene repression. Mol Cell, 2009. 35(3): p. 340-51.

246. Passtoors, W.M., et al., Transcriptional profiling of human familial longevity indicates a role for ASF1A and IL7R. PLoS One, 2012. 7(1): p. e27759.

247. Sawatsubashi, S., et al., A histone chaperone, DEK, transcriptionally coactivates a nuclear receptor. Genes Dev, 2010. 24(2): p. 159-70.

248. Gamble, M.J. and R.P. Fisher, SET and PARP1 remove DEK from chromatin to permit access by the transcription machinery. Nat Struct Mol Biol, 2007. 14(6): p. 548-55.

249. von Lindern, M., et al., Characterization of the translocation breakpoint sequences of two DEK-CAN fusion genes present in t(6;9) acute myeloid leukemia and a SET-CAN fusion gene found in a case of acute undifferentiated leukemia. Genes Chromosomes Cancer, 1992. 5(3): p. 227-34.

250. Fornerod, M., et al., Relocation of the carboxyterminal part of CAN from the nuclear envelope to the nucleus as a result of leukemia-specific chromosome rearrangements. Oncogene, 1995. 10(9): p. 1739-48.

251. Khodadoust, M.S., et al., Melanoma proliferation and chemoresistance controlled by the DEK oncogene. Cancer Res, 2009. 69(16): p. 6405-13.

252. Han, S., et al., Clinicopathological significance of DEK overexpression in serous ovarian tumors. Pathol Int, 2009. 59(7): p. 443-7.

253. Orlic, M., et al., Expression analysis of 6p22 genomic gain in retinoblastoma. Genes Chromosomes Cancer, 2006. 45(1): p. 72-82.

254. Shibata, T., et al., DEK oncoprotein regulates transcriptional modifiers and sustains tumor initiation activity in high-grade neuroendocrine carcinoma of the lung. Oncogene, 2010. 29(33): p. 4671-81.

Page 139: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

124

255. Carro, M.S., et al., DEK Expression is controlled by E2F and deregulated in diverse tumor types. Cell Cycle, 2006. 5(11): p. 1202-7.

256. Wise-Draper, T.M., et al., The human DEK proto-oncogene is a senescence inhibitor and an upregulated target of high-risk human papillomavirus E7. J Virol, 2005. 79(22): p. 14309-17.

257. Wise-Draper, T.M., et al., Apoptosis inhibition by the human DEK oncoprotein involves interference with p53 functions. Mol Cell Biol, 2006. 26(20): p. 7506-19.

258. Kavanaugh, G.M., et al., The human DEK oncogene regulates DNA damage response signaling and repair. Nucleic Acids Res, 2011. 39(17): p. 7465-76.

259. Privette Vinnedge, L.M., et al., The human DEK oncogene stimulates beta-catenin signaling, invasion and mammosphere formation in breast cancer. Oncogene, 2011. 30(24): p. 2741-52.

260. Wise-Draper, T.M., et al., Overexpression of the cellular DEK protein promotes epithelial transformation in vitro and in vivo. Cancer Res, 2009. 69(5): p. 1792-9.

261. McGarvey, T., et al., The acute myeloid leukemia-associated protein, DEK, forms a splicing-dependent interaction with exon-product complexes. J Cell Biol, 2000. 150(2): p. 309-20.

262. Soares, L.M., et al., Intron removal requires proofreading of U2AF/3' splice site recognition by DEK. Science, 2006. 312(5782): p. 1961-5.

263. Ageberg, M., et al., Identification of a novel and myeloid specific role of the leukemia-associated fusion protein DEK-NUP214 leading to increased protein synthesis. Genes Chromosomes Cancer, 2008. 47(4): p. 276-87.

264. Salomoni, P. and A.F. Khelifi, Daxx: death or survival protein? Trends Cell Biol, 2006. 16(2): p. 97-104.

265. McDowell, T.L., et al., Localization of a putative transcriptional regulator (ATRX) at pericentromeric heterochromatin and the short arms of acrocentric chromosomes. Proc Natl Acad Sci U S A, 1999. 96(24): p. 13983-8.

266. Ishov, A.M., O.V. Vladimirova, and G.G. Maul, Heterochromatin and ND10 are cell-cycle regulated and phosphorylation-dependent alternate nuclear sites of the transcription repressor Daxx and SWI/SNF protein ATRX. J Cell Sci, 2004. 117(Pt 17): p. 3807-20.

267. Jiao, R., et al., Physical and functional interaction between the Bloom's syndrome gene product and the largest subunit of chromatin assembly factor 1. Mol Cell Biol, 2004. 24(11): p. 4710-9.

268. Heaphy, C.M., et al., Altered telomeres in tumors with ATRX and DAXX mutations. Science, 2011. 333(6041): p. 425.

Page 140: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

125

269. Schwartzentruber, J., et al., Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature, 2012. 482(7384): p. 226-31.

270. Ding, L., et al., Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature, 2012. 481(7382): p. 506-10.

271. Dunn, G.P., et al., Interferon-gamma and cancer immunoediting. Immunol Res, 2005. 32(1-3): p. 231-45.

272. Boehm, U., et al., Cellular responses to interferon-gamma. Annu Rev Immunol, 1997. 15: p. 749-95.

273. Reith, W. and B. Mach, The bare lymphocyte syndrome and the regulation of MHC expression. Annu Rev Immunol, 2001. 19: p. 331-73.

274. Marine, J.C., et al., SOCS1 deficiency causes a lymphocyte-dependent perinatal lethality. Cell, 1999. 98(5): p. 609-16.

275. Alexander, W.S., et al., SOCS1 is a critical inhibitor of interferon gamma signaling and prevents the potentially fatal neonatal actions of this cytokine. Cell, 1999. 98(5): p. 597-608.

276. Deffrennes, V., et al., Constitutive expression of MHC class II genes in melanoma cell lines results from the transcription of class II transactivator abnormally initiated from its B cell-specific promoter. J Immunol, 2001. 167(1): p. 98-106.

277. Swanberg, M., et al., MHC2TA is associated with differential MHC molecule expression and susceptibility to rheumatoid arthritis, multiple sclerosis and myocardial infarction. Nat Genet, 2005. 37(5): p. 486-94.

278. Holling, T.M., et al., Epigenetic silencing of MHC2TA transcription in cancer. Biochem Pharmacol, 2006. 72(11): p. 1570-6.

279. Hobart, M., et al., IFN regulatory factor-1 plays a central role in the regulation of the expression of class I and II MHC genes in vivo. J Immunol, 1997. 158(9): p. 4260-9.

280. Sauvageau, M. and G. Sauvageau, Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer. Cell Stem Cell, 2010. 7(3): p. 299-313.

281. Soshnikova, N. and D. Duboule, Epigenetic regulation of Hox gene activation: the waltz of methyls. Bioessays, 2008. 30(3): p. 199-202.

282. Kia, S.K., et al., SWI/SNF mediates polycomb eviction and epigenetic reprogramming of the INK4b-ARF-INK4a locus. Mol Cell Biol, 2008. 28(10): p. 3457-64.

283. Soshnikova, N. and D. Duboule, Epigenetic regulation of vertebrate Hox genes: a dynamic equilibrium. Epigenetics, 2009. 4(8): p. 537-40.

Page 141: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

126

284. Matzke, M.A. and J.A. Birchler, RNAi-mediated pathways in the nucleus. Nat Rev Genet, 2005. 6(1): p. 24-35.

285. Baeg, G.H., R. Zhou, and N. Perrimon, Genome-wide RNAi analysis of JAK/STAT signaling components in Drosophila. Genes Dev, 2005. 19(16): p. 1861-70.

286. Kittler, R., et al., Genome-wide resources of endoribonuclease-prepared short interfering RNAs for specific loss-of-function studies. Nat Methods, 2007. 4(4): p. 337-44.

287. Lu, J., et al., A genome-wide RNA interference screen identifies putative chromatin regulators essential for E2F repression. Proc Natl Acad Sci U S A, 2007. 104(22): p. 9381-6.

288. Pal-Bhadra, M., U. Bhadra, and J.A. Birchler, RNAi related mechanisms affect both transcriptional and posttranscriptional transgene silencing in Drosophila. Mol Cell, 2002. 9(2): p. 315-27.

289. Yu, J.Y., S.L. DeRuiter, and D.L. Turner, RNA interference by expression of short-interfering RNAs and hairpin RNAs in mammalian cells. Proc Natl Acad Sci U S A, 2002. 99(9): p. 6047-52.

290. Muller, P., et al., Identification of JAK/STAT signalling components by genome-wide RNA interference. Nature, 2005. 436(7052): p. 871-5.

291. Brideau, C., et al., Improved statistical methods for hit selection in high-throughput screening. J Biomol Screen, 2003. 8(6): p. 634-47.

292. Makarenkov, V., et al., An efficient method for the detection and elimination of systematic error in high-throughput screening. Bioinformatics, 2007. 23(13): p. 1648-57.

293. Malo, N., et al., Statistical practice in high-throughput screening data analysis. Nat Biotechnol, 2006. 24(2): p. 167-75.

294. Chang, C.H., et al., Class II transactivator (CIITA) is sufficient for the inducible expression of major histocompatibility complex class II genes. J Exp Med, 1994. 180(4): p. 1367-74.

295. O'Keefe, G.M., et al., IFN-gamma regulation of class II transactivator promoter IV in macrophages and microglia: involvement of the suppressors of cytokine signaling-1 protein. J Immunol, 2001. 166(4): p. 2260-9.

296. Marignani, P.A., F. Kanai, and C.L. Carpenter, LKB1 associates with Brg1 and is necessary for Brg1-induced growth arrest. J Biol Chem, 2001. 276(35): p. 32415-8.

297. Welsh, M., et al., The tyrosine kinase FRK/RAK participates in cytokine-induced islet cell cytotoxicity. Biochem J, 2004. 382(Pt 1): p. 261-8.

298. Alsayed, Y., et al., IFN-gamma activates the C3G/Rap1 signaling pathway. J Immunol, 2000. 164(4): p. 1800-6.

Page 142: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

127

299. Steimle, V., et al., Regulation of MHC class II expression by interferon-gamma mediated by the transactivator gene CIITA. Science, 1994. 265(5168): p. 106-9.

300. Kimura, T., et al., Involvement of the IRF-1 transcription factor in antiviral responses to interferons. Science, 1994. 264(5167): p. 1921-4.

301. Platanias, L.C., Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol, 2005. 5(5): p. 375-86.

302. Qin, Z. and T. Blankenstein, A cancer immunosurveillance controversy. Nat Immunol, 2004. 5(1): p. 3-4; author reply 4-5.

303. Golbabapour, S., et al., Gene silencing and Polycomb group proteins: an overview of their structure, mechanisms and phylogenetics. OMICS, 2013. 17(6): p. 283-96.

304. Goodrich, J., et al., A Polycomb-group gene regulates homeotic gene expression in Arabidopsis [see comments]. Nature, 1997. 385(6620): p. 44-51.

305. Gunster, M.J., et al., Differential expression of human Polycomb group proteins in various tissues and cell types. J Cell Biochem Suppl, 2001. Suppl 36: p. 129-43.

306. Kohler, C. and U. Grossniklaus, Epigenetic inheritance of expression states in plant development: the role of Polycomb group proteins. Curr Opin Cell Biol, 2002. 14(6): p. 773-9.

307. Oktaba, K., et al., Dynamic regulation by polycomb group protein complexes controls pattern formation and the cell cycle in Drosophila. Dev Cell, 2008. 15(6): p. 877-89.

308. Sparmann, A. and M. van Lohuizen, Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer, 2006. 6(11): p. 846-56.

309. Wang, J., et al., Imprinted X inactivation maintained by a mouse Polycomb group gene. Nat Genet, 2001. 28(4): p. 371-5.

310. Gil, J., D. Bernard, and G. Peters, Role of polycomb group proteins in stem cell self-renewal and cancer. DNA Cell Biol, 2005. 24(2): p. 117-25.

311. Martinez, A.M. and G. Cavalli, The role of polycomb group proteins in cell cycle regulation during development. Cell Cycle, 2006. 5(11): p. 1189-97.

312. Simon, J.A., Polycomb group proteins. Curr Biol, 2003. 13(3): p. R79-80.

313. Verreault, A., et al., Nucleosome assembly by a complex of CAF-1 and acetylated histones H3/H4. Cell, 1996. 87(1): p. 95-104.

314. Xu, M. and B. Zhu, Nucleosome assembly and epigenetic inheritance. Protein Cell, 2010. 1(9): p. 820-9.

Page 143: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

128

315. Khavari, P.A., et al., BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature, 1993. 366(6451): p. 170-4.

316. Muchardt, C. and M. Yaniv, A human homologue of Saccharomyces cerevisiae SNF2/SWI2 and Drosophila brm genes potentiates transcriptional activation by the glucocorticoid receptor. Embo J, 1993. 12(11): p. 4279-90.

317. Liu, R., et al., Regulation of CSF1 promoter by the SWI/SNF-like BAF complex. Cell, 2001. 106(3): p. 309-18.

318. Staibano, S., et al., Chromatin assembly factor-1 (CAF-1)-mediated regulation of cell proliferation and DNA repair: a link with the biological behaviour of squamous cell carcinoma of the tongue? Histopathology, 2007. 50(7): p. 911-9.

319. Ransom, M., B.K. Dennehey, and J.K. Tyler, Chaperoning histones during DNA replication and repair. Cell, 2010. 140(2): p. 183-95.

320. Martini, E., et al., Recruitment of phosphorylated chromatin assembly factor 1 to chromatin after UV irradiation of human cells. J Cell Biol, 1998. 143(3): p. 563-75.

321. Craig, J.M., Heterochromatin--many flavours, common themes. Bioessays, 2005. 27(1): p. 17-28.

322. Sumer, H., et al., Effects of scaffold/matrix alteration on centromeric function and gene expression. J Biol Chem, 2004. 279(36): p. 37631-9.

323. Sakamoto, Y., et al., Overlapping roles of the methylated DNA-binding protein MBD1 and polycomb group proteins in transcriptional repression of HOXA genes and heterochromatin foci formation. J Biol Chem, 2007. 282(22): p. 16391-400.

324. Ahmad, A., Y. Takami, and T. Nakayama, WD dipeptide motifs and LXXLL motif of chicken HIRA are essential for interactions with the p48 subunit of chromatin assembly factor-1 and histone deacetylase-2 in vitro and in vivo. Gene, 2004. 342(1): p. 125-36.

325. Kaufman, P.D., et al., The p150 and p60 subunits of chromatin assembly factor I: a molecular link between newly synthesized histones and DNA replication. Cell, 1995. 81(7): p. 1105-14.

326. Tchenio, T., J.F. Casella, and T. Heidmann, A truncated form of the human CAF-1 p150 subunit impairs the maintenance of transcriptional gene silencing in mammalian cells. Mol Cell Biol, 2001. 21(6): p. 1953-61.

327. Cao, Q., et al., Repression of E-cadherin by the polycomb group protein EZH2 in cancer. Oncogene, 2008. 27(58): p. 7274-84.

328. Wang, C., et al., Polycomb group protein EZH2-mediated E-cadherin repression promotes metastasis of oral tongue squamous cell carcinoma. Mol Carcinog, 2013. 52(3): p. 229-36.

Page 144: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

129

329. Avvakumov, N., A. Nourani, and J. Cote, Histone chaperones: modulators of chromatin marks. Mol Cell. 41(5): p. 502-14.

330. Ransom, M., B.K. Dennehey, and J.K. Tyler, Chaperoning histones during DNA replication and repair. Cell. 140(2): p. 183-95.

331. Huang, H., et al., Drosophila CAF-1 regulates HP1-mediated epigenetic silencing and pericentric heterochromatin stability. J Cell Sci. 123(Pt 16): p. 2853-61.

332. Chan, C.S., L. Rastelli, and V. Pirrotta, A Polycomb response element in the Ubx gene that determines an epigenetically inherited state of repression. EMBO J, 1994. 13(11): p. 2553-64.

333. Wang, L., et al., Hierarchical recruitment of polycomb group silencing complexes. Mol Cell, 2004. 14(5): p. 637-46.

334. Arnold, P., et al., Modeling of epigenome dynamics identifies transcription factors that mediate Polycomb targeting. Genome Res, 2013. 23(1): p. 60-73.

335. Brockdorff, N., Noncoding RNA and Polycomb recruitment. RNA, 2013. 19(4): p. 429-42.

336. Klose, R.J., et al., Chromatin sampling--an emerging perspective on targeting polycomb repressor proteins. PLoS Genet, 2013. 9(8): p. e1003717.

337. Ku, M., et al., Genomewide analysis of PRC1 and PRC2 occupancy identifies two classes of bivalent domains. PLoS Genet, 2008. 4(10): p. e1000242.

338. Deaton, A.M. and A. Bird, CpG islands and the regulation of transcription. Genes Dev, 2011. 25(10): p. 1010-22.

339. Lee, M.G., et al., Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science, 2007. 318(5849): p. 447-50.

340. Yuan, W., et al., Dense chromatin activates Polycomb repressive complex 2 to regulate H3 lysine 27 methylation. Science, 2012. 337(6097): p. 971-5.

341. Simon, J.A. and R.E. Kingston, Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol Cell, 2013. 49(5): p. 808-24.

342. Moldovan, G.L., B. Pfander, and S. Jentsch, PCNA, the maestro of the replication fork. Cell, 2007. 129(4): p. 665-79.

343. Hervouet, E., et al., Disruption of Dnmt1/PCNA/UHRF1 interactions promotes tumorigenesis from human and mice glial cells. PLoS One, 2010. 5(6): p. e11333.

344. Uwada, J., et al., The p150 subunit of CAF-1 causes association of SUMO2/3 with the DNA replication foci. Biochem Biophys Res Commun. 391(1): p. 407-13.

Page 145: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

130

345. Riising, E.M., et al., The polycomb repressive complex 2 is a potential target of SUMO modifications. PLoS One, 2008. 3(7): p. e2704.

346. Wu, H., et al., Dual functions of Tet1 in transcriptional regulation in mouse embryonic stem cells. Nature, 2011. 473(7347): p. 389-93.

347. Jin, B., et al., DNMT1 and DNMT3B modulate distinct polycomb-mediated histone modifications in colon cancer. Cancer Res, 2009. 69(18): p. 7412-21.

348. Zhao, J., et al., Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science, 2008. 322(5902): p. 750-6.

349. Plath, K., et al., Role of histone H3 lysine 27 methylation in X inactivation. Science, 2003. 300(5616): p. 131-5.

350. Corn, P.G., et al., E-cadherin expression is silenced by 5' CpG island methylation in acute leukemia. Clin Cancer Res, 2000. 6(11): p. 4243-8.

351. Graff, J.R., et al., Methylation patterns of the E-cadherin 5' CpG island are unstable and reflect the dynamic, heterogeneous loss of E-cadherin expression during metastatic progression. J Biol Chem, 2000. 275(4): p. 2727-32.

352. Long, H.K., N.P. Blackledge, and R.J. Klose, ZF-CxxC domain-containing proteins, CpG islands and the chromatin connection. Biochem Soc Trans, 2013. 41(3): p. 727-40.

353. Ridgway, P. and G. Almouzni, CAF-1 and the inheritance of chromatin states: at the crossroads of DNA replication and repair. J Cell Sci, 2000. 113 ( Pt 15): p. 2647-58.

354. Sirbu, B.M., et al., Analysis of protein dynamics at active, stalled, and collapsed replication forks. Genes Dev, 2011. 25(12): p. 1320-7.

355. Kliszczak, A.E., et al., DNA mediated chromatin pull-down for the study of chromatin replication. Sci Rep, 2011. 1: p. 95.

356. Leung, K.H., M.A. El Hassan, and R. Bremner, A rapid and efficient method to purify proteins at replication forks under native conditions. Biotechniques, 2013. 55(4): p. 204-6.

Page 146: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

131

Appendices

Apx1. The list of activator and inhibitor hits identified by the primary and secondary Wt and Suppressor screens.

Apx 1.1 The list of activator hits identified by the primary Wt screen. Listed below are the top 1% of the all activator hits, i.e., those which reduced luciferase activity after knock down, obtained with each method of B score, M score and control base normalization. See Fig 2.4.6 and text for details.

!

Page 147: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

132

Apx 1.2 The list of inhibitor hits identified by the primary Wt screen. Listed below are the top 1% of the inhibitor hits, those which induced luciferase activity after knock down, obtained with each method of B score, M score and control base normalization. See Fig 2.4.6 and text for details.

Page 148: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

133

Apx 1.3 The list of inhibitor hits identified by the primary Suppressor screen. Listed below are the top 1% of the inhibitor hits, those which induced luciferase activity after knock down, in the absence of BRG1, obtained with each method of B score, M score and control base normalization. See Fig 2.4.6 and text for details

Page 149: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

134

Apx 1.4 Secondary Wt screen, verified activator hits

Page 150: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

135

Apx 1.5 Secondary Wt screen, validated inhibitor hits

Apx 1.6 Secondary supp screen, validated inhibitor hits

Page 151: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

136

Apx2. List of SiRNAs

AllStarNegative QIAGEN 1027280 CHAF1A #1 Thermo Scientific L-019938-00 CHAF1A #2 QIAGEN SI03163699 CHAF1A #3 QIAGEN SI04219999 CHAF1A #4 QIAGEN SI04274914 PCNA#1 Thermo Scientific L-003289-00 PCNA#2 QIAGEN SI02653287 PCNA#3 QIAGEN SI02653357 PCNA#4 QIAGEN SI04436131 CHAF1B QIAGEN SI00077224 RBBP4 Thermo Scientific L-012137-00 POLD1 Thermo Scientific L-019687-00 POLE Thermo Scientific M-020132-01 HIRA Thermo Scientific L-013610-00 RAD9A Thermo Scientific M-003295-03 ARGONAUTE1 Thermo Scientific L-004638-00 ARGONAUT2 Thermo Scientific L-004639-00 DNMT1 Thermo Scientific L-004605-00 HP1a Thermo Scientific L-00429600 HP1b Thermo Scientific L-009716-00 HP1g Thermo Scientific L-010033-00 EHMT2(G9A) Thermo Scientific L-006937-00 SETDB1 Thermo Scientific L-020070-00 MBD1 Thermo Scientific L-008359-00

Page 152: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

137

Apx3. List of primers for RT-PCR!

Gene Region Chromosome position

Forward (5' to 3') Reverse (5'-3')

GBP2 Last exon AATGGCAGTTGTTTGACACTCTGA GCAAAATTTGCCTGTCCAGC

Promoter GTGTCAGGACCTGGGTTAGG CCAGTTTGAGAAGCATGTGG

GBP3 Last exon ATTACACAGTGCATAATTGTTACCATGT ACCTTAATGTGACACCTGAGACCTT

Promoter ACTGTGTGCTGGGCATTAAG GGAGGCATCACATTAATTCT

GBP4 Last exon ATATGCCATGGGCCTTTTCA CCGGGTTGTTTTAAGAAGCCT

Promoter AAACTCAAATTTCTTCCCTG AGACTCCTGAAACTCAGGTT

IFITM2 Last exon GAGTCCTGCATCAGCCCTTTA TGAATGCCATTGTAGAAAAGCG

Promoter CCAGCAAAGGACAAGGGATG AGGAGAATGGCGTGAACCGG

IRF1 Last exon AGGCCATTCCCTGTGCACCGTAGCA GTCCAGCTTCTCTGCACCATATCCA

Promoter GATCAGAGCTAGCCCACCCCTA GCCGTTGCAGGGTCTAATAG

IFI27 last exon TCTCCGGATTGACCAAGTTCA CAGGGAGCTAGTAGAACCTCGC

Promoter CCTGGTGCTTTCTCTTCCGC CCACAAAAGCACTGCAAGGA

6-16 last exon CAGCAGCGTCGTCATAGGTAAT TCCTCATCCTCCTCACTATCGAG

STAT1 Last exon CCTTATCACTGACACAAAAAGTAGATTAAGA ATAGTTGTGGTAGCAGTAGTGGAAAAAC

BPSB9 Last exon ATTGCTCTGGCCATGAGCC CCACACCGGCAGCTGTAATAG

HLA-E Last exon TTTGCAAGGGCCTCTGAATC TCCTCACATTGTGCTAACAGGG

HLA-G Last exon CTGAACTGCATTCCTTCCCC CAGCCCCTTTTCTGGAACAG

PITX2 Last exon AGGACTGCTGCCTTGTATGTTTAA TGGCCAGGACATCTCAGTCAC

MNDA Exon6 AACTTCGACTCTTCTGCCTTCAA GCTGTGACTTCCACACACCAGT

BRM Last exon GGGTGGGTCTAATTTGGTAA TGGCTTCTCCTGGTTATCAG

promoter CATCCCGCGCAGTTTCTCTG TGTCACTCGCTCAGCTCAGC

CIITA Last exon ACGTCTGACAGGCAATGCTG GGGTCCTAGCCAACTATTCCG

-70.1 kb TAAGAAATCTGTCTGTGGA TTCACAGAATTCCTCCGAAC

-50.6 kb CAGCTCATGTCCCACCCAGT AACAAACATGTCAGGCCACAGT

-16.4 kb TTCTGCAACTAGGTAACACC ATAGGTTGGATTACATGATC

-7.9 kb, PII AGTTGAACTGGCACATGGGC CTCTTGGAATTGGGAAGGCA

-0.3 kb, pIV TCACGGTTGGACTGAGTTGG CCTGAGTTGCAGGGAGCTTG

+10 GGACGTAATCTCAGCGCCTG TGTTAACGGCAACTCTGGGAG

+59.1 kb CAGCCTGTCCTCTTCTGCTCACA CGTGTTATACCCATGCCCTTGCAA

+82.6 kb TTAGAGAAAGGCACTGGATGGTCTGT GATACTTGTCTGTACACAGCCTAGCGG

Page 153: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

138

Apx3. List of primers for RT-PCR

!

!

!

!

!

!

!

!

!

!

!

!

Gene Region Chromosome position

Forward (5' to 3') Reverse (5'-3')

CHAF1A Last exon CCACTGGGTGCATCCTGAGA TACACTGCCTCGTTAGGTCCAGGG

PCNA Last exon CCAGTTCAACTAACTTTTGC CTGACTTTGGACTTTATTCT

CHAF1B Last exon TCCAGAAATATGGATGCTGTTG TGTGCACTTTCACGAGGATG

RbAp48 Last exon AGAATACTAAGGAATCCAGTTG AGAGGTTCAAGTTGGAGGTA

POLD Last exon CCGGGAGTCTGAGCTGTATC CTTGCGCATGTAGAAGATGG

POLE Last exon AATTTCACAGGGCACCAAAC GCGATGGCCTCAGACAGTAG

HIRA Last exon TTTCCTTTGGCCGATAATCA CATTCCCCCAAAACAGTCTC

RAD9A Last exon AGACTCCCAAGCGGCTCT CAGGTCGTGGGTCTCCAG

ARGONAUT1 Last exon AATGAAATTACTTTCCTGTGCACAC AGAGACATTTCCCCATCCAT

ARGONAUT2 Last exon TGAACTAAGGAGCAGTGGCAGA CCTATAGGACAAATCTGATG

DNMT1 Last exon AAGCTGTTGTGTGAGGTTCG TTCCACTCATACAGTGGTAGATTTG

HP1a Last exon TATGGGGGAGTTTTAGCTGT GCATCAGATGTCAGTTATGG

HP1b Last exon CTTTCATATTGGGCAGTGGT CGTAGCTCTAGATGCAAGTC

HP1g Last exon GCCTTTACAGTAGAAATAGAAATGC ACTGTGGAAACATTCCTGTG

EHMT2(G9A) Last exon CTGCCCCCTGTCAACACATG

SETDB1 Last exon CAGAACTTACTTGGGACTACA GACAGTTCAAGAAGGGTTGG

MBD1 Last exon CCTCAGACTCTTAATTATGC GTCAAACCAAATAACCAGTA

E-Cadherin Last exon GAACTCAGCCAAGTGTAAAAGCC GAGTCTGAACTGACTTCCGC

promoter CGCGCTAGCGCGGCCGCATGGCTCACACCTGAAA CGCAGATCTACCCTCTAGCCTGGAGTTGC

HPRT Last exon TTGCTCGAGATGTGATGAAGG TGATGTAATCCAGCAGGTCAGC

ACTIN Last exon TCCTAAAAGCCACCCCACTTCT GGGAGAGGACTGGGCCATT

Page 154: CHAF1A Regulates PRC2-mediated Epigenetic Memory · PDF fileChromatin and epigenetics ... Gene regulation by PRC2 and TrxG ... Fig 3.4.8 General effect of CHAF1A in PRC2-mediated gene

139

Apx4. List of Abs

!

Target protein Catalog Number source CHAF1A SC-28085 Santa Cruz PCNA SC-56 Santa Cruz B-ACTIN A-5441 Sigma b-Tubulin 2146 Cell Signalling Technology BRM 2146 Santa Cruz LAMINA/C SC-7293 Santa Cruz STAT1 SC-345 Santa Cruz IRF1 SC-497 Santa Cruz P-STAT1 9167S Cell Signalling g-H2X 05-636 Millipore SUZ12 ab12073 Abcam EZH2 3147S Cell Signalling Technology H3k27me3 07449 Millipore H3 ab1791 Abcam