this dissertation was sponsored by fundação para a ciência ... thesis mariluz... · trabalho...

180
This dissertation was sponsored by Fundação para a Ciência e Tecnologia. Apoio financeiro da FCT e do FSE no âmbito do Quadro Comunitário de apoio, BD nº SFRH/BD/33567/2008

Upload: ledat

Post on 05-Nov-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

This dissertation was sponsored by

Fundação para a Ciência e Tecnologia.

Apoio financeiro da FCT e do FSE no

âmbito do Quadro Comunitário de apoio,

BD nº SFRH/BD/33567/2008

Page 2: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório
Page 3: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

i

Declaraçao

Declaro que esta dissertação de candidatura ao grau de Doutor

é da minha autoria e que os dados aqui incluídos são o resultado de

trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril

de 2013 no laboratório do Dr Lars Jansen, Instituto Gulbenkian de

Ciência em Oeiras, Portugal. Este doutoramento foi realizado no

âmbito do Programa Doutoral do Instituto Gulbenkian de Ciência PGD

2008. Todas as colaborações estão indicadas em cada capítulo, na

secção de Acknowledgements. Esta dissertação teve o apoio

financeiro da FCT BD nº SFRH/BD/33567/2008 e dos projectos BIA-

BCM/100557/2008, BIAPRO/100537/2008 e EMBO instalação.

Do trabalho desenvolvido durante este período resultaram as

seguintes publicações:

Mariluz Gómez-Rodríguez and Lars E.T. Jansen. Basic properties of

epigenetic systems: lessons from the centromere. Current Opinion in

Genetics & Development. 23(2):219-227.

Dani L. Bodor, Mariluz Gómez Rodríguez, Nuno Moreno and Lars

E.T. Jansen. Analysis of protein turnover by quantitative SNAP-based

pulse–chase imaging (2012). Current Protocols in Cell Biology

55:8.8.1.-8.8.34.

Jan H. Bergmann, Mariluz Gómez Rodríguez, Nuno M.C. Martins,

Hiroshi Kimura, David A. Kelly, Hiroshi Masumoto, Vladimir Larionov,

Lars E.T. Jansen, and William C. Earnshaw (2011). Epigenetic

engineering shows H3K4me2 is required for HJURP targeting and

Page 4: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

ii

CENP-A assembly on a synthetic human kinetochore. Embo Journal

30, 328–340.

Page 5: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

iii

Acknowledgements

I would like to start with my supervisor, Professor Lars Jansen.

Dear Lars, I am thankful for the opportunity to work in your group. I

could not be more pleased by having the challenge to work on a

fundamental question with an excellent and fantastic professor like

you. Your support, confidence, motivation, demand, guidance,

leadership are attributes that immensely contributed to this process.

Thanks a lot!

I thank the past and present members of EpiLab: Nuno,

Mariana, João, Luis, Ana and Ana Filipa. Dani, the criticisms and very

importantly: your willingness to help out whenever, were very important

during these years, thank you. I do also want to thanks the group of

evaluation of the program (PGD2008) professors: Antonio Coutinho,

Henrique Teotónio, Mónica Dias and Moises Mallo for your welcome,

demands and feedback.

There is a group that I define as: aid service for graduate

student in troubles. Who are they? Cell Cycle Regulation, Plant Stress

Signaling and Plant Molecular Biology groups, together with Zeng Ho

wing’s technician: Paulo Duarte and Sónia Rosa. Hope you guys have

lost count of the number of times that I bothered for help. But, that is

the consequence for being kind, patience, hard workers, enthusiastic

and cool. Many thanks guys! Besides the people at Zeng Ho wing,

especially to the ones that often brought joy like Gaston, Zita, Ana Rita

and Filipe.

Special thanks to Claudia, Raquel, Vera, Inês, Thiago, Pierre,

Céu and Ana Luisa for your invaluable help but most of all your

friendship. Your friendship was a gift, I couldn’t be luckier during these

years by having you around.

Page 6: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

iv

Gracias a mi familia su apoyo, sus palabras de aliento, su

comprensión, su generosidad, su oración; a ustedes: aquí estamos,

para nosotros y por nosotros.

Al otro lado del Atlántico de Sur a Norte, en Europa y Asia:

Jimmy, Sandra, Olga Lucía, Erick, Luis Alfredo, Jose Edwin, Marta,

Vivianne, Saeeda, Sanjay, Nancy, Martalucía, Juan Carlos, Genoveva,

Fanny, Mónica, Alejandro, y la alma máter: Pontificia Universidad

Javeriana, especialmente a la Vicerrectoria Académica; a todos

ustedes muchas gracias.

Page 7: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

v

Abstract

Cell proliferation and differentiation into distinct cell types

during development requires the preservation of cellular phenotypes

during cell divisions. How the cell maintains its identity and how cells

with the same genetic information display different heritable

phenotypes or gene expression profiles are fundamental questions in

biology. The mitotic and/or meiotic inheritance of changes in gene

expression that are not due to changes in DNA sequences is known as

epigenetic inheritance. The transmission of the epigenetic state is

dependent on molecular markers that are predicted to be transmissible

across cell divisions, have the capacity to template their own

duplication and propagate under control of the cell cycle. There are a

range of markers that control gene expression in cis involved in

epigenetic phenomena, including DNA or histone modifications, DNA

or histone binding proteins and histone variants. Although histone

modifications are widely studied, their inheritance and potential

underlying mechanism of propagation are controversial. A key aspect

to elucidate the role of histone proteins and their modification as an

epigenetic mark is the dynamic equilibrium between histone turnover

and the dynamics of the modifications of those histones. The basic unit

of chromatin is the nucleosome. The nucleosome is formed by an

octamer of four core histones (H2A, H2B, H3 and H4) wrapped by

147bp of DNA. With the exception of H4 all histones have variants and

their preferential localization serves different roles in preserving

epigenetic identity.

The major H3 variants are H3.3 and centromere protein A

(CENP-A). Unlike canonical H3.1, H3.3 is deposited at promoters,

sites of active transcription, enhancers and subtelomeric regions

throughout the cell cycle; whereas CENP-A is deposited into

centromeric chromatin during G1 phase. The dynamics of histones has

Page 8: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

vi

been analyzed using powerful methods that include metabolic pulse

labeling of nascent proteins, fluorescence recovery after

photobleaching, conditional expression of tagged histones and the use

of self-labeling tags. Nevertheless, methods for quantitative histone

turnover at specific loci have been lacking. In chapter 2, I describe the

development of TimeChIP, a pulse labeling strategy coupled to

chromatin precipitation that provides temporal information of histone

occupancy at high resolution. We show that using the self-labeling

SNAP-tag, we can isolate pulse labeled histones at different times

following their labeling for the analysis of their dynamics. This method

provides temporal resolution, distinction between old and new pools of

proteins and high spatial resolution in living cells. We employ this

method for the analysis of CENP-A in proof of principle experiments

and show that CENP-A is stably maintained in centromeric chromatin

and assembled in nucleosomes following its targeting in G1 phase.

In chapter 3, we used the TimeChIP strategy to determine to

what extend histones H3 variants H3.1 and H3.3 can be maintained at

different loci of mitotically dividing cells. We find that canonical H3.1 is

locally retained at genes and non-gene loci, whereas H3.3 exhibits a

faster turnover. Genome-wide analysis demonstrates that H3.3

turnover at active genes correlates with the rate of transcription.

However, there is a strikingly high degree of histone retention of both

H3.1 and H3.3 for the duration of the cell cycle, even at gene bodies of

highly transcribed genes. Moreover, we find that transcription

activation at an inducible gene locus does not lead to faster histone

turnover. Conversely, inhibition of RNA polymerase II leads to an

increased retention of H3.1 and H3.3 during the cell cycle.

This work describes the development of a versatile strategy for

the analysis of histone dynamics: turnover and assembly. We used this

method to demonstrate for the first time that while subsets of H3.1 and

Page 9: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

vii

H3.3 nucleosomes are dynamic at active gene loci, a significant pool is

retained in cis during the extent of a cell cycle. Our findings are

consistent with a model in which nucleosomes contribute to the

inheritance of epigenetic information and make predictions about the

dynamics and retention of histone modifications.

Page 10: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

viii

Sumário

A proliferação e diferenciação das células durante o

desenvolvimento requerem a preservação dos fenótipos celulares

durante a divisão celular. O modo como uma célula mantém a sua

identidade e o modo como células com a mesma informação genética

conseguem exibir diferentes fenótipos hereditários ou diferentes perfis

de expressão génica são questões fundamentais em biologia. A

herança epigenética é definida como a hereditariedade mitótica e

meiótica de alterações na expressão dos genes que são

independentes da sequência de DNA. A transmissão do estado

epigenético é dependente de marcadores moleculares que se

esperam ser transmissíveis através das divisões celulares, que têm a

capacidade de servir de modelo para a sua própria duplicação e cuja

propagação está dependente do controlo do ciclo celular. Existem

uma série de marcadores que controlam a expressão génica em cis

envolvidos em fenómenos epigenéticos tais como modificações de

DNA ou de histonas, proteínas de ligação ao DNA ou a histonas e

variantes de histonas. Apesar das modificações em histonas serem

amplamente estudadas, a sua herança e o possível modelo

subjacente à sua transmissão hereditária são ainda muito

controversos. De modo a elucidar o papel das histonas e suas

modificações enquanto marca epigenética é fundamental perceber o

equilíbrio entre o seu turnover e a dinâmica das suas modificações. A

unidade básica da cromatina é o nucleossoma que consiste em 147

pares de bases de ADN enrolados em redor de um octâmero de

quatro histonas centrais (H2A, H2B, H3 e H4). Com a excepção da

H4, todas as histonas têm variantes e as suas diferentes localizações

determinam papéis distintos na preservação da identidade

epigenética.

Page 11: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

ix

As principais variantes da histona H3 são as H3.3 e a proteína

A do centrómero (CENP-A). Ao contrário da proteína canónica H3.1,

a H3.3 é depositada em promotores, em locais de transcrição activa e

em regiões intensificadoras e subteloméricas durante todo o ciclo

celular. Por sua vez, a proteína CENP-A é depositada na cromatina

centromérica durante a fase G1. Neste estudo, a dinâmica das

histonas foi analisada utilizando métodos poderosos que incluem

pulse labeling (marcação com recurso a compostos especiais)

metabólico de proteínas nascentes, recuperação de fluorescência

após a fotodegradação, expressão condicional de histonas com tags e

a utilização de self-labeling tags. No entanto, não foram possíveis

métodos para quantificar o turnover de histonas em loci específicos.

No capítulo 2, descrevemos o desenvolvimento do método TimeChIP,

uma estratégia de pulse labeling associada à precipitação da

cromatina, que fornece informação temporal em alta resolução da

localização de histonas. Mostramos que o uso da self-labeling SNAP-

tag permite isolar histonas marcadas em diferentes tempos, seguindo

a sua marcação de modo a analisar a sua dinâmica. Este método

permite distinguir entre pools novos e antigos de proteínas,

oferecendo alta resolução temporal e espacial em células vivas. Este

método foi aplicado para a análise da CENP-A, onde mostramos que

esta proteína se encontra estável na cromatina centromérica e é

incorporada em nucleossomas após a sua focalização na fase G1.

No capítulo 3, descrevemos a utilização do método TimeChIP

para determinar até que ponto as variantes de histonas H3, H3.1 e

H3.3, podem ser mantidas em diferentes loci de células em mitose. Os

nossos resultados mostram que a proteína canónica H3.1 é mantida

localmente em loci génicos e não-génicos, enquanto que a H3.3

apresenta um turnover mais rápido. Uma análise genómica em larga

escala mostra que o turnover da H3.3 em genes activos se

Page 12: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

x

correlaciona com a taxa de transcrição. No entanto, existe um elevado

grau de retenção de ambas variantes (H3.1 e H3.3) durante todo o

ciclo celular, mesmo em conjuntos de genes altamente transcritos.

Adicionalmente, descobrimos que a activação da transcrição de um

locus de um gene indutível não resulta num turnover mais rápido das

histonas. Por outro lado, a inibição da RNA polimerase II leva a um

aumento da retenção das variantes H3.1 e H3.3 durante o ciclo

celular.

Este trabalho descreve o desenvolvimento de uma estratégia

versátil para a análise da dinâmica de histonas: seu turnover e

montagem (assembly). A utilização deste método permitiu mostrar

pela primeira vez que pequenos conjuntos de nucleossomas

compostos por H3.1 e H3.3 são dinâmicos em loci de genes activos

mas que uma parte significativa é retida em cis durante a extensão de

um ciclo celular. Os nossos resultados estão de acordo com um

modelo em que os nucleossomas contribuem para a herança de

informação epigenética e permitem fazer previsões sobre a dinâmica e

a retenção de modificações das histonas.

Page 13: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xi

List of Abreviations

ACTB β-Actin

AHA Azidohomoalanine

ATRX α-thalassemia X linked mental retardation protein

BG O6-benzylguanine

CAF1 Chromatin Association Factor-1

CATCH-IT Covalent Attachment of Tags to Capture Histone and

Identify Turnover

CATD CENP-A Targeting Domain

CCAN Constitutive Centromere Associated Network

CCNA Cyclin A

CDK Cyclin Dependent Kinase

CENP-A Centromeric Protein A

ChIP Chromatin Immunoprecipitation

CiA Chromatin in vivo Assay

DAXX Death Associated Protein

FPALM Fluorescence Photoactivation Localization Microscopy

FRAP Fluorescence Recovery After Photobleaching

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

hAGT human O6-Alkylguanine-DNA Alkyltransferase

HFD Histone Fold Domain

Page 14: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xii

HIRA Histone Regulator A complex

HJURP Holliday Junction Recognizing Protein

LacO Lac Operon

lncRNAs long non coding RNAs

MS Mass Spectrometry

MYC Myc proto–oncogen protein

MyoD Myoblast Determination protein

ncRNA non-coding RNAs

NF-ƙβ Nuclear Transcription factor-ƙβ

PAFPs Photoactivatable Fluorescent Proteins

PCNA Proliferating Cell Nuclear Antigen

PRC Polycomb Repressive Complex

PRE Polycomb Response Elements

RITE Recombination-Induced Tag Exchange

RPKM Reads per kilobase per million

RPL13A Ribosomal protein L13a

RPLP0 Ribosomal protein P0

Sat2 Pericentromeric Satellite 2 repeats

SILAC Stable Isotope Labeling of Amino Acids in Cell Culture

TBP TATA binding protein

Page 15: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xiii

TrxG/PcG Trithorax and Polycomb protein

TSS Transcription Start Site

TTS Transcription Termination Site

XCi X Chromosome Inactivation

Page 16: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xiv

Page 17: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xv

Table of Contents

Declaraçao ........................................................................................... i

Acknowledgements ............................................................................. iii

Abstract ............................................................................................... v

Sumário ............................................................................................ viii

List of Abreviations ............................................................................. xi

Table of Contents .............................................................................. xv

Chapter 1 – General Introduction .........................................................1

1. Epigenetic inheritance ...............................................................3

1.1.1 Epigenetic Inheritance in trans .............................................4

1.1.2 Epigenetic Inheritance in cis ................................................5

1.1.2.1 DNA methylation ...............................................................8

1.1.2.2 Polycomb proteins ............................................................9

1.1.2.3 Histone modifications ...................................................... 11

1.1.2.4. X chromosome inactivation ............................................ 16

1.1.2.5. Histone variants ............................................................. 17

1.1.2.5.1 CENP-A ....................................................................... 18

1.1.2.5.2. H3.3 ............................................................................ 23

2. Measuring protein dynamics .................................................... 25

2.1 Fluorescence recovery after photobleaching (FRAP) ............ 25

2.2 Inducible fluorescent proteins ............................................... 27

2.3 Self labeling tags .................................................................. 28

2.4 Recombination-induced tag exchange (RITE) ....................... 29

2.5 Metabolic labeling of proteins ................................................ 30

Page 18: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xvi

2.5.1 Stable isotope labeling by amino acids in cell culture

(SILAC) ....................................................................................... 30

2.5.2. Covalent attachment of tags to capture histone and identify

turnover (CATCH-IT) .................................................................. 32

3. Aims of this thesis ................................................................... 34

References ..................................................................................... 35

Chapter 2 – TimeChIP ....................................................................... 49

AUTHOR CONTRIBUTION ............................................................ 51

SUMMARY ..................................................................................... 51

INTRODUCTION ............................................................................ 52

MATERIALS AND METHODS ........................................................ 59

Cell lines and constructs ............................................................. 59

DNA transfections ....................................................................... 59

Cell synchronization .................................................................... 60

SNAP quench-chase-pulse labeling ............................................ 60

Soluble Nucleosome Preparation ................................................ 61

SNAP-Nucleosomes Purification ................................................. 61

Quantification of bound nucleosomes ......................................... 62

Immunofluorescence .................................................................. 63

Microscopy ................................................................................. 63

Immunoblotting ........................................................................... 64

Flow cytometry............................................................................ 64

RESULTS ...................................................................................... 65

TimeChIP strategy ...................................................................... 65

Proof of Principle ........................................................................ 72

Page 19: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xvii

Pulse-chase with TimeChIP ........................................................ 74

Quench-chase-pulse with TimeChIP ........................................... 75

DISCUSSION AND CONCLUSIONS ............................................. 81

CENP-A nucleosome dynamics along the cell cycle ................... 83

ACKNOWLEDGEMENTS ............................................................... 86

REFERENCES ............................................................................... 87

Chapter 3 – H3.1 and H3.3 Turnover ............................................... 103

AUTHOR CONTRIBUTION .......................................................... 105

SUMMARY ................................................................................... 105

INTRODUCTION .......................................................................... 106

MATERIALS AND METHODS ...................................................... 109

SNAP labeling and drug treatments .......................................... 109

Immunofluorescence ................................................................ 110

Microscopy ............................................................................... 111

Flow cytometry.......................................................................... 111

Next-generation sequencing ..................................................... 111

RNA extraction and qRT-PCR .................................................. 112

RESULTS .................................................................................... 113

Quantitative retention of ancestral H3.1 and H3.3 ..................... 113

Local retention of H3.1 .............................................................. 118

Genomic distribution and turnover of H3.3 ................................ 122

Differential histone retention within active gene ........................ 124

Dynamics of H3.1 and H3.3 during transcription activation ....... 125

Dynamics of H3.1 and H3.3 during transcription inhibition ........ 128

Page 20: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

xviii

DISCUSSION ............................................................................... 132

ACKNOWLEDGEMENTS ............................................................. 139

REFERENCES ............................................................................. 140

Chapter 4 – General Discussion ...................................................... 147

Can qualitative differences between nucleosomes result in

differential stability? ...................................................................... 150

Transcription drives histone turnover ............................................ 152

The role of histones as epigenetic markers .................................. 154

References ................................................................................... 158

Page 21: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

1

Chapter 1 – General Introduction

Page 22: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

2

Page 23: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

3

1. Epigenetic inheritance

The term epigenetics has had different meanings throughout the

last half century. Generally speaking the concept of epigenetics refers

to heritable changes that occur independent of genetic changes.

Conrad H. Waddington coined the term (1942) to refer to the study of

“causal mechanisms” by which the genes of the genotype bring about

phenotypic effects during development. Later, D.L. Nanney introduced

the term epigenetic control systems as “auxiliary mechanisms with

different principles of operation that are involved in determining which

specificities are to be expressed in any particular cell”, and that such

systems were “presumably limited by the information contained in the

genetic library…”An epigenetic change should not result in a

permanent loss of information and a return to a previous condition of

expression is always theoretically possible” (Haig, 2004). Currently

epigenetics is more strictly defined as “the study of mitotically and/or

meiotically heritable changes in gene function that cannot be explained

by changes in DNA sequences”

Maintenance of epigenetic information relies on the ability to

persist through cell divisions in the absence of the initial inducing

signals. The ability of an epigenetic mechanism to sustain itself is

characterized by: 1) stability, where the agent responsible for the

transmission of the information is expected to self maintain across the

cell cycle; 2) self-duplication, the ability to copy information into novel

structures so as to maintain cellular information through cell division

and growth; and 3) cell cycle control, a temporal regulation of the

propagation of the epigenetic system (Gómez-Rodríguez and Jansen,

2013). Mechanistically, an epigenetic state can in principle be self-

sustainable by trans-acting or cis-acting molecular signatures.

Page 24: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

4

1.1.1 Epigenetic Inheritance in trans

Trans-acting epigenetic signals are maintained by positive feed-

back mechanisms involving transcription factors and non-coding RNAs

(ncRNAs). In the case of transcription factors, there are many

examples; classic cases include the phage lambda and the cell type

specific master regulator MyoD in eukaryotes. The phage lambda

codes for two transcription factors, Cro and lambda repressor.

Although the two proteins are not homologous at the amino acid

sequence level, they recognize the same operator sites. Binding of one

or the other, leads to two physiologically distinct states, lysogeny and

lytic growth. Cro autoregulates its own production positively while the

lambda repressor does so positively and negatively. The bi-regulation

of the repressor is given by the cooperative binding of its dimers to

adjacent sites generating a state poised to respond to a transient

signal and switch state. The epigenetic nature of the binary phage

lambda states comes from the positive feedback loop of either the

repressor or the Cro transcription factor that activates expression of its

own gene and is stable for many cell divisions as the respective

transcription factor is distributed to daughter cells. (Johnson et al.,

1978, 1981; Ptashne et al., 1976). Similarly, the basic-helix-loop-helix

(bHLH) transcription factor MyoD binds to private E boxes of target

genes and induces myogenesis in a wide variety of cell types in

mammals. Importantly, in addition to its role as an activator of

downstream genes, it functions in a positive autoregulatory loop

increasing the levels of its own expression thereby stabilizing

myogenic commitment (Chanoine et al., 2004; Fong et al., 2012;

Lassar et al., 1989; Thayer et al., 1989).

A growing number of ncRNAs are found to be involved in

epigenetic phenomena but the underlying mechanisms are largely

unknown. For instance, in plants, Dicer like protein 3 (DCL-3)

Page 25: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

5

generates mobile 24 – nucleotide (24-nt) sRNAs (Molnar et al., 2010).

24-nt sRNA moves between cells directing RNA-dependent DNA

methylation in the recipient cell that control transcriptional gene

silencing of transposon elements from three separate genomic loci

(Melnyk et al., 2011; Molnar et al., 2010).

1.1.2 Epigenetic Inheritance in cis

Epigenetic states that are maintained in cis are typically

chromatin based. Chromatin is a dynamic DNA - protein complex in

which the genetic information is packaged inside eukaryotes cells. Its

organization directly impacts on control of gene expression. Chromatin

as a high order structure shows different levels of organization: 1) the

basic unit of chromatin is the nucleosome which is formed by an

octamer of four core histones (H3, H4, H2A and H2B) wrapped with

147bp of DNA and separated by a 10-80bp linker DNA associated with

linker histone H1. Together, this complex forms a 10nm diameter fiber,

2) in vitro, a helical fiber of 30nm in diameter has been observed

containing 6-11 nucleosomes per turn. 3) chromatin fibers that interact

within or between larger fibers (Higher order in vivo chromatin has

been described as a “polymer melt” state where nucleosomes interact

across chromatin regions and not in a linear-fashion between

neighbors), 4) chromosome territories and 5) chromosomes (de Graaf

and van Steensel, 2013; Hübner et al., 2013) (Figure 1.1)

Figure 1.1 Chromatin organization in the mammalian nucleus. a)

Chromosomes are organized in chromosome territories. b) Chromosome

Page 26: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

6

territories are comprised of so-called fractal globules that interact in cis or can

interdigitate with adjacent chromosome territories. c) Chromatin fibers interact

(i) within a fractal globule, (ii) between fractal globules of the same

chromosome territory, or (iii) between adjacent chromosome territories. d)

Chromatin may form a 30nm fiber. e) Chromatin is resolved as a 10nm beads

on a string fiber consisting of nucleosomes. Adapted from Hübner et al., 2013.

The accessibility to the DNA is, at its most basic level,

determined by the nucleosome. The octamer is arranged along a

twofold dyad symmetry axis, the intersection point with the middle of

DNA fragment. The typical histone protein features three α-helices (α1,

α2 and α3) separated by two loops (L1 and L2) that constitute the

histone fold domain (HFD) and an amino-tail. The HFDs fold together

in antiparallel pairs: H3 with H4 and H2A with H2B. The assembly of

the eukaryotic nucleosome begins with an (H3-H4)2 tetramer, held

together by a strong four-helix bundle between the two H3 molecules,

which is followed by the addition of two H2A-H2B dimers that form

weak four-helix bundles between H4 and H2B (Luger et al., 1997;

Talbert and Henikoff, 2010).

Figure 1.2 Architecture of nucleosome core particle. The four histone

dimers H3, H4, H2A and H2B are colored in blue, green red and yellow,

Page 27: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

7

respectively. The four histone dimers are arranged about a twofold dyad

symmetry axis, which also intersects the middle of the DNA fragment.

Adapted from Biswas et al., 2011.

The canonical histones, that are among the slowest evolving

proteins known, are encoded by gene clusters whose expression is

tightly coupled to DNA replication. DNA replication is highly regulated

and occurs in two steps: licensing and initiation. During the licensing

step, the prereplicative complex (pre-RC) is formed at multiple origins

of replication in G1 phase, where it loads ORC, Cdc6, Cdt1 and the

inactive replicative helicase Mcm2-7 complex (Remus and Diffley,

2009). In the initiation step, active helicase containing the CMG

(Cdc45-Mcm2-7-GINS) complex unwinds the double-stranded DNA for

the loading of DNA polymerases. The initiation step is regulated by the

S phase specific cyclin-dependent kinase (S-CDK) and Cdc-7-Dbf4-

dependent kinase (DDK) (Tanaka and Araki, 2010). Assembly of newly

synthesized genomic DNA requires large amounts of histones

produced during S phase and ancestral histones that are transferred

from the parental strand to the daughter strands (Groth et al., 2007;

Osley, 1991). Assembly of (H3-H4)2 tetramers is mediated by the

histone chaperone chromatin assembly factor-1 (CAF-1) that binds to

the proliferating cell nuclear antigen (PCNA) (Shibahara and Stillman,

1999) and followed by deposition of two H2A-H2B dimers to complete

the nucleosome.

Cis epigenetic states that are propagated in chromatin are

maintained by so-called epigenetic markers that include DNA

methylation, histone variants and their modifications, DNA or histone

binding proteins and ncRNAs.

Page 28: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

8

1.1.2.1 DNA methylation

Cytosine DNA methylation is a stable and heritable long term

silencing mark important for processes such as gene and transposon

silencing, imprinting and X chromosome inactivation in most of

eukaryotes. There are three different nucleotide sequence context

where DNA methylation has found to occur: symmetrically in CG and

CHG, and asymmetrically in CHH (where H= C, T or A). In mammals

DNA methylation occurs symmetrically in the CG context. The majority

of CG dinucleotides across the genome are methylated except for high

density CG regions called CpG islands at active promoters that are

typically unmethylated. The unmethylated state is important for gene

activation. The acquisition of methylation during development results in

the establishment of long term repression (Bird, 2002; Lee et al.,

2010). The establishment is driven by the DNA methyltransferases

DNMT3A and DNMT3B during the blastocyst stage and re-

establishment in gametogenesis of embryonic development (Law and

Jacobsen, 2010).

DNA methylation is semiconservatively transmitted during the cell

cycle because of the nature of DNA replication. A DNA sequence

carrying symmetrical methylation marks on both strands gives rise to

two hemi-methylated double strands. The DNA methyl transferase

DNMT1 methylates new CpGs during DNA replication at sites where

the parental strand already carries a methyl group. DNMT1 is recruited

by a ubiquitin-like plant homeodomain and RING finger domain 1

(UHRF1) to hemimethylated DNA and through a transient interaction

with proliferating cell nuclear antigen (PCNA) associates with

replication machinery, where PCNA enhances the efficiency of the

catalytic activity of DNMT1 (Bird, 2002; Law and Jacobsen, 2010;

Schermelleh et al., 2007; Spada et al., 2007). Consequently, DNMT1

is responsible for the heritable maintenance of DNA methylation in a

Page 29: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

9

reading and writing manner. This occurs during S phase through the

UHRF1 interaction as well as in G2 and M phases (Easwaran et al.,

2004). DNMT1 is positively regulated along the cell cycle by CDK1, 2

and 5 which phosphorylate Ser154 in humans (Lavoie and St-Pierre,

2011).

1.1.2.2 Polycomb proteins

Another important class of factors implicated in carrying epigenetic

states are non-histone chromatin-binding proteins. Among the most

extensively studied are the Trithorax and Polycomb (TrxG/PcG) protein

family. TrxG and PcG were originally identified in D. melanogaster due

to their roles in tuning HOX genes expression by promoting histone

modifications (Simon and Kingston, 2009). In flies, TrxG and PcG

associate to chromatin by complex DNA elements named Trithorax

and Polycomb response elements (TRE/PRE), which are located

kilobases from the promoters they control. These sites are depleted of

nucleosomes and form looping interactions with their target repressed

genes in the case of PcG and non repressed genes in Trx in

Drosophila (Petruk et al., 2012; Simon and Kingston, 2013). Although,

TrxG and PcG were initially found on HOX genes, approaches like

genome wide chromatin immunoprecipitation (ChIP) studies have

identified target genes besides HOX in Drosophila and mammals

(Simon and Kingston, 2009).

PcG proteins are involved in silencing of genes in a non-

constitutive conditional manner. In development, PcG is implicated in

holding developmentally controlled genes in a poised but inactive state

in mouse and human embryonic stem cells (ES). The system is

composed of many complexes, Polycomb repressive complex 1

(PRC1), PRC2, PRC1 variant, PHO-RC and PR-RUB, of which the

best characterized are PRC1 and PRC2. PRC1 silences target genes,

in part, by monoubiquitylation of histone H2AK119u1 (Wang et al.,

Page 30: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

10

2004), that inhibits the recruitment of FACT, blocking RNA polymerase

II release at an early stage of elongation (Zhou et al., 2008) and

creating a compacting state in chromatin. Experiments using the

mammalian SV40 replication system show that PRC1 is maintained on

the template after replication fork passage and binds tightly to single

stranded DNA (Francis et al., 2009; Lo et al., 2012) which may

contribute to the heritable nature of PRC1 mediated silencing.

PRC2 carries the major methyltransferase for H3K27 methylation,

a modification critical for maintaining repressed gene expression

programs throughout development (Pengelly et al., 2013).

The mechanism for PRC2 targeting has not been elucidated,

except the presence of Polycomb response elements (PRE) located

many kilobases from the promoters they control in Drosophila. In

mammals, recruitment is even more poorly defined. In addition to DNA

elements similar to PREs, unmethylated CpG islands and lncRNAs are

believed to target PcG functions in mammals (Simon and Kingston,

2013). Once, PRC2 is recruited to chromatin, the presence of pre-

existing H3K27me3 marks on neighboring nucleosomes activates the

methyltransferase activity of PRC2 contained in the E(z)/Ezh2 subunit,

to carry out further methylation on unmethylated H3K27 (Hansen et al.,

2008; Margueron et al., 2009; Xu et al., 2010a). This suggest that

PRC2 self-replicates by acting as adaptor that reads (recognize) and

writes (methylates) its own signal to be sustained (Gómez-Rodríguez

and Jansen, 2013).

In vivo FRAP experiments in Drosophila show that Pc and Ph,

members of the PRC2 complex, have a relative short residence time of

2-6 minutes, suggesting that PcG complexes are highly dynamic

where the chromatin state is inherited in a dynamic fashion rather than

a stable fashion (Ficz et al., 2005). In vivo experiments in Drosophila

Page 31: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

11

embryos show that Trx, PRC1 and PRC2 associate transiently with

PCNA, indicating that these PcG proteins are maintained during S

phase (Petruk et al., 2012). Regarding maintenance of PcG proteins

during mitosis, evidence comes from FRAP studies in Drosophila stem

and differentiated cells. PcG proteins binds mitotic chromatin with up to

300 fold longer residence times than in interphase potentially

contributing to the mitotic propagation of the silent state. This retention

is governed by phosphorylation of H3 at Ser 38 (H3K27me3Ser38)

(Fonseca et al., 2012).

The PRC2 complex is cell cycle regulated at its catalytic subunit.

Ezh2 contains an evolutionally conserved consensus CDK

phosphorylation motif. Phosphorylation at this motif correlates with the

oscillatory activity of CDK1 and 2 during the cell cycle, without

affecting assembly and histone methyltransferase (HMT) activity of

PRC2 (Chen et al., 2010), thereby CDK1 and 2 act as positive factors

for the maintenance of PRC2.

1.1.2.3 Histone modifications

In addition to the above mentioned histone methylation events,

other histone modifications contribute to chromatin dynamics and

epigenetic states. Histone modifications, either on the surface of the

DNA wrapped core of the nucleosome or on the amino-terminal tail

that extends from the nucleosome surface, have been described

(Table 2). They can be functionally categorized in: 1) modifications that

disrupt chromatin structure. These include negatively charged

acetylation and phosphorylation that neutralize the positively charged

histones thereby generating repulsion between histones and DNA.

This results in the relaxation of histone-DNA contacts for processes

like transcription, replication and DNA repair. In contrast, deacetylases

have the ability to induce condensation of chromatin important for

chromosome segregation. 2) Regulation of chromatin binding factors

Page 32: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

12

that act as effector proteins of the modification. Such factors are

characterized by Chromo, Tudor, MBT, PhD finger domains that bind

to methyl-lysine modifications, Bromo domains bind to acetyl-lysine

modifications and 14-3-3 domains bind phosphorylated H3S10.

Numerous domains can recognize the same histone modification and

create binding platforms for various factors (Bannister and Kouzarides,

2011; Goutte-Gattat et al., 2013; Kouzarides, 2007; Zentner and

Henikoff, 2013).

Table 1. Different Classes of Modifications Identified on Histones

Chromatin Modifications

Modified Residues

Regulated Functions

Acetylation K-ac Transcription, repair, replication, condensation

Methylation (lysines) K-me1,K-me2,K-me3

Transcription, repair

Methylation (arginines) R-me1, R-me2a, R-me2s

Transcription

Phosphorylation S-ph, T-ph Transcription, repair, condensation

Ubiquitinylation K-ub Transcription, repair Sumoylation K-su Transcription ADP ribosyaltion E-ar Transcription Deimination R > Cit Transcription Proline Isomerization P-cis > P-trans Transcription Glycosylation T-OGlcNAc S-

OGlcNAc Transcription

Histone tail clipping Transcription Modified from Kouzarides, 2007.

A large number of histone modifications are associated with

epigenetic phenomena; nevertheless, few cases have been defined as

heritable epigenetic markers. The methyl marks H3K9, H3K27 and

H4K20 are involved in constitutive heterochromatin formation, gene

silencing, DNA damage repair and mitotic chromosome condensation.

All three marks come in three different flavors (mono, di or tri-

methylation) that have different functional consequences, the tri-

methylated state being the most strongly associated with the silent

chromatin state. These marks have a slow turnover rate compared

Page 33: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

13

with acetylation and phosphorylation and are marks postulated to be

epigenetic in the sense that they drive an in cis heritable chromatin

state (Barth and Imhof, 2010; Zee et al., 2010).

Table 2. Turnover rates for histones carrying different post translational modifications

Histone Modification Half-life Acetylation All histones <15min H2A 80% 2-3 min H2B 75% 3 min, 20% 40 min H3 60-70% 3 min, 25% 30 min H4 50% 2-3 min, 45% 40 min Phosphorylation H1 Short pulse: 3h H2A 60% 40 min; fast, 30 min; slow H2B Not determined H3 Fast, 30 min; slow, 2-3h H4 Fast, 30 min; slow, 2-3h Methylation Half-life (days) Overall H3 1.298 +/- 0.007 H3K4me1 0.959 +/- 0.129 H3K9me1 0.342 +/- 0.001 H3K9me2 1.031 +/- 0.060 H3K18me1 1.207 +/- 0.116 H3K27me1 0.470 +/- 0.005 H3K27me2 1.145 +/- 0.001 H3K27me3 3.128 +/- 0.032 H3K36me1 0.751 +/- 0.085 H3K36me2 0.571 +/- 0.000 H3K79me1 1.105 +/- 0.070 H3K79me2 3.609 +/- 0.283 Overall H4 1.385 H4K20me1 0.297 +/- 0.005 H4K20me2 1.467 +/- 0.001 H4K20me3 4.809 +/- 1.483 H4R3me1 2.788 +/. 1.806 Overall H1.4 0.976 +/- 0.065 H1.4K25me1 1.294 +/- 0.166 From Barth and Imhof, 2010.

H4K20 methylation, an evolutionarily conserved chromatin

modification, is linked with repression of transcription, DNA damage

repair and X inactivation in mammals. Initial methylation of H4K20 is

mediated by SET8/PRSet7, whereas further H4K20 methylation to

H4K20me2 and H4K20me3 is performed by SUV4-20H1 and SUV4-

Page 34: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

14

20H2 enzymes (Jørgensen et al., 2013). The methyltransferase Set8

has been found to be associated with chromatin and/or the PCNA

replication clamp during replication (Huen et al., 2008). H4K20me1 is

present throughout the cell cycle but PRSet7, which is regulated during

the cell cycle, has been detected only during G2 and early M phase.

This indicates that the presence of the methyl mark does not need the

continued presence of the methyl transferase. Consistently,

persistence of H4K20me has been observed in Drosophila embryos

lacking PRSet7 (Trojer and Reinberg, 2006), suggesting that the

marker is stable retained. Moreover, H4k20me1 is required for Suv4-

20 dependent establishment of H4K20me2 that is recognized by the

ORC1 component of the origin of replication complex in vitro, and

might cooperate in marking replication origins (Kuo et al., 2012).

Stable isotope labeling of amino acids in cell culture (SILAC) in

combination with mass spectrometry (MS) in HeLa cells has shown

that H4K20me3 and H3K27me3 have the slowest rates of formation

(Zee et al., 2010). Studies of the turnover of histone modifications

indicates that H3K27me3 has a half life of 3 days (Zee et al., 2010).

The steady-state of H3K27 is achieved by the action of demethylases

and the methyltransferase PRC2. Once established, H3K27me3

recruits PRC2 to sites of DNA replication, facilitating the maintenance

of H3K27me3 via the action of EZH2 containing PRC2. Thus

H3K27me3 is replicated onto nascent deposited histones (Hansen et

al., 2008). Despite the reported association of PRC2 and PCNA during

replication, a recent report by Petruk et al., showed that at least in

Drosophila embryos, S phase nuclei are depleted of H3K27me3 as

well as H3K4me3 marks which appear to be replenished only later,

during G2 phase. They therefore suggest that these marks are unlikely

to be the only epigenetic mark that propagates the transcriptional state

of TrxG and PcG target genes, at least in Drosophila.

Page 35: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

15

Another hallmark of silenced chromatin is H3K9 methylation. Di

and trimethylation of H3 lysine 9 like H3K27me3 is propagated by a

positive feedback loop, in which Heterochromatin Protein 1 (HP1)

binds specifically to methylated H3K9, forms oligomers to bridge

neighboring nucleosomes and recruit Suv39h1/2, KMT1F and SETDB1

histone methyltransferases to methylate neighboring nucleosomes

thereby spreading the silent state (Bannister and Kouzarides, 2011;

Bannister et al., 2001; Lachner et al., 2001). Moreover, the association

of SETDB1 with the HP1-CAF1(chromatin association factor) complex

and the interaction between DNMT1 and the HMT G9a that associates

with PCNA during DNA synthesis, suggest that H3K9me3 propagation

is maintained during DNA replication (Estève et al., 2006; Loyola et al.,

2009). Hathaway et al., developed a chromatin in vivo assay (CiA)

system with the ability to induce heterochromatin formation through the

recruitment of HP1 to examine the epigenetic properties of H3K9me3

through cell division of embryonic stem cells. HP1 tethering leads to

suppression of gene activity, spreading of heterochromatin over a 10kb

domain after 5 days of HP1 recruitment independent of sequence

elements. H3K9 methylation is stable in the absence of HP1 with a

turnover rate similar to global histone turnover in HeLaS3 cells and

rates of histone H3 displacement form chromatin in Drosophila S2 cells

(Deal et al., 2010; Dodd et al., 2007; Hathaway et al., 2012; Zee et al.,

2010).

The transmission of histone modifications through a closed positive

feed-back loop, where reader and writers propagate the histone mark,

has been simulated in a mathematical model based on the silent

mating type of Schizosaccharomyces pombe. The region that contains

the mating type cassettes is normally in a silenced state but mutants

which have a portion of the silenced region depleted and replaced by a

reporter gene, generate a bistable condition, flipping between silenced

Page 36: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

16

and active state. This bistability is under the control of activating

acetylations and silencing methylations that are dynamically produced

by their respective transferases and removed by deacetylases and

demethylases, respectively (Dodd et al., 2007). The model showed

that 1) positive feedback loops in nucleosome modifications are an

effective and robust mechanism for epigenetic memory, 2) histone

modification can be highly dynamic without compromising stability, and

3) bistability, requires cooperativity that results from the ability of

modified nucleosomes to not only stimulated addition of same

modification but also to stimulate removal of competing modifications

(Dodd et al., 2007).

The role of histone modifications associated with transcriptional

activation to maintain an active chromatin state is less explored. The

turnover rates of histone modifications estimated by SILCA-MS show

that active modification marks turn over ~2.5 fold faster in comparison

with repressive marks, suggesting that cells require more fine temporal

control over activated genes than repressed genes (Zee et al., 2010).

Analysis of histone modifications at gene promoters and coding

regions in mammalian cell lines show that H3K4ac, H3K4me and

H3K79me persist upon inhibition of transcription and through mitosis

(Kouskouti and Talianidis, 2005).

1.1.2.4. X chromosome inactivation

In mammals, the imbalance in X-linked genes between XX and

XY individuals is regulated by silencing of a large percentage of genes

on female X chromosomes called X chromosome inactivation (XCi).

The initiation and maintenance of the inactive chromosome (Xi) is

controlled by the x-inactivation center that produces Xist, a ~17kb

ncRNA. Once the inactive X coated by Xist which is transcribed in cis

by unknown mechanisms, transcription marks across the chromosome

are lost and the Polycomb Repressive Complex 2 (PRC2) is recruited

Page 37: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

17

followed by broad distribution of H3K27me3, and enrichment of the

histone H2A variant macroH2A. As a result promoters of X-linked

genes eventually undergo DNA methylation at later stages to stably

maintain silencing (Calabrese et al., 2012; Heard and Disteche, 2006).

X chromosome inactivation is a classic example of in cis chromatin-

mediated epigenetic silencing that does not affect the homologous X

chromosome that shares the same nucleus.

1.1.2.5. Histone variants

With the exception of H4 all histones have variants that are not

linked specifically to DNA replication. These variant histones differ in

amino acid sequence from the canonical histones (Figure 1.3) and are

encoded by separate genes that are expressed throughout the cell

cycle. Different variants are enriched in specific chromatin domains

rather than the broad distribution across all chromatin as is the case

for canonical replication-coupled histones (Talbert and Henikoff, 2010).

An important property of histone variants is that by creating specialized

nucleosomes, they generate different functional chromatin domains

with distinct functions by influencing nucleosome stability or through

downstream effectors that bind to specific variants (Kurumizaka et al.,

2013). In humans, eight H3 variants have been identified: H3.1, H3.2,

H3.3 H3.4 (H3T), H3.5, H3.X, H3.Y and CENP-A. The major H3

variants are CENP-A and H3.3. Two of these H3 variants, CENP-A

and H3.3 that are relevant for the work described in this thesis will be

introduced in more detail below.

Page 38: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

18

Figure 1.3 Sequence alignments of human H3 variants. Alignment of

amino acid sequences corresponding to human H3 variants. Sequences are

compared in relation to the H3.3 variant and the amino acid differences are

highlighted. H3.1 and H3.2 differences are highlighted in purple, H3t in gray,

H3.X and H3.Y in yellow, and CENP-A in light blue. The position numbers of

amino acids that are different between H3.3 and H3.1/2 are indicated. The

positions of the N-terminal tail and of the α-helixes of the histone-fold motif are

shown (Szenker et al., 2011).

1.1.2.5.1 CENP-A

The histone H3 variant CENP-A uniquely marks centromeric

chromatin and is found in virtually all eukaryotes investigated thus far.

It shares only about 50% amino acid identity with canonical H3

(Kurumizaka et al., 2013). The centromere is a chromosomal region

that functions as a platform for the assembly of the kinetochore, which

drives segregation of chromosomes to daughter cell during cell division

(Allshire and Karpen, 2008; Cleveland et al., 2003). In most

eukaryotes, centromeres are propagated on repetitive satellite DNA

which in humans corresponds to α-satellite, a complex family of long

Page 39: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

19

tandemly repeated monomers of 171bp that constitute 5% of total

human DNA (Henikoff et al., 2001; Waye and Willard, 1986). However,

α-satellites are not sufficient for the function and maintenance of

centromeres, as is evident from neocentromeres, ectopic centromeres

that are stably transmitted through mitosis and meiosis (Tyler-Smith et

al., 1999; Vafa and Sullivan, 1997; Warburton et al., 1997).

While specific DNA sequences are non-essential, centromere

function strongly depends on CENP-A. Depletion of CENP-ACID in

Drosophila, leads to mislocalization of most kinetochore proteins while

the depletion of kinetochore proteins does not affect the localization of

CENP-ACID. Indeed, CENP-ACID mislocalization and formation of

ectopic kinetochores is observed when CENP-ACID is overexpressed

(Blower and Karpen, 2001; Heun et al., 2006). These findings strongly

suggest that CENP-A provides identity and position of the centromere

in dividing cells.

CENP-A is also structurally divergent of canonical H3

nucleosomes by inducing negative supercoil, left-handed wrapping and

by containing a specific domain at the HFD (Black et al., 2004; Sekulic

et al., 2010). The CENP-A targeting domain (CATD), that

encompasses the L1 and α2 helix of CENP-A, is pivotal for the

definition of centromeric chromatin. It has been shown that when the

CATD domain is placed within the HFD of canonical H3, the chimeric

histone generated is sufficient to target to centromeres (Black et al.,

2004; Panchenko et al., 2011). Moreover, it has been shown that the

CATD is the target sequence for Holliday junction recognizing protein

(HJURP), the chaperone for prenucleosomal CENP-A. HJURP

contributes to the efficient incorporation into centromeres (Foltz et al.,

2009) and transmit stability throughout the histone fold domains of

CENP-A and H4 (Panchenko et al., 2011).

Page 40: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

20

Throughout the entire cell cycle, CENP-A nucleosomes are

present at centromeres. Experiments using fluorescent pulse labeling

techniques based on SNAP-tagging and fluorescence recovery after

photobleaching (FRAP), whose details will be introduced in section 2,

have shown that in human cells, Xenopus extracts, Chicken cells and

Drosophila, CENP-A is targeted to centromeres during late

telophase/early G1 phase of the cell cycle (Bernad et al., 2011;

Dunleavy et al., 2012; Jansen et al., 2007; Schuh et al., 2007; Silva et

al., 2012). In human cells CENP-A has been found to be stably

retained throughout multiple cell divisions and segregates in a semi

conservative manner during DNA replication (Bodor et al., 2013;

Hemmerich et al., 2008; Jansen et al., 2007). Such high stability is

consistent with maintaining an epigenetic memory of centromere

identity.

Recently, direct evidence for the epigenetic nature of the

centromere has been shown by means of the induction of formation of

a new centromere. This was achieved by the artificial tethering of

CENP-ACID in Drosophila S2 cells and tethering of HJURP in human

culture cells to a non-centromeric site (Barnhart et al., 2011;

Mendiburo et al., 2011). Barnhart et al. forced the CENP-A chaperone

HJURP onto a LacO (Lac operon) domain at a non-centromeric locus.

This was found to be sufficient to nucleate CENP-A chromatin and

form a functional centromere. Mendiburo et al. used a similar approach

to show that the ectopic targeting of CENP-ACID is sufficient to recruit

nascent untagged CENP-A and create an epigenetic feed-back loop

that triggers heritable kinetochore formation for several subsequent

cell divisions.

As mentioned above, contrary to canonical histones, nascent

CENP-A is loaded in late telophase/early G1 phase in human cells

(Jansen et al., 2007). Prenucleosomal CENP-A is targeted to

Page 41: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

21

centromeres by HJURP (Foltz et al., 2009) through an interaction

either with the constitutive centromere complex (CCAN) or the Mis18

complex. Among the components of CCAN, CENP-N are CENP-C are

the factors that recognize the CENP-A nucleosome directly through the

CATD domain and the carboxyl terminus of CENP-A respectively, for

the recruitment of the CCAN to the CENP-A nucleosome (Carroll et al.,

2009, 2010). In fission yeast it has been reported that the recruitment

of HJURP requires the Mis18 complex (Pidoux et al., 2009; Williams et

al., 2009), but there is no evidence of a direct interaction between

Mis18 and HJURP. A possible link between the Mis18 complex and

HJURP are the RbAp46/48 proteins that interact with both the Mis18

complex and HJURP (Dunleavy et al., 2009). Alternatively, Mis18 may

change centromeric chromatin structure to facilitate the recruitment of

HJURP. In support of this view, the Mis18 complex has been

suggested to affect the acetylation state of chromatin (Fujita et al.,

2007; Ohzeki et al., 2012), and recruitment of HJURP to a human

artificial chromosome depends on the transcriptionally active chromatin

mark with H3K4me2 (Bergmann et al., 2011). Furthermore, the

requirement for Mis18 in the initiation of de novo centromere assembly

on α-satellite DNA can be bypassed by targeting histone acetyl

transferases to the satellite array (Ohzeki et al., 2012). Combined, this

evidence points to an elaborated protein network that possibly involves

changes in chromatin structure that constitutes the feed-back loop for

the self templating of CENP-A (Figure 1.4).

Page 42: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

22

Figure 1.4 Model of self-templating of CENP-A chromatin. CENP-N and

CENP-C bind directly to CENP-A nucleosomes that leads to the recruitment of

the Mis18 complex in early G1 phase. Changes of local centromeric

chromatin marks or structure leads to the recruitment of HJURP that deposits

new CENP-A. Ancestral CENP-A is labeled pink, new CENP-A in red and

local chromatin that may include H3 nucleosomes are labeled green. Adapted

from Gómez-Rodríguez and Jansen, 2013.

The assembly of nascent CENP-A is tightly coupled to the cell

cycle. HJURP binds to prenucleosomal CENP-A in late S/G2 phases

of the cell cycle and chaperones new CENP-A–H4 until its assembly in

early G1 phase (Dunleavy et al., 2009; Foltz et al., 2009; Jansen et al.,

2007); preventing misincorporation of CENP-A. The cell cycle is

controlled by Cdk2/cyclin A and Cdk1/cyclin B complexes during S, G2

and mitotic phases. By blocking CDK activity and visualizing newly

synthesized CENP-A in human culture cells, Silva et al., proposed that

CDKs hold the CENP-A assembly machinery in an inactive state until

mitotic exit for deposition of CENP-A (Silva et al., 2012). This model

was substantiated by the finding that CDK inhibition leads to premature

recruitment of Mis18α and Mis18BP1HsKNL2 to centromeres in G2 phase

Page 43: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

23

and concomitant CENP-A assembly. Depletion of Mis18α,

Mis18BP1HsKNL2 or HJURP in combination with CDK inhibition prevents

assembly of CENP-A in G1 and G2 phases. Thus, cell cycle restricted

loading of nascent CENP-A provides a direct coupling between the S-

phase turnover, cell division and subsequent assembly after mitosis,

potentially ensuring a proper homeostasis of centromere size across

cell division cycles (Gómez-Rodríguez and Jansen, 2013).

1.1.2.5.2. H3.3

In contrast to CENP-A, histone H3.3 differs from canonical H3

by just four amino acids which render the incorporation of H3.3

independent of DNA replication. Genome wide analysis of H3.3

distribution showed that H3.3 is deposited at chromatin regions that

are actively transcribed and in promoter regions coinciding with

enrichment of H3K4me1 and RNA polymerase II (Mito et al., 2005).

H3.3 assembly is not cell cycle regulated, but can occur

throughout the cell cycle, thereby replacing a fraction of the

nucleosome pool assembled during S phase (Gómez-Rodríguez and

Jansen, 2013; Wu et al., 1982). Recently, Deal et al., suggested that

H3.3 turnover is fast at active genes and regulatory elements (Deal et

al., 2010) but H3.3 in bulk is stable indicating that not the entire pool is

turned over. H3.3 histones are deposited by the Histone regulator A

(HIRA) complex in a replication-independent manner (Tagami et al.,

2004). HIRA forms a complex with Cabin1 and Ubinuclein (UBN1),

which copurifies with H3.3 (Tagami et al., 2004).

In addition to HIRA, studies in ES cells have described the α-

thalassemia X linked mental retardation protein - death associated

protein (ATRX-DAXX) complex, and DEK to be involved in the H3.3

deposition at telomeres, pericentromeres and centromeres (Drané et

al., 2010; Goldberg et al., 2010; Sawatsubashi et al., 2010). The

Page 44: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

24

function of H3.3 in the organization of centromeres and telomeres

chromatin is unknown (Goldberg et al., 2010). Whether it is associated

to transcription or needed for cell division and genome stability needs

to be established.

A direct role of H3.3 in maintaining memory of a chromatin

state is, as of yet, poorly defined. One study has implicated H3.3 in

memory. Nuclear transfer experiments in Xenopus laevis showed that

missexpression of the donor-originated MyoD gene occurs upon

initiation of zygotic transcription in nuclear transfer embryos. Memory

of MyoD target gene expression is maintained in the absence of

transcription during 24 mitotic divisions and this epigenetic memory is

dependent upon H3.3 lysine 4 methylation (Ng and Gurdon, 2008).

Page 45: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

25

2. Measuring protein dynamics

Key to our understanding of the contribution of chromatin to

epigenetic memory is a determination of the dynamics of chromatin

components. Below I will review different strategies that have been

used to determine different aspects of chromatin dynamics, stability

and turnover.

2.1 Fluorescence recovery after photobleaching (FRAP)

The green fluorescence protein (GFP) technology has made it

possible to apply methods such as Fluorescence recovery after

photobleaching (FRAP) and Fluorescence correlation spectroscopy

(FCS) to probe dynamic behavior of proteins in vivo. Fluorescence

recovery after photobleaching (FRAP) was developed by Axelrod and

colleagues as a quantitative technique to study dynamics of protein

mobility in living cells by measuring the rate of fluorescence recovery

at a previously bleached site (Ishikawa-Ankerhold et al., 2012). In a

FRAP experiment, fluorescent molecules in a small region of the cell

are irreversible photobleached using a high-powered laser beam.

Subsequently, movement of the surrounding non-bleached fluorescent

molecules into the photobleached area leads to recovery of

fluorescence with a particular velocity, which is recorded at low laser

power (Figure 2.1.) (Ishikawa-Ankerhold et al., 2012).

Page 46: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

26

Figure 2.1 Fluorescence recovery after photobleaching. Typical plot of

fluorescence intensity in a region of interest versus time after photobleaching

a fluorescent protein. The prebleach is compared with the asymptote of the

recovery curve to calculate the mobile and immobile fractions. The diffusion

rate of the fluorescent protein is determined from the recovery curve. Adapted

from Lippincott-Schwartz et al., 2001.

Recovery of fluorescence into the bleached area occurs as a

result of the diffusional exchange between bleached and unbleached

molecules. Quantitative FRAP studies define two kinetic parameters:

the mobile fraction (M), which is the fraction of fluorescent proteins that

can diffuse into the bleached region during the time course of the

experiment; and the immobile fraction, which is the fraction of

molecules that cannot exchange between bleached and nonbleached

regions (Lippincott-Schwartz et al., 2001). Knowledge of the rate of

molecular exchange can provide important insights into the properties

and interactions of molecules within the cellular environment. FRAP,

has recently been applied to 3D environments like the cytoplasm or

Page 47: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

27

inside the nucleus to determine the binding and diffusion parameters of

proteins in these environments. FRAP has been used to measure the

residence time of canonical histones, which exhibit a longer residence

time that most chromatin-associated proteins (see chapter 2 and

Kimura and Cook, 2001; Thiriet and Hayes, 2005).

2.2 Inducible fluorescent proteins

Photoactivatable fluorescent proteins (PAFPs) are capable of

displaying pronounced changes in their spectral properties in response

to irradiation with light of a specific wavelength and intensity (Lukyanov

et al., 2005). Rather than observing the recovery of fluorescence into

the bleached area (as in FRAP), the movement of photoactivated

molecules away from the activation spot is assessed (Sparkes et al.,

2011). The main advantage of PAFPs is the capability for the

performance of pulse-chase labeling, which contributes to elucidate

protein transport pathways, intricate connections between

compartments, protein turnover and super-resolution imaging

(Lippincott-Schwartz and Patterson, 2009).

More than 20 different PAFPs have been described, which can

be categorized in three classes: 1) irreversibly switches from a dark to

bright fluorescent state, 2) irreversible photoconversion from one

fluorescent color to another or 3) reversibly photoconversion, enabling

on/off switching capability. The first group includes PA-GFP a

photoactivatable variant of GFP and PA-mRFP1 and PAmCherry1 that

is derived from DsRed that are less phototoxic due to their low energy

red light emission. The irreversible photoconverters class has the

advantage that molecules are visible prior to photoconversion which

enables tracking and selection of specific regions to photoactivate.

Among these are EosFP and Dendra2 that convert from green to red

and PS-CFP2 a photoswitchable cyan FP that photoconverts from

cyan to green. The class of reversible highlighters with on/off switching

Page 48: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

28

is characterized by their ability to be repeatedly photoconverted

between two states using light of two distinct wavelengths. They

include FP595, Dronpa PA-Cherry and DsRed timer among others

(Lippincott-Schwartz and Patterson, 2009). Different applications in

diffraction limited and super-resolution imaging, can be achieved by

the combination of optical and biochemical characteristics of the

PAFPs. These include their brightness level, oligomeric state, contrast

ratio, rate of spontaneous conversion into an activated state and rate

of photobleaching (Lippincott-Schwartz and Patterson, 2009).

2.3 Self labeling tags

Self-labeling proteins are those that are capable to bind

irreversibly or with high affinity and specificity to a synthetic probe in

vitro or in vivo (Keppler et al., 2004). Self labeling protein tags are

used as a fusion to a protein of interest. Self labeling proteins include

SNAP, SNAPf, CLIP, CLIPf, Halo and TMP. The most well established

system is the SNAP-tag (Gautier et al., 2008; Keppler et al., 2004).

SNAP is a stable suicide-enzyme protein-fusion tag that catalyzes its

own covalent binding to cell permeable O6-benzylguanine (BG)

derivatives and thereby permits the labeling of SNAP-tag fusion

proteins with a variety of different synthetic probes. It was generated in

a stepwise manner from human O6-alkylguanine-DNA alkyltransferase

(hAGT) by introduction of 19 point mutations and deletion of 25 C-

terminal residues. Relative to hAGT, SNAP-tag possess a 52-fold

higher reactivity toward BG derivatives, does not bind to DNA and is

expressed as well in cells as on cell surfaces (Mollwitz et al., 2012).

The main advantages of self labeling tagging are: an alternative

for detection of proteins in organisms that are not suitable for the

expression of autofluorescent proteins, the ability to distinguish young

and old copies of a protein by labeling at different time points with

different fluorophores, simultaneous and specific labeling of self-

Page 49: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

29

labeling fusion proteins in vitro and in living cells (Bodor et al., 2012;

Gaietta et al., 2002; Gautier et al., 2008; Jansen et al., 2007).

Although, this system is not suited for the analysis of proteins with high

turnover, it has been used for in vivo analysis of chromatin dynamic

assembly of H3 histones. SNAP-tag quench-chase-pulse and pulse-

chase experiments (see chapter 2) have contributed to the elucidation

of the stability of CENP-A chromatin and the cell cycle timing of CENP-

A assembly that is restricted to G1 phase (Jansen et al., 2007). In

addition, it has facilitated the characterization of factors involved in

H3.1 and H3.3 deposition (Ray-Gallet et al., 2002).

2.4 Recombination-induced tag exchange (RITE)

Epitope tags are used to study protein expression and

localization using specific antibodies raised against peptides that are

used as tags. Detection of tagged proteins provides a steady state

snapshot of protein levels. Recently, genetically encoded epitope tags

have been used to develop a versatile strategy to study protein

dynamics called Recombination-induced tag exchange (RITE)

(Verzijlbergen et al., 2010). RITE is a genetic method that induces a

permanent epitope-tag switch in the coding sequence of a protein of

interest after a transient induction of a site-specific recombinase. RITE

is composed of two parts: a tandem-tag cassette that can be

integrated in frame of the gene of interest for conditional C-terminal

tagging, and a stably integrated and constitutively expressed hormone-

dependent Cre recombinanse for the control of epitope switching.

RITE cassettes encode for one epitope tag between two LoxP

recombination sites and a second, epitope tag downstream of the

second LoxP site. The tagged proteins are encoded by a single gene

and under the control of the endogenous promoter (Terweij et al.,

2013; Verzijlbergen et al., 2010). Cre induced recombination leads to

the switch in expression of the protein of interest with one tag to being

Page 50: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

30

expressed with another tag. In effect, this allows for the detection of

lifetime and fate of both the old pre-switch pool of protein and the new

post-switch pool of protein. Advantages of RITE are selective tagging

and following of one protein of interest, monitoring of old and newly

synthesized proteins and identification of proteins controlling protein

turnover (Verzijlbergen et al., 2010). The method is not suited for the

analysis of proteins with high turnover. The epitope tags used in RITE

also allows for application of proteomic methods, DNA based high-

throughput screens, protein dynamics in single cells by live imaging

(Radman-Livaja et al., 2011; Terweij et al., 2013; Verzijlbergen et al.,

2010). The use of RITE has allowed to determine directly the dynamics

of histones in yeast. This analysis has shown that nucleosomes are

dynamically retained for multiple cell divisions, that the rate of turnover

is dependent of transcription and that ancestral histones accumulate

towards the 5’ end of long and less transcribed genes (Radman-Livaja

et al., 2011; Verzijlbergen et al., 2010).

2.5 Metabolic labeling of proteins

2.5.1 Stable isotope labeling by amino acids in cell culture

(SILAC)

Mass spectrometry (MS) is an analytical strategy to identify

known and novel PTMs on proteins, as well as determine the relative

quantity of proteins (Sanz-Medel et al., 2008). Mass spectrometric

measurements are carried out in the gas phase on ionized analytes.

A mass spectrometer measures the mass to charge ratio (m/z)

of freely moving gas-phase ions in electric and/or magnetic fields

(Soldi et al., 2013). A mass spectrometry based proteomics

experiment consists of five stages: 1) protein isolation that might

include one dimensional gel electrophoresis, 2) enzymatic degradation

of proteins to peptides, 3) separation of peptides by high-pressure

Page 51: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

31

liquid chromatography and elution into an electrospray ion source,

where they are nebulized in small, highly charged droplets. After

evaporation, multiply protonated peptides enter the mass spectrometer

4) a mass spectrum of the peptides eluting at this time point is taken,

and 5) generation of a prioritized list of these peptides for

fragmentation (Aebersold and Mann, 2003).

Recently, a stable isotope labeling strategy called stable

isotope labeling by amino acids in cell culture (SILAC) has been

described (Ong et al., 2002). In SILAC, a growth medium is prepared

where natural (light) amino acids are replaced by heavy SILAC amino

acids. Cells grown in this medium incorporate the heavy amino acids.

When light and heavy cell populations are mixed, they remain

distinguishable by MS (Ong et al., 2002; Soldi et al., 2013). This allows

for quantitative measurements of changes in protein levels or post

translational modifications between experimental and control

conditions. It also allows heavy labeling to be used as a pulse label to

determine protein turnover by mass spectrometry. The advantages of

SILAC are: 1) there is no need of peptide labeling step which minimize

the loss of starting material, 2) no differences in labeling efficiency

between samples because incorporation is virtually 100%, 3) peptides

from the same protein can be compare due to the uniform labeling, 4)

labeling of peptides is specific to its sequence and the mass differential

between two states can be specified more directly and 5) quantification

of small proteins is possible (Ong et al., 2002). SILAC has been used

to profile protein levels, profile PTM dynamics during the cell cycle, in

spike in strategies and in pulse experiments to measure the turnover of

PTMs and histone variants (Soldi et al., 2013). SILAC has been

applied to profile the bulk dynamics of histone modifications by labeling

newly synthesized histones with a heavy isotope. In this way the

presence and quantity of new and old pool histones can be determined

Page 52: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

32

by MS (See chapter 2 and Ong et al., 2002). This strategy revealed

that H3.1 nucleosomes remain largely octameric throughout the cell

cycle whereas a proportion of H3.3 nucleosomes split and old H3.3

histones form hybrid nucleosomes with nascent H3.3 (Xu et al.,

2010b).

2.5.2. Covalent attachment of tags to capture histone and

identify turnover (CATCH-IT)

CATCH-IT is a modified pulse labeling method specifically

designed for estimating nucleosome turnover rates. It is based on

metabolically labeling newly synthesized histones with an amino acid

analog that can be subsequently coupled to an affinity tag (Deal and

Henikoff, 2010). In CATCH-IT native histones are pulse labeled with

the methionine (Met) surrogate azidohomoalanine (AHA). AHA is a

small bioorthogonal functional group used to tag proteins, glycans and

lipids in cells. It is stable at physiological temperatures and

incorporated by the biosynthetic machinery of the cell. After pulse

labeling AFA labeled proteins are subsequently ligated to biotin which

allows their detection (Dieterich et al., 2006). In the case of analysis of

chromatin dynamics, isolated nucleosomes containing newly

synthesized histones are affinity purified with streptavidin, and washed

stringently to remove non-histone proteins as well as H2A-H2B dimers.

Finally, the affinity purified DNA from chromatin that is pulse labeled or

allowed to turn over during a chase period is hybridized to high density

tiling microarrays to create a map of nucleosome occupancy that is

dependent on its rate of turnover following the initial pulse (Deal et al.,

2010). The advantage of CATCH-IT is that no transgenes or tags are

needed. However, the method does not allow for the detection of a

specific protein nor is it suitable to measure fast dynamics. The use of

CATCH-IT showed that newly synthesized H3/H4 is incorporated at

Page 53: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

33

active genes and enhancers in Drosophila and is turned over with a

half-life ~1-1.5 hours (Deal and Henikoff, 2010).

Modified from Bodor et al., 2012.

Method Description Advantages Disadvantages Scale Time

scales

FRAP Measures local protein turnover after photobleaching

Allows analysis at very short timescales

Not ideal for long timescales (hours.days); phototoxicity. Few cells can be analyzed

Singles cells

Seconds.minutes

Inducible

florescent

proteins

Measures local protein turnover after activation or conversion

Allows analysis at short to medium timescales

Not ideal for long timescales (hours.days); phototoxicity. Few cells can be analyzed

Singles cells

Seconds to hours

Self-labeling tags Enzymes that catalyze their covalent binding to small compounds

Allows measurements at long timescales; allows analysis of old vs new protein pools.

Does not allow measurement at short timescales, high fluorescent background in live cells.

Single cells/bulk

Hours-days

RITE Floxed protein tag that is replaced by a different tag upon Cre-expression

Allows analysis of old vs new protein pools

Depends on efficiency of Cre.recombination. Does not allow measurement at short timescales.

Single cells/bulk

Hours-days

Metabolic

labeling of

protein

(SILAC,CATCH-IT)

Pulse labeling using labeled amino acids or amino acid analogs

Not requirement for transgenes or tags

All proteins are labeled simultaneously, thus requiring downstream techniques to purify proteins of interest

Bulk Minutes-days

Table 3 Methods to Analyze Protein Turnover

Page 54: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

34

3. Aims of this thesis

Chromatin states can potentially be propagated through multiple

cell divisions thereby contributing to epigenetic inheritance. However,

the mechanistic basis for chromatin inheritance is not clear, nor is the

dynamics and the rate of turnover of histones at specific loci well

established. Histones and their post translational modifications are

candidates to mark epigenetic states. Gaining insight into the dynamic

state of chromatin is important for the understanding of the epigenetic

inheritance.

Early experiments in bulk chromatin suggest that histones are

stable and survive critical processes challenging stability such as DNA

replication and transcription, thereby supporting the argument that

histones have a role in epigenetic inheritance. However, whether

histones are stably maintained at all loci is not known. Indeed stability

has been questioned with the finding of residence times of nascent

histones on the order of 1.5h at active genes and promoters of

Drosophila cells. The determination of the genomic localization and

timescales of histone turnover is pivotal to elucidate the molecular

basis for the propagation of chromatin.

In chapter 2, I introduce TimeChIP, an assay that allows capturing

histone dynamics whose main advantage is that it allows for the

distinction of old and new pools of histones and high spatial resolution.

In chapter 3, we show for the first time that a population of ancestral

H3.1 and H3.3 histones is stably retained across the cell cycle in a

human cell line. Finally in chapter 4, I discuss these results and

present an outlook to the future based on the results and conclusions

of this work.

Page 55: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

35

References

Aebersold, R., and Mann, M. (2003). Mass spectrometry-based proteomics. Nature 422, 198–207.

Allshire, R.C., and Karpen, G.H. (2008). Epigenetic regulation of centromeric chromatin: old dogs, new tricks? Nat. Rev. Genet. 9, 923–937.

Bannister, A.J., and Kouzarides, T. (2011). Regulation of chromatin by histone modifications. Cell Res. 21, 381–395.

Bannister, A.J., Zegerman, P., Partridge, J.F., Miska, E.A., Thomas, J.O., Allshire, R.C., and Kouzarides, T. (2001). Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain. Nature 410, 120–124.

Barnhart, M.C., Kuich, P.H.J.L., Stellfox, M.E., Ward, J.A., Bassett, E.A., Black, B.E., and Foltz, D.R. (2011). HJURP is a CENP-A chromatin assembly factor sufficient to form a functional de novo kinetochore. J. Cell Biol. 194, 229–243.

Barth, T.K., and Imhof, A. (2010). Fast signals and slow marks: the dynamics of histone modifications. Trends Biochem. Sci. 35, 618–626.

Bergmann, J.H., Rodríguez, M.G., Martins, N.M.C., Kimura, H., Kelly, D.A., Masumoto, H., Larionov, V., Jansen, L.E.T., and Earnshaw, W.C. (2011). Epigenetic engineering shows H3K4me2 is required for HJURP targeting and CENP-A assembly on a synthetic human kinetochore. EMBO J. 30, 328–340.

Bernad, R., Sánchez, P., Rivera, T., Rodríguez-Corsino, M., Boyarchuk, E., Vassias, I., Ray-Gallet, D., Arnaoutov, A., Dasso, M., Almouzni, G., et al. (2011). Xenopus HJURP and condensin II are required for CENP-A assembly. J. Cell Biol. 192, 569–582.

Bird, A. (2002). DNA methylation patterns and epigenetic memory. Genes Dev. 16, 6–21.

Biswas, M., Voltz, K., Smith, J.C., and Langowski, J. (2011). Role of histone tails in structural stability of the nucleosome. PLoS Comput. Biol. 7, e1002279.

Black, B.E., Foltz, D.R., Chakravarthy, S., Luger, K., Woods, V.L., Jr, and Cleveland, D.W. (2004). Structural determinants for generating centromeric chromatin. Nature 430, 578–582.

Page 56: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

36

Blower, M.D., and Karpen, G.H. (2001). The role of Drosophila CID in kinetochore formation, cell-cycle progression and heterochromatin interactions. Nat. Cell Biol. 3, 730–739.

Bodor, D.L., Rodríguez, M.G., Moreno, N., and Jansen, L.E.T. (2012). Analysis of protein turnover by quantitative SNAP-based pulse-chase imaging. Curr. Protoc. Cell Biol. Editor. Board Juan S Bonifacino Al Chapter 8, Unit8.8.

Bodor, D.L., Valente, L.P., Mata, J.F., Black, B.E., and Jansen, L.E.T. (2013). Assembly in G1 phase and long-term stability are unique intrinsic features of CENP-A nucleosomes. Mol. Biol. Cell 24, 923–932.

Calabrese, J.M., Sun, W., Song, L., Mugford, J.W., Williams, L., Yee, D., Starmer, J., Mieczkowski, P., Crawford, G.E., and Magnuson, T. (2012). Site-specific silencing of regulatory elements as a mechanism of X inactivation. Cell 151, 951–963.

Carroll, C.W., Silva, M.C.C., Godek, K.M., Jansen, L.E.T., and Straight, A.F. (2009). Centromere assembly requires the direct recognition of CENP-A nucleosomes by CENP-N. Nat. Cell Biol. 11, 896–902.

Carroll, C.W., Milks, K.J., and Straight, A.F. (2010). Dual recognition of CENP-A nucleosomes is required for centromere assembly. J. Cell Biol. 189, 1143–1155.

Chanoine, C., Della Gaspera, B., and Charbonnier, F. (2004). Myogenic regulatory factors: Redundant or specific functions? Lessons from Xenopus. Dev. Dyn. 231, 662–670.

Chen, S., Bohrer, L.R., Rai, A.N., Pan, Y., Gan, L., Zhou, X., Bagchi, A., Simon, J.A., and Huang, H. (2010). Cyclin-dependent kinases regulate epigenetic gene silencing through phosphorylation of EZH2. Nat. Cell Biol. 12, 1108–1114.

Cleveland, D.W., Mao, Y., and Sullivan, K.F. (2003). Centromeres and Kinetochores: From Epigenetics to Mitotic Checkpoint Signaling. Cell 112, 407–421.

Deal, R.B., and Henikoff, S. (2010). Capturing the dynamic epigenome. Genome Biol. 11, 218.

Deal, R.B., Henikoff, J.G., and Henikoff, S. (2010). Genome-Wide Kinetics of Nucleosome Turnover Determined by Metabolic Labeling of Histones. Science 328, 1161–1164.

Page 57: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

37

Dieterich, D.C., Link, A.J., Graumann, J., Tirrell, D.A., and Schuman, E.M. (2006). Selective identification of newly synthesized proteins in mammalian cells using bioorthogonal noncanonical amino acid tagging (BONCAT). Proc. Natl. Acad. Sci. U. S. A. 103, 9482–9487.

Dodd, I.B., Micheelsen, M.A., Sneppen, K., and Thon, G. (2007). Theoretical Analysis of Epigenetic Cell Memory by Nucleosome Modification. Cell 129, 813–822.

Drané, P., Ouararhni, K., Depaux, A., Shuaib, M., and Hamiche, A. (2010). The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev. 24, 1253–1265.

Dunleavy, E.M., Roche, D., Tagami, H., Lacoste, N., Ray-Gallet, D., Nakamura, Y., Daigo, Y., Nakatani, Y., and Almouzni-Pettinotti, G. (2009). HJURP Is a Cell-Cycle-Dependent Maintenance and Deposition Factor of CENP-A at Centromeres. Cell 137, 485–497.

Dunleavy, E.M., Beier, N.L., Gorgescu, W., Tang, J., Costes, S.V., and Karpen, G.H. (2012). The cell cycle timing of centromeric chromatin assembly in Drosophila meiosis is distinct from mitosis yet requires CAL1 and CENP-C. PLoS Biol. 10, e1001460.

Easwaran, H.P., Schermelleh, L., Leonhardt, H., and Cardoso, M.C. (2004). Replication-independent chromatin loading of Dnmt1 during G2 and M phases. EMBO Rep. 5, 1181–1186.

Estève, P.-O., Chin, H.G., Smallwood, A., Feehery, G.R., Gangisetty, O., Karpf, A.R., Carey, M.F., and Pradhan, S. (2006). Direct interaction between DNMT1 and G9a coordinates DNA and histone methylation during replication. Genes Dev. 20, 3089–3103.

Ficz, G., Heintzmann, R., and Arndt-Jovin, D.J. (2005). Polycomb group protein complexes exchange rapidly in living Drosophila. Development 132, 3963–3976.

Foltz, D.R., Jansen, L.E.T., Bailey, A.O., Yates III, J.R., Bassett, E.A., Wood, S., Black, B.E., and Cleveland, D.W. (2009). Centromere-Specific Assembly of CENP-A Nucleosomes Is Mediated by HJURP. Cell 137, 472–484.

Fong, A.P., Yao, Z., Zhong, J.W., Cao, Y., Ruzzo, W.L., Gentleman, R.C., and Tapscott, S.J. (2012). Genetic and epigenetic determinants of neurogenesis and myogenesis. Dev. Cell 22, 721–735.

Page 58: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

38

Fonseca, J.P., Steffen, P.A., Müller, S., Lu, J., Sawicka, A., Seiser, C., and Ringrose, L. (2012). In vivo Polycomb kinetics and mitotic chromatin binding distinguish stem cells from differentiated cells. Genes Dev. 26, 857–871.

Francis, N.J., Follmer, N.E., Simon, M.D., Aghia, G., and Butler, J.D. (2009). Polycomb Proteins Remain Bound to Chromatin and DNA during DNA Replication In Vitro. Cell 137, 110–122.

Fujita, Y., Hayashi, T., Kiyomitsu, T., Toyoda, Y., Kokubu, A., Obuse, C., and Yanagida, M. (2007). Priming of Centromere for CENP-A Recruitment by Human hMis18α, hMis18β, and M18BP1. Dev. Cell 12, 17–30.

Gaietta, G., Deerinck, T.J., Adams, S.R., Bouwer, J., Tour, O., Laird, D.W., Sosinsky, G.E., Tsien, R.Y., and Ellisman, M.H. (2002). Multicolor and electron microscopic imaging of connexin trafficking. Science 296, 503–507.

Gautier, A., Juillerat, A., Heinis, C., Corrêa, I.R., Jr, Kindermann, M., Beaufils, F., and Johnsson, K. (2008). An engineered protein tag for multiprotein labeling in living cells. Chem. Biol. 15, 128–136.

Goldberg, A.D., Banaszynski, L.A., Noh, K.-M., Lewis, P.W., Elsaesser, S.J., Stadler, S., Dewell, S., Law, M., Guo, X., Li, X., et al. (2010). Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 140, 678–691.

Gómez-Rodríguez, M., and Jansen, L.E. (2013). Basic properties of epigenetic systems: lessons from the centromere. Curr. Opin. Genet. Dev. 23, 219–227.

Goutte-Gattat, D., Shuaib, M., Ouararhni, K., Gautier, T., Skoufias, D.A., Hamiche, A., and Dimitrov, S. (2013). Phosphorylation of the CENP-A amino-terminus in mitotic centromeric chromatin is required for kinetochore function. Proc. Natl. Acad. Sci. U. S. A. 110, 8579–8584.

De Graaf, C.A., and van Steensel, B. (2013). Chromatin organization: form to function. Curr. Opin. Genet. Dev. 23, 185–190.

Groth, A., Corpet, A., Cook, A.J.L., Roche, D., Bartek, J., Lukas, J., and Almouzni, G. (2007). Regulation of Replication Fork Progression Through Histone Supply and Demand. Science 318, 1928–1931.

Page 59: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

39

Haig, D. (2004). The (Dual) Origin of Epigenetics. Cold Spring Harb. Symp. Quant. Biol. 69, 67–70.

Hansen, K.H., Bracken, A.P., Pasini, D., Dietrich, N., Gehani, S.S., Monrad, A., Rappsilber, J., Lerdrup, M., and Helin, K. (2008). A model for transmission of the H3K27me3 epigenetic mark. Nat. Cell Biol. 10, 1291–1300.

Hathaway, N.A., Bell, O., Hodges, C., Miller, E.L., Neel, D.S., and Crabtree, G.R. (2012). Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460.

Heard, E., and Disteche, C.M. (2006). Dosage compensation in mammals: fine-tuning the expression of the X chromosome. Genes Dev. 20, 1848–1867.

Hemmerich, P., Weidtkamp-Peters, S., Hoischen, C., Schmiedeberg, L., Erliandri, I., and Diekmann, S. (2008). Dynamics of inner kinetochore assembly and maintenance in living cells. J. Cell Biol. 180, 1101–1114.

Henikoff, S., Ahmad, K., and Malik, H.S. (2001). The Centromere Paradox: Stable Inheritance with Rapidly Evolving DNA. Science 293, 1098–1102.

Heun, P., Erhardt, S., Blower, M.D., Weiss, S., Skora, A.D., and Karpen, G.H. (2006). Mislocalization of the Drosophila Centromere-Specific Histone CID Promotes Formation of Functional Ectopic Kinetochores. Dev. Cell 10, 303–315.

Hübner, M.R., Eckersley-Maslin, M.A., and Spector, D.L. (2013). Chromatin organization and transcriptional regulation. Curr. Opin. Genet. Dev. 23, 89–95.

Huen, M.S.Y., Sy, S.M.-H., Deursen, J.M. van, and Chen, J. (2008). Direct Interaction between SET8 and Proliferating Cell Nuclear Antigen Couples H4-K20 Methylation with DNA Replication. J. Biol. Chem. 283, 11073–11077.

Ishikawa-Ankerhold, H.C., Ankerhold, R., and Drummen, G.P.C. (2012). Advanced fluorescence microscopy techniques--FRAP, FLIP, FLAP, FRET and FLIM. Mol. Basel Switz. 17, 4047–4132.

Jansen, L.E.T., Black, B.E., Foltz, D.R., and Cleveland, D.W. (2007). Propagation of centromeric chromatin requires exit from mitosis. J. Cell Biol. 176, 795–805.

Page 60: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

40

Johnson, A., Meyer, B.J., and Ptashne, M. (1978). Mechanism of action of the cro protein of bacteriophage lambda. Proc. Natl. Acad. Sci. U. S. A. 75, 1783–1787.

Johnson, A.D., Poteete, A.R., Lauer, G., Sauer, R.T., Ackers, G.K., and Ptashne, M. (1981). λ Repressor and cro—components of an efficient molecular switch. Nature 294, 217–223.

Jørgensen, S., Schotta, G., and Sørensen, C.S. (2013). Histone H4 lysine 20 methylation: key player in epigenetic regulation of genomic integrity. Nucleic Acids Res. 41, 2797–2806.

Keppler, A., Kindermann, M., Gendreizig, S., Pick, H., Vogel, H., and Johnsson, K. (2004). Labeling of fusion proteins of O6-alkylguanine-DNA alkyltransferase with small molecules in vivo and in vitro. Methods San Diego Calif 32, 437–444.

Kimura, H., and Cook, P.R. (2001). Kinetics of Core Histones in Living Human Cells Little Exchange of H3 and H4 and Some Rapid Exchange of H2b. J. Cell Biol. 153, 1341–1354.

Kouskouti, A., and Talianidis, I. (2005). Histone modifications defining active genes persist after transcriptional and mitotic inactivation. EMBO J. 24, 347–357.

Kouzarides, T. (2007). Chromatin Modifications and Their Function. Cell 128, 693–705.

Kuo, A.J., Song, J., Cheung, P., Ishibe-Murakami, S., Yamazoe, S., Chen, J.K., Patel, D.J., and Gozani, O. (2012). The BAH domain of ORC1 links H4K20me2 to DNA replication licensing and Meier-Gorlin syndrome. Nature 484, 115–119.

Kurumizaka, H., Horikoshi, N., Tachiwana, H., and Kagawa, W. (2013). Current progress on structural studies of nucleosomes containing histone H3 variants. Curr. Opin. Struct. Biol. 23, 109–115.

Lachner, M., O’Carroll, D., Rea, S., Mechtler, K., and Jenuwein, T. (2001). Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410, 116–120.

Lassar, A.B., Buskin, J.N., Lockshon, D., Davis, R.L., Apone, S., Hauschka, S.D., and Weintraub, H. (1989). MyoD is a sequence-specific DNA binding protein requiring a region of myc homology to bind to the muscle creatine kinase enhancer. Cell 58, 823–831.

Page 61: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

41

Lavoie, G., and St-Pierre, Y. (2011). Phosphorylation of human DNMT1: Implication of cyclin-dependent kinases. Biochem. Biophys. Res. Commun. 409, 187–192.

Law, J.A., and Jacobsen, S.E. (2010). Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat. Rev. Genet. 11, 204–220.

Lee, T.-F., Zhai, J., and Meyers, B.C. (2010). Conservation and divergence in eukaryotic DNA methylation. Proc. Natl. Acad. Sci. U. S. A. 107, 9027–9028.

Lippincott-Schwartz, J., and Patterson, G.H. (2009). Photoactivatable fluorescent proteins for diffraction-limited and super-resolution imaging. Trends Cell Biol. 19, 555–565.

Lippincott-Schwartz, J., Snapp, E., and Kenworthy, A. (2001). Studying protein dynamics in living cells. Nat. Rev. Mol. Cell Biol. 2, 444–456.

Lo, S.M., Follmer, N.E., Lengsfeld, B.M., Madamba, E.V., Seong, S., Grau, D.J., and Francis, N.J. (2012). A Bridging Model for Persistence of a Polycomb Group Protein Complex through DNA Replication In Vitro. Mol. Cell 46, 784–796.

Loyola, A., Tagami, H., Bonaldi, T., Roche, D., Quivy, J.P., Imhof, A., Nakatani, Y., Dent, S.Y.R., and Almouzni, G. (2009). The HP1alpha-CAF1-SetDB1-containing complex provides H3K9me1 for Suv39-mediated K9me3 in pericentric heterochromatin. EMBO Rep. 10, 769–775.

Luger, K., Mäder, A.W., Richmond, R.K., Sargent, D.F., and Richmond, T.J. (1997). Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389, 251–260.

Lukyanov, K.A., Chudakov, D.M., Lukyanov, S., and Verkhusha, V.V. (2005). Innovation: Photoactivatable fluorescent proteins. Nat. Rev. Mol. Cell Biol. 6, 885–891.

Margueron, R., Justin, N., Ohno, K., Sharpe, M.L., Son, J., Drury, W.J., 3rd, Voigt, P., Martin, S.R., Taylor, W.R., De Marco, V., et al. (2009). Role of the polycomb protein EED in the propagation of repressive histone marks. Nature 461, 762–767.

Melnyk, C.W., Molnar, A., Bassett, A., and Baulcombe, D.C. (2011). Mobile 24 nt Small RNAs Direct Transcriptional Gene Silencing in the Root Meristems of Arabidopsis thaliana. Curr. Biol. 21, 1678–1683.

Page 62: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

42

Mendiburo, M.J., Padeken, J., Fülöp, S., Schepers, A., and Heun, P. (2011). Drosophila CENH3 Is Sufficient for Centromere Formation. Science 334, 686–690.

Mito, Y., Henikoff, J.G., and Henikoff, S. (2005). Genome-scale profiling of histone H3.3 replacement patterns. Nat. Genet. 37, 1090–1097.

Mollwitz, B., Brunk, E., Schmitt, S., Pojer, F., Bannwarth, M., Schiltz, M., Rothlisberger, U., and Johnsson, K. (2012). Directed evolution of the suicide protein O6-alkylguanine-DNA alkyltransferase for increased reactivity results in an alkylated protein with exceptional stability. Biochemistry (Mosc.) 51, 986–994.

Molnar, A., Melnyk, C.W., Bassett, A., Hardcastle, T.J., Dunn, R., and Baulcombe, D.C. (2010). Small Silencing RNAs in Plants Are Mobile and Direct Epigenetic Modification in Recipient Cells. Science 328, 872–875.

Ng, R.K., and Gurdon, J.B. (2008). Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription. Nat. Cell Biol. 10, 102–109.

Ohzeki, J., Bergmann, J.H., Kouprina, N., Noskov, V.N., Nakano, M., Kimura, H., Earnshaw, W.C., Larionov, V., and Masumoto, H. (2012). Breaking the HAC Barrier: Histone H3K9 acetyl/methyl balance regulates CENP-A assembly. EMBO J. 31, 2391–2402.

Ong, S.-E., Blagoev, B., Kratchmarova, I., Kristensen, D.B., Steen, H., Pandey, A., and Mann, M. (2002). Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics MCP 1, 376–386.

Osley, M.A. (1991). The regulation of histone synthesis in the cell cycle. Annu. Rev. Biochem. 60, 827–861.

Panchenko, T., Sorensen, T.C., Woodcock, C.L., Kan, Z.-Y., Wood, S., Resch, M.G., Luger, K., Englander, S.W., Hansen, J.C., and Black, B.E. (2011). Replacement of histone H3 with CENP-A directs global nucleosome array condensation and loosening of nucleosome superhelical termini. Proc. Natl. Acad. Sci. U. S. A. 108, 16588–16593.

Pengelly, A.R., Copur, Ö., Jäckle, H., Herzig, A., and Müller, J. (2013). A histone mutant reproduces the phenotype caused by loss of histone-modifying factor Polycomb. Science 339, 698–699.

Page 63: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

43

Petruk, S., Sedkov, Y., Johnston, D.M., Hodgson, J.W., Black, K.L., Kovermann, S.K., Beck, S., Canaani, E., Brock, H.W., and Mazo, A. (2012). TrxG and PcG Proteins but Not Methylated Histones Remain Associated with DNA through Replication. Cell 150, 922–933.

Pidoux, A.L., Choi, E.S., Abbott, J.K.R., Liu, X., Kagansky, A., Castillo, A.G., Hamilton, G.L., Richardson, W., Rappsilber, J., He, X., et al. (2009). Fission Yeast Scm3: A CENP-A Receptor Required for Integrity of Subkinetochore Chromatin. Mol. Cell 33, 299–311.

Ptashne, M., Backman, K., Humayun, M.Z., Jeffrey, A., Maurer, R., Meyer, B., and Sauer, R.T. (1976). Autoregulation and function of a repressor in bacteriophage lambda. Science 194, 156–161.

Radman-Livaja, M., Verzijlbergen, K.F., Weiner, A., van Welsem, T., Friedman, N., Rando, O.J., and van Leeuwen, F. (2011). Patterns and Mechanisms of Ancestral Histone Protein Inheritance in Budding Yeast. PLoS Biol 9, e1001075.

Ray-Gallet, D., Quivy, J.-P., Scamps, C., Martini, E.M.-D., Lipinski, M., and Almouzni, G. (2002). HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol. Cell 9, 1091–1100.

Remus, D., and Diffley, J.F.X. (2009). Eukaryotic DNA replication control: lock and load, then fire. Curr. Opin. Cell Biol. 21, 771–777.

Sanz-Medel, A., Montes-Bayón, M., del Rosario Fernández de la Campa, M., Encinar, J.R., and Bettmer, J. (2008). Elemental mass spectrometry for quantitative proteomics. Anal. Bioanal. Chem. 390, 3–16.

Sawatsubashi, S., Murata, T., Lim, J., Fujiki, R., Ito, S., Suzuki, E., Tanabe, M., Zhao, Y., Kimura, S., Fujiyama, S., et al. (2010). A histone chaperone, DEK, transcriptionally coactivates a nuclear receptor. Genes Dev. 24, 159–170.

Schermelleh, L., Haemmer, A., Spada, F., Rösing, N., Meilinger, D., Rothbauer, U., Cardoso, M.C., and Leonhardt, H. (2007). Dynamics of Dnmt1 interaction with the replication machinery and its role in postreplicative maintenance of DNA methylation. Nucleic Acids Res. 35, 4301–4312.

Schuh, M., Lehner, C.F., and Heidmann, S. (2007). Incorporation of Drosophila CID/CENP-A and CENP-C into Centromeres during Early Embryonic Anaphase. Curr. Biol. 17, 237–243.

Page 64: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

44

Sekulic, N., Bassett, E.A., Rogers, D.J., and Black, B.E. (2010). The structure of (CENP-A-H4)(2) reveals physical features that mark centromeres. Nature 467, 347–351.

Shibahara, K., and Stillman, B. (1999). Replication-dependent marking of DNA by PCNA facilitates CAF-1-coupled inheritance of chromatin. Cell 96, 575–585.

Silva, M.C.C., Bodor, D.L., Stellfox, M.E., Martins, N.M.C., Hochegger, H., Foltz, D.R., and Jansen, L.E.T. (2012). Cdk Activity Couples Epigenetic Centromere Inheritance to Cell Cycle Progression. Dev. Cell 22, 52–63.

Simon, J.A., and Kingston, R.E. (2009). Mechanisms of polycomb gene silencing: knowns and unknowns. Nat. Rev. Mol. Cell Biol. 10, 697–708.

Simon, J.A., and Kingston, R.E. (2013). Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol. Cell 49, 808–824.

Soldi, M., Cuomo, A., Bremang, M., and Bonaldi, T. (2013). Mass spectrometry-based proteomics for the analysis of chromatin structure and dynamics. Int. J. Mol. Sci. 14, 5402–5431.

Spada, F., Haemmer, A., Kuch, D., Rothbauer, U., Schermelleh, L., Kremmer, E., Carell, T., Längst, G., and Leonhardt, H. (2007). DNMT1 but not its interaction with the replication machinery is required for maintenance of DNA methylation in human cells. J. Cell Biol. 176, 565–571.

Sparkes, I.A., Graumann, K., Martinière, A., Schoberer, J., Wang, P., and Osterrieder, A. (2011). Bleach it, switch it, bounce it, pull it: using lasers to reveal plant cell dynamics. J. Exp. Bot. 62, 1–7.

Szenker, E., Ray-Gallet, D., and Almouzni, G. (2011). The double face of the histone variant H3.3. Cell Res. 21, 421–434.

Tagami, H., Ray-Gallet, D., Almouzni, G., and Nakatani, Y. (2004). Histone H3.1 and H3.3 Complexes Mediate Nucleosome Assembly Pathways Dependent or Independent of DNA Synthesis. Cell 116, 51–61.

Talbert, P.B., and Henikoff, S. (2010). Histone variants — ancient wrap artists of the epigenome. Nat. Rev. Mol. Cell Biol. 11, 264–275.

Page 65: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

45

Tanaka, S., and Araki, H. (2010). Regulation of the initiation step of DNA replication by cyclin-dependent kinases. Chromosoma 119, 565–574.

Terweij, M., van Welsem, T., van Deventer, S., Verzijlbergen, K.F., Menendez-Benito, V., Ontoso, D., San-Segundo, P., Neefjes, J., and van Leeuwen, F. (2013). Recombination-induced tag exchange (RITE) cassette series to monitor protein dynamics in Saccharomyces cerevisiae. G3 Bethesda Md 3, 1261–1272.

Thayer, M.J., Tapscott, S.J., Davis, R.L., Wright, W.E., Lassar, A.B., and Weintraub, H. (1989). Positive autoregulation of the myogenic determination gene MyoD1. Cell 58, 241–248.

Thiriet, C., and Hayes, J.J. (2005). Replication-independent core histone dynamics at transcriptionally active loci in vivo. Genes Dev. 19, 677–682.

Trojer, P., and Reinberg, D. (2006). Histone Lysine Demethylases and Their Impact on Epigenetics. Cell 125, 213–217.

Tyler-Smith, C., Gimelli, G., Giglio, S., Floridia, G., Pandya, A., Terzoli, G., Warburton, P.E., Earnshaw, W.C., and Zuffardi, O. (1999). Transmission of a fully functional human neocentromere through three generations. Am. J. Hum. Genet. 64, 1440–1444.

Vafa, O., and Sullivan, K.F. (1997). Chromatin containing CENP-A and α-satellite DNA is a major component of the inner kinetochore plate. Curr. Biol. 7, 897–900.

Verzijlbergen, K.F., Menendez-Benito, V., van Welsem, T., van Deventer, S.J., Lindstrom, D.L., Ovaa, H., Neefjes, J., Gottschling, D.E., and van Leeuwen, F. (2010). Recombination-induced tag exchange to track old and new proteins. Proc. Natl. Acad. Sci. U. S. A. 107, 64–68.

Wang, H., Wang, L., Erdjument-Bromage, H., Vidal, M., Tempst, P., Jones, R.S., and Zhang, Y. (2004). Role of histone H2A ubiquitination in Polycomb silencing. Nature 431, 873–878.

Warburton, P.E., Cooke, C.A., Bourassa, S., Vafa, O., Sullivan, B.A., Stetten, G., Gimelli, G., Warburton, D., Tyler-Smith, C., Sullivan, K.F., et al. (1997). Immunolocalization of CENP-A suggests a distinct nucleosome structure at the inner kinetochore plate of active centromeres. Curr. Biol. CB 7, 901–904.

Page 66: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 1 – General Introduction

46

Waye, J.S., and Willard, H.F. (1986). Structure, organization, and sequence of alpha satellite DNA from human chromosome 17: evidence for evolution by unequal crossing-over and an ancestral pentamer repeat shared with the human X chromosome. Mol. Cell. Biol. 6, 3156–3165.

Williams, J.S., Hayashi, T., Yanagida, M., and Russell, P. (2009). Fission yeast Scm3 mediates stable assembly of Cnp1/CENP-A into centromeric chromatin. Mol. Cell 33, 287–298.

Wu, R.S., Tsai, S., and Bonner, W.M. (1982). Patterns of histone variant synthesis can distinguish G0 from G1 cells. Cell 31, 367–374.

Xu, C., Bian, C., Yang, W., Galka, M., Ouyang, H., Chen, C., Qiu, W., Liu, H., Jones, A.E., MacKenzie, F., et al. (2010a). Binding of different histone marks differentially regulates the activity and specificity of polycomb repressive complex 2 (PRC2). Proc. Natl. Acad. Sci. U. S. A. 107, 19266–19271.

Xu, M., Long, C., Chen, X., Huang, C., Chen, S., and Zhu, B. (2010b). Partitioning of Histone H3-H4 Tetramers During DNA Replication–Dependent Chromatin Assembly. Science 328, 94–98.

Zee, B.M., Levin, R.S., Xu, B., LeRoy, G., Wingreen, N.S., and Garcia, B.A. (2010). In vivo residue-specific histone methylation dynamics. J. Biol. Chem. 285, 3341–3350.

Zentner, G.E., and Henikoff, S. (2013). Regulation of nucleosome dynamics by histone modifications. Nat. Struct. Mol. Biol. 20, 259–266.

Zhou, W., Zhu, P., Wang, J., Pascual, G., Ohgi, K.A., Lozach, J., Glass, C.K., and Rosenfeld, M.G. (2008). Histone H2A monoubiquitination represses transcription by inhibiting RNA polymerase II transcriptional elongation. Mol. Cell 29, 69–80.

Page 67: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

47

Page 68: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

48

Page 69: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

49

Chapter 2 – TimeChIP

Page 70: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

50

Page 71: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

51

AUTHOR CONTRIBUTION

All the experiments were planned by the author and the

supervisor Lars E.T. Jansen. All the experiments in this chapter were

executed by the author.

SUMMARY

Proteins are dynamic entities. The steady-state of proteins is

defined by the combination of synthesis, localization, maintenance and

degradation. Protein stability is likely an important parameter in

protein-based epigenetic memory. Histones have been proposed to act

as markers of epigenetic memory. Bulk analysis of histone dynamics

suggests that histones turnover slow. Although, recently a genome

wide analysis of newly synthesized histones indicates that histones

turnover rapidly, at least at active genes, challenging the role of

histones as epigenetic markers. To measure the dynamics of histones,

we developed a strategy based on the SNAP system for the

performance of pulse-chase experiments, we call TimeChIP. We show

that TimeChIP facilitates the study of histone dynamics because of its

temporal resolution, analysis in intact cells, differentiation of old and

new pools of histone and the ability to determine turnover at high

spatial resolution across the genome.

Page 72: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

52

INTRODUCTION

Proteins undergo continuous changes in their interactions,

stability and location with consequences for their functions. One

striking example of such a consequence is the loss of post-

translational modifications and need for re-establishment once the

protein is replenished. The spatio-temporal dynamics of proteins

involves coordination of synthesis and assembly of nascent proteins.

Of special relevance to this thesis is protein inheritance across cell

divisions which is directly impacted by protein turnover rates. For the

analysis of different features of protein dynamics there are an array of

methodologies which differ in the degree of both spatial and temporal

resolution. Bulk measures of protein turnover are based on differential

chemical labeling. Proteins are in vivo labeled e.g. with isotope labeled

amino acids and followed for a certain period of time. Then, the

distribution of new and old proteins is determined in bulk. Traditionally

this was done by measuring density differences after isolation of

proteins of interest (Jackson and Chalkley, 1974; Jackson et al., 1975).

A modern incarnation of this is the use of non-radioactive isotopes for

stable isotope labeling of amino acids in cell culture that can be

detected by mass spectrometry. This method is rendered quantitative

when internally controlled with stable isotope labeling by amino acid in

cell culture (SILAC) (Ong et al., 2002). Although such assays are

highly quantitative they do not provide information about the

localization of the protein and the temporal resolution is low.

In contrast to such bulk measures, methodologies have been

developed that assay turnover at specific loci. These include

microscopy-based methods such as photoactivation or photobleaching

of fluorescent proteins fused to the protein of interest. In fluorescence

recovery after photobleaching (FRAP) an area of a cell expressing a

fluorescently tagged protein is rapidly and irreversible bleached and

Page 73: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

53

the influx of unbleached molecules (recovery of fluorescence) is

measured as a function of time (Ishikawa-Ankerhold et al., 2012).

FRAP has been used for the analysis of histone dynamics, where after

photobleaching a small area of nuclei, H3-GFP and H4-GFP enter the

bleached area more slowly than H2B-GFP. Once incorporated into

replication foci during S phase, H3 and H4 are stably maintained and

can even persist through mitosis (Kimura and Cook, 2001).

Photoactivation experiments function reciprocally to FRAP where a

population of dark inactive molecules is locally activated. In

fluorescence photoactivation localization microscopy (FPALM)

activated molecules are switched on by photoactivation and yield a

signal over a dark background. Its tracking over time provides spatial

and temporal dynamics of the protein of interest (Hess et al., 2006).

Albeit, FRAP provides a high temporal resolution and FPALM

subcellular specificity, both are phototoxic, and restricted to short

timescales. Furthermore, although specific nuclear domains can be

analyzed, dynamics cannot be analyzed at genome sequence

resolution.

To circumvent these limitations, novel methods have recently

been developed specifically to determine dynamics of chromatin that

include recombination-induced tag exchange (RITE). This method

allows to measure ancestral and newly synthesized proteins pools

encoded by the same transgene in budding yeast (Verzijlbergen et al.,

2010). In RITE the protein of interest is encoded by a transgene

containing an epitope tag and a stop codon flanked by loxP

recombination sites followed by a second epitope tag. When Cre

recombinase activity is induced the sequence encoding the first tag is

removed and expression of second tag commences. This, in effect

creates a pulse labeling condition where the differentially tagged

proteins represent old and new protein. Accordingly, the two tagged

Page 74: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

54

proteins can be affinity purified and relative protein occupancy can be

mapped across the genome by microarrays or sequencing. Besides

the simultaneous detection of old and new protein, this method allows

for the measurement of dynamics at long time scales at DNA

sequence resolution. The ability to measure dynamics at short

timescales is limited by the efficiency of Cre-recombination. In addition,

histone dynamics is measured in relative terms derived from the ration

between old on new protein. RITE experiments have shown the

dynamics of replication-independent histone H3 in budding yeast,

whose rate of exchange vary between cell cycle phases. Particularly in

G1 phase half of old histones are replaced and continued histone

deposition was maintained during at least three cell divisions

(Verzijlbergen et al., 2010). Importantly, when considering retention,

ancestral histones accumulate toward 5’ end of long and less

transcribed genes as opposed to turning over uniformly across the

genome. Consistently, a mathematical model which considers histone

turnover, histone spreading dependent of replication and transcription-

dependent passback, predicted that maternal histones are

reincorporated within two nucleosomes of their original site (Radman-

Livaja et al., 2011). A technique with a similar application as that of

RITE is a method called covalent attachment of tags to capture

histones and identify turnover (CATCH-IT). In CATCH-IT all nascent

proteins are metabolically labeled with an amino-acid analog that can

subsequently be coupled to a biotin affinity tag (Deal et al., 2010).

Following biotinylation, chromatin is extracted and fragmented.

Sequences associated with biotinylated chromatin are analyzed. This

method provides high spatial information down to nucleotide sequence

resolution but is limited to the measurement of nascent protein

incorporation and turnover at a short timescale. CATCH-IT has been

used to measure the incorporation of nascent histones across the

Drosophila genome. Somewhat surprisingly, using CATCH-IT Deal et

Page 75: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

55

al., found that new synthesized H3 has a half-life ~1-1.5 hours at active

genes and enhancers (Deal et al., 2010). Similar to what has been

observed in yeast by RITE (Verzijlbergen et al., 2010), this study

demonstrated that the rate of histone turnover correlates with the rate

of gene expression.

A powerful approach for protein dynamics studies is the use of

self-labeling tags that can overcome some of the limitations of the

methodologies described above. The SNAP-tag is derived from the

human DNA repair protein O6-alkylguanine-DNA alkyltransferase

(hAGT) that catalyzes its own covalent binding to cell permeable O6-

benzylguanine (BG) derivatives (Keppler et al., 2004) (Figure 2.1). The

self-labeling nature of the SNAP-tag makes it ideally suited for pulse

labeling studies. The SNAP-tag can be used to label proteins in living

cells as well as in vitro. Importantly, SNAP reactive substrates can be

conjugated with different molecules that include fluorescent dyes as

well as e.g. biotin or Digoxin. This renders this tag applicable to both

fluorescence and well as biochemical techniques where e.g.

biotinylation allows for the immobilization of a protein of interest. Thus

in SNAP pulse-chase experiments, the SNAP tagged protein is pulse

labeled with BG bearing a dye molecule, then chase for a period where

the pulse labeled protein is turned over and replenished by new

unlabeled protein. Alternatively, in SNAP quench-chase-pulse

experiments, the SNAP-tagged protein is labeled with unconjugated

BG (quench), chased for a certain time where new protein is

synthesized and subsequently labeled with a dye-conjugated BG

molecule (Figure 2.2). Experiments using the SNAP tag have provided

insight in the dynamics of histone H3 variants.

Initially this method was used to characterize the dynamics of

the histone H3 variant CENP-A whose localization is restricted to the

centromere. This strict localization makes CENP-A an ideal substrate

Page 76: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

56

for fluorescence-based approaches. Pulse-chase experiments

demonstrated that CENP-A is a stable protein at centromeres, diluted

only by DNA replication and newly synthesized CENP-A is targeted to

centromeres in a strict cell cycle regulated manner in early G1 phase

(Bodor et al., 2013; Jansen et al., 2007). In contrast to CENP-A, H3.3,

another H3 variant, was initially proposed to be deposited throughout

the cell cycle. Importantly, assembly does not dependent on DNA

synthesis in S phase while canonical H3 is loaded (Ahmad and

Henikoff, 2002). In contrast to CENP-A, the histone H3 variants H3.1

and H3.3 display very different dynamics. While assembly of H3.1 is

restricted to S phase by direct coupling to the DNA replication

machinery, (Shibahara and Stillman, 1999; Smith and Stillman, 1989),

H3.3 is dynamically assembly at active genes, promoters and

subtelomeric regions and is also detected at centromeres (Deal et al.,

2010; Goldberg et al., 2010; Ray-Gallet et al., 2011). A recent study

used fluorescence SNAP-technology to characterize H3.1 and H3.3

dynamics across the cell cycle based on the quench-chase-pulse

strategy described above. This confirmed the strict S-phase

dependency of H3.1 assembly and the dynamic assembly of H3.3

(Ray-Gallet et al., 2011). However, such microscopy-based analysis

does not have the resolution to study the stability of histone H3.1 or

H3.3 at specific loci.

Page 77: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

57

Figure 2.1 SNAP-tag. SNAP-tag (brown) is fused to the protein of interest

(green). Self-labeling with benzylguanine results in a covalent irreversible

bond between the benzyl moiety and a reactive cysteine in SNAP with the

consequent releases of guanine.

To gain insight into locus-specific assembly and turnover of

specific histone variants, we sought to develop a strategy based on

pulse labeling of SNAP-tagged histones, followed by chromatin

isolation and fragmentation, immobilization of nucleosomes dependent

on the SNAP pulse label and DNA isolation to determine occupancy at

specific loci. We call this method TimeChIP as it resembles a

Chromatin immunoprecipitation (ChIP) protocol but provides temporal

information of histone occupancy. We aimed to generate a system to

determine the site specific dynamics of histone H3 variants across the

cell cycle in human somatic cells.

Page 78: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

58

Figure 2.2 SNAP labeling strategies. A) Pulse-chase: cells that produce and

turnover SNAP-tagged protein (light green spheres, represent protein

population) are labeled with SNAP substrate at time 0 (dark green spheres).

Then, the protein is chased by allowing cells to continue to synthesize SNAP

protein that is not labeled, while the pulse labeled pool turns over. B) Quench-

chase-pulse: cells that produce and turnover SNAP-tagged protein are

quenched with an unconjugated SNAP substrate (no dye, grey spheres) at

time 0, blocking subsequent labeling. Then, cells continue to synthesize

SNAP protein that is not labeled for a certain period and new SNAP protein is

specifically labeled with SNAP substrate bearing a dye.

Page 79: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

59

MATERIALS AND METHODS

Cell lines and constructs

HeLa cell lines stably expressing H3.1-SNAP-3XHA, H3.3-

SNAP-3XHA or CENP-A-SNAP-3XHA (Jansen et al., 2007; Ray-Gallet

et al., 2011) were cultured in DMEM medium supplemented with 10%

newborn calf serum, 2mM L/Glutamine, 100/mL Penicillin and

100µg/mL Streptomycin (all from Gibco), at 37°C and 5% CO2. The

constructs 3xHA-SNAPf-2xPreScission-CENP-A, 3xHA-SNAP-

2xPreScission-CENP-A, 3xHA-CLIPf-2xPreScission-CENP-A and

3xHA-CLIP-2xPreScission-CENP-A named pLJ471, pLJ509, pLJ473

and pLJ492 respectively, were constructed by a PCR-generated

fragment carrying a triple HA-tag, SNAPf, SNAP, CLIPf or CLIP-tag

and a double PreScission protease site cloned into the NdeI and

BamHI sites of pSS26m. The resulting ORF was subcloned into the

EcoRV and BamHI sites of pCEMS1-CLIP-CENP-A (pLJ310),

replacing the CLIP fragment.

The SNAP-3XHA fragment from pSS26m-SNAP-3XHA

(pLJ222) was subcloned into EcoRV and NotI sites of pCEMS1-CLIP

(pLJ299), replacing CLIP, resulting in pLJ447. T98G SNAP-3XHA

cells were cultured in DMEM medium supplemented with 10%

newborn calf serum, 2mM L/Glutamine, 100/mL Penicillin and

100µg/mL Streptomycin (all from Gibco), at 37°C and 5% CO2 and

selected with 0,5mg/mL G418.

DNA transfections

T98G cells were transfected with 200ng of DNA, 1µl Plus

Reagent and 1.25µl of Lipofectamine (Invitrogen) in Optimem reduced

serum media (Gibco) according to manufactures instructions.

Page 80: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

60

Cell synchronization

HeLa CENP-A-SNAP-3XHA cells were synchronized by double

Thymidine block. Cells were treated with 2mM of Thymidine (Sigma)

for 17 hours, washed twice in medium and released in medium

containing 24µM of Deoxycytidine (Sigma) for 9 hours. Subsequently,

cells were treated again with Thymidine for 16 hours and released into

medium containing Deoxycytidine.

SNAP quench-chase-pulse labeling

Synchronized HeLa cells expressing CENP-A-SNAP were

pulse labeled by addition of 5µM BTP (SNAP-Cell Block New England

Biolabs) in presence of Thymidine in growth medium for 30 minutes at

37°C for irreversible non-fluorescent labeling of the preexisting CENP-

A-SNAP pool. 3 hours after the quench, cells were treated with 9µM

RO-3306 (Calbiochem) for 8 hours and followed for 3 hours treatment

with 24µM MG132 (Sigma).

Pulse labeling of intact cells was performed with 10µM BG-

Biotin or 2µM TMR Star (SNAP-Cell TMR-Star New England Biolabs),

in growth medium for 1 hour or 15 minutes, respectively at 37ºC. After

each labeling step (fluorescent and non-fluorescent) cells were

washed twice with medium and reincubated at 37ºC to allow excess

SNAP substrate to be release from cells. After 30 minutes, cells were

washed again once in medium.

Pulse labeling of permeabilized cells: 2-3 x 107 cells were

collected, resuspended in chromosome isolation buffer (CIB buffer)

and pulse labeled with 10µM BG-Biotin (New England Biolabs) for 1

hour at room temperature.

Page 81: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

61

Soluble Nucleosome Preparation

Chromatin was prepared by a modification of the method of

Yoda and Ando, 2004. The following volumes and numbers are given

for one purification (~5 x 107 cell equivalent): cells were equilibrated in

15mL CIB buffer (3.75mM Tris pH7.5, 20mM KCl, 0.5mM EDTA,

0.5mM DTT, 0.05mM Spermidine (Sigma), 0.125mM Spermine

(Sigma), 0.1% recrystallized Digitonin (Sigma), 1mM PMSF, 1:1000

Protease inhibitor cocktail (Sigma), aprotinin (Sigma)), dounced 10x

with tight pestle and centrifuged at 300g for 5 min at 4ºC. Dounce

homogenization and centrifugation was repeated once more. Pelleted

nuclei were washed in 15mL washing buffer (20mM Hepes sodium

pH7.7, 20mM KCl, 0.5mM EDTA, 0.5mM DTT, 0.5mM PMSF, 1:1000

Protease inhibitor cocktail (Sigma) and aprotinin (Sigma)), and

centrifuged at 300g for 5 min at 4ºC. Pelleted nuclei were washed

again in 7.5mL washing buffer with 0.3M NaCl and centrifuged at 500g

for 10 min at 4ºC. Chromatin resuspended in 500µL washing buffer

was digested with 800U/mL Micrococcal nuclease (Roche) in the

presence of 0.3M NaCl and 3mM CaCl2 while rotating for 1 hour at

room temperature. The reaction was quenched with 5mM EGTA,

0.05% NP40 and centrifuged at 10000g for 15 min at 4ºC. Soluble

nucleosomes were recovered.

SNAP-Nucleosomes Purification

The soluble nucleosomes concentration was adjusted to 1 A276

units in 340µL washing buffer A with 0.3M NaCl. 0.2mg Streptavidin

Magnetic Beads (Thermo Scientific) were washed twice with washing

buffer B (20mM Hepes sodium pH7.7, 20mM KCl, 0.5mM EDTA,

0.5mM DTT, 0.5mM PMSF, 1:1000 Protease inhibitor cocktail (Sigma),

aprotinin (Sigma), 0.5%NP40 and 0.5M NaCl), while rotating for 5 min

at room temperature. Beads were blocked with washing buffer B

supplemented with 50mg BSA and 200µg/mL yeast tRNA (Invitrogen)

Page 82: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

62

while rotating for 1 hour at 4ºC. SNAP biotinylated nucleosomes were

recovered by adding 300µL of the adjusted fraction to the pretreated

beads, and rotated for 1 hour at 4ºC, while the 40µL of nucleosomes

were saved as input fraction. After 3 washes with washing buffer B

with rotation for 5 min at room temperature each, 250µL TNES buffer

with 100µg/mL RNaseA (Invitrogen) was added directly to the beads

and incubate for 10 minutes at room temperature. The resulting RNase

treated samples were subjected to a phenol-chloroform extraction

followed by purification on a Qiagen column. For protein analysis,

beads were directly eluted in 2X Sample Buffer, incubated for 10

minutes at 100ºC and centrifuged.

Quantification of bound nucleosomes

The recovered DNA was quantified with a Quant-iT PicoGreen

dsDNA assay kit (Invitrogen). Quantification of nucleosome occupancy

at specific loci was performed by real-time PCR. The recovered DNA

was quantified in triplicate by real time PCR using iTaq Universal

SYBR Green supermix (Bio-Rad) and the 7900HT Fast Real-time PCR

System (Applied Biosystems). Quantification of DNA was achieved by

comparing qPCR signals to those obtained from a standard curve

derived from 10 fold serial dilutions of the input DNA which also served

as a measure of the linear dose response of the qPCR reaction.

In addition to the qPCR standard curve, a TimeChIP standard

curve was generated for each pulse-chase experiment to determine

the dose-response of the qPCR to the fraction of biotinylated

nucleosomes. The TimeChIP standard curve was generated by mixing

of equal amounts of in vivo pulse labeled cells expressing SNAP-

tagged histones mixed with unlabeled cells in a 2 fold ratio series. The

quantitative value of TimeChIPed DNA of the chase samples at each

locus was interpolated on the TimeChIP standard curve. The

Page 83: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

63

TimeChIP signal was defined as the fraction of biotin retained relative

to the standard curve.

Immunofluorescence

HeLa cells were grown on glass coverslips coated with poly-L-

Lysine (Sigma) and fixed with 4% formaldehyde (Thermo Scientific) for

10 minutes. Cells were extracted after fixation and processed for

immunofluorescence using standard procedures. Cells were stained

with 1µg/mL anti-HA (clone HA11, Covance) and FITC-conjugated

anti-mouse (Jackson Immunoresearch Laboratories). Samples were

stained with DAPI (4’,6-diamidino-2-phenylindole; Sigma) before

mounting in Mowiol (Calbiochem).

Microscopy

Widefield fluorescence microscopy was performed using a

DeltaVision Core System (Applied Precision) that controls an inverted

microscope (Olympus, IX-71), coupled to a Cascade 2 EMCCD

camera (Photometrics). 512 by 512 pixel images were collected at 1x

binning using a 100x, 1.4 NA oil immersion objective (UPIanSApo) at

0,2µm axial sections spanning the entire nucleus. Quantified images

were acquired using the same exposure conditions for each

fluorescent channel. TRITC and FITC images of a uniformly slide were

automatically flatfield- and camera-noise-corrected using softWoRx

(Applied Precision). All images presented are maximum intensity

projections of deconvolved pictures.

Centromeric TMR-Star fluorescence intensity was quantified

using CRaQ, a macro specifically developed for ImageJ (NIH). For

specific details about methods and parameters of CRaQ see Bodor et

al., 2012.

Page 84: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

64

Immunoblotting

Proteins were separated in 12% SDS/PAGE gel and

transferred to Hybond PVDF membranes (GE Healthcare) using

standard procedures. Blots were probed with anti-HA (Covance) at

1:2000 dilution. Anti-mouse HRP conjugated secondary antibody was

purchased from Jackson Immunoresearch Laboratories.

Flow cytometry

HeLa CENP-A-SNAP cells (106) were harvested and fixed 1

hour at 4ºC with 70% ethanol. Cells were washed twice in PBS

containing 3% BSA (Sigma) and incubated for 30 minutes at room

temperature with 5µg/mL propidium iodide (Sigma) and 200µg/mL of

RNaseA in PBS containing 3% BSA. Subsequently flow cytometry

analysis was performed on FACScan (Becton Dickinson) using

CellQuest software.

Page 85: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

65

RESULTS

TimeChIP strategy

To determine aspects of protein dynamics in living cells,

common approaches include fluorescence recovery after photo

bleaching (FRAP), photoactivation, among other novel techniques (see

introduction above) that asses turnover at specific loci at a relatively

short time frame. Alternatively, high resolution localization to a specific

genomic region is achieved through chromatin immuno precipitation

(ChIP), but this approach cannot assess protein dynamics. A strategy

that combines both protein dynamics and localization at high genomic

resolution is currently lacking. We sought to develop a biochemical

approach to determine histone dynamics in living cells at specific loci

using the SNAP-tag system (TimeChIP, see figure 2.3)

Page 86: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

66

Figure 2.3 Outline of the TimeChIP assay. A) Cells stably expressing H3.1-

or H3.3-SNAP-HA are in vivo pulse labeled with BG-biotin at time 0 (blue).

Cells continue to synthesize H3-SNAP protein that is not labeled, while the

H3-SNAP biotinylated pool turns over. Cells are harvested and nuclei are

isolated by mechanical forces using a Dounce homogenizer in the presence

of 0.1% Digitonin. Salt extracted chromatin is fragmented to

mononucleosomes with Micrococcal nuclease and soluble biotinylated H3-

SNAP nucleosomes are affinity purified with Streptavidin magnetic beads. B)

Detection of pulse labeled histone. Soluble H3.3-SNAP nucleosomes either

pulse labeled or unlabeled were purified and separated by SDS-PAGE,

blotted and detected using an anti-HA antibody. C) DNA associated with

purified pulse labeled H3.3-SNAP nucleosomes can be identified by

Page 87: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

67

quantitative (q) PCR. Biotinylation dependent enrichment for the ACTB gene

is shown.

HeLa cells stably expressing histone-SNAP-3XHA cells were in

vivo pulse labeled with benzylguanine (BG) conjugated with a biotin

moiety. The specific and tight binding of biotin-streptavidin makes it a

widely used system for labeling, purification and immobilization of

protein complexes. Once cells were collected, chromatin was extracted

and fragmented into mononucleosomes by MNase treatment. Histone-

SNAP-biotinylated nucleosomes were captured on Streptavidin

magnetic beads. Recovery of SNAP-tagged protein as well as the

presence of co-purified DNA by western blot and spectrophotometry

respectively was assessed followed by the determination of the

localization of tagged histones at a particular locus. The resulting

protocol is outlined in figure 2.3. Unlike strategies based upon

antibody-antigen specificity, TimeChIP allows to purify the protein of

interest depending on self-labeling of the SNAP-tag with high affinity

given by biotin-streptavidin interaction. To minimize non-specific

background binding of DNA, we used yeast tRNA to block the

Streptavidin solid phase. Finally, the isolation of chromatin in a native

manner provides single nucleosome resolution to the assay.

To make TimeChIP a suitable assay for the study of

nucleosome dynamics, the following parameters were assessed:

labeling efficiency, mononucleosome isolation and purification. In

contrast to fluorescent BG substrates, nonfluorescent BG or BTP

enters cells efficiently (Bodor et al., 2012). We tested in vivo and in

vitro labeling of CENP-A-SNAP cells using the BG-biotin substrate to

determine the efficiency of labeling. CENP-A-SNAP cells were labeled

in cell culture media at 37ºC for 1 hour either as intact cells or as

permeabilized cells in buffer containing nonionic detergent at room

temperature for 1 hour as well. After purification of biotinylated

Page 88: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

68

proteins, the protein levels were detected by western blot. We found

that labeling of CENP-A-SNAP in conditions where the cell membrane

is permeabilized with nonionic detergents is improved (Figure 2.4 A).

This suggests that cell permeability is a rate limiting step in SNAP

labeling with BG-biotin. Furthermore, we tested a novel variant of BG-

biotin, CP-biotin, whose structure is modified to facilitate crossing cell

membrane (synthesized by Ivan Correa, New England Biolabs,

Ipswich, MA). We found that the amount of H3.3 purified increases

~1.5 fold when H3.3-SNAP is labeled with CP-biotin in comparison

with BG-biotin (Figure 2.4 B).

Recently, new variants of both the SNAP and CLIP tags have

been developed (named SNAPf and CLIPf, respectively) that show an

improved reaction kinetics (Pellett et al., 2011; Sun et al., 2011). We

fused these novel tags to CENP-A. Cells transiently transfected with

CENP-A-SNAP, CENP-A-CLIP, CENP-A-SNAPf or CENP-A-CLIPf

fusion proteins were in vivo fluorescence pulse labeled with BG

carrying TMR-STAR at different concentrations and incubation times.

The activity of the tags was determined by measuring the TMR-Star

fluorescence relative to HA fluorescence intensity and normalized to

the standard conditions. HA is encoded by the SNAP transgene and

therefore provides a measure of the protein level independent of

labeling activity. While CLIPf showed only a modest improvement over

CLIP, SNAPf performed ~3 to 5 fold better across different

concentrations of substrates and incubation times (Figure 2.5). Next,

we sought to create stable S3 HeLa and HeLa cell lines expressing

HA-SNAPf-PreScission-CENP-A but efforts to isolate cell lines bearing

properly localized tagged-CENP-A failed suggesting that the tag

interfered with CENP-localization.

Obtaining a homogeneous preparation of soluble

mononucleosomes is an important factor in the successful application

Page 89: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

69

of this assay. It is known that the histone octamer can be extracted

from DNA at 2M NaCl, while lower concentrations result in the

dissociation of H2A-H2B dimers leaving behind the H3-H4 tetramer

(Eickbush and Moudrianakis, 1978). We used 300mM NaCl to extract

chromatin which leaves the full nucleosome intact but removes non-

chromatin proteins and low-affinity chromatin interacting proteins.

Histone octamers protect ~147bp of DNA from Micrococcal nuclease

(MNase) digestion (Axel, 1975; Luger et al., 1997). We used MNase

protection as parameter for the definition of nucleosomes isolated at

0.3M NaCl as well as a method to isolate mono or oligo nucleosomes

(Figure 2.4 C). By titrating the MNase concentration and incubation

time we optimized conditions to maximize the yield of

mononucleosomes. Finally, although the biotin-streptavidin purification

system is based on the tight and essentially irreversible complex that

biotin forms with streptavidin; we tested the efficiency of binding of the

biotinylated SNAP-tag to streptavidin magnetic beads. T98G cells

transiently expressing SNAP-HA tags were in vivo biotinylated and

soluble labeled SNAP-HA (non histone fused) was immobilized on

streptavidin magnetic beads. In order to test whether all the biotin-

labeled protein binds to streptavidin, the unbound fraction was re-

incubated with fresh streptavidin beads. Recovery of any SNAP-HA

from this unbound fraction would indicate the initial binding reaction did

not deplete all protein during the first binding step. We found that all

biotinylated SNAP-HA protein binds efficiently to streptavidin, as no

additional protein could be recovered using fresh streptavidin beads

(Figure 2.4 D). In addition we tested whether all biotinylated proteins

are eluted from the beads by re-eluting beads a second time (Figure

2.4 D).

Page 90: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

70

Figure 2.4 TimeChIP optimization. A) Western blot analysis of CENP-A-

SNAP labeling of intact cells vs. permeabilized cells. Cells were either

unlabeled or labeled with BG-biotin for 1 hour. Soluble nucleosomes were

prepared and bound to streptavidin beads. Bound material was eluted,

separated by SDS-PAGE, blotted and detected using an anti-HA antibody.

Alternatively (right), 0.1% recrystallized Digitonin was included during the

biotinylation reaction to permeabilized cells. Percentage indicates the % of

sample loaded. B) Experiment to compare the efficiency of biotinylation using

BG-biotin or CP-biotin. Intact H3.3-SNAP expressing cells were pulse labeled

under identical conditions with either CP-biotin, BG-biotin or were left

unlabeled. Chromatin was extracted and solubilized by MNase. Nucleosomes

Page 91: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

71

were bound to streptavidin beads and the quantification of co-bound DNA was

determined by qPCR for the ACTB gene. C) Generation of soluble

mononucleosomes. Salt extracted chromatin was digested with Micrococcal

Nuclease in a 2 fold dilution series. Following digestion, DNA was isolated

and analyzed by agarose gel electrophoresis and run on a Bioanalyzer for

quantification. Size of fragments is indicted as well as concentration of

mononucleosome peak. D) Determination of binding efficiency. Left: T98G

cells expressing SNAP-HA were either unlabeled or BG-biotin pulse labeled.

Soluble extract was bound to streptavidin beads. Input, unbound and bound

fractions were separated on SDS-PAGE and analyzed by Western blot to

detect in vivo biotinylated SNAP-HA using an anti-HA antibody. Both primary

and secondary elutions were analyzed (compare lanes 6 & 7). The unbound

fraction (lane 5 and 8) was tested for the presence of residual biotinylated

SNAP-HA by reincubation with streptavidin to detect biotinylated SNAP-HA

not purified during the first purification. Lane 9 shows that very little SNAP-HA

binds to streptavidin in the second purification (indicating that unbound SNAP-

HA is unlabeled). Right: Pulse labeling of T98G cells expressing SNAP-HA

cells with TMR-Star show SNAP-HA is diffusely localized.

Page 92: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

72

Figure 2.5 Evaluation of SNAPf-tag performance. A) HeLa cells transiently

transfected with either CENP-A-SNAP, CENP-A-SNAPf, CENP-A-CLIP or

CENP-A-CLIPf fusion proteins were labeled with TMR-Star at different

concentrations and incubation times and processed for immunofluorescence

with anti-HA. Representative images of cells are shown with TMR-Star signals

in green and DAPI (DNA) in blue. B) TMR-Star and HA fluorescence intensity

were determined using CRaQ. TMR/HA ratios are a measure of SNAPf and

CLIPf activity. Results are plotted as fold difference normalized against

signals obtained after incubation with 2µM TMR-Star for 15 minutes.

Proof of Principle

Following the initial pilot experiments to determine the feasibility

of purifying biotin-pulse labeled SNAP from cells, we adopted the

method to apply to the purification of histone H3 variants. Histone

variants are implicated in epigenetic memory, however their dynamics

at specific loci is poorly defined (Gómez-Rodríguez and Jansen, 2013).

The reported dynamics of H3 variants varies from less than an hour to

days (Bodor et al., 2013; Deal et al., 2010; Jansen et al., 2007; Kimura

and Cook, 2001; Radman-Livaja et al., 2011; Xu et al., 2010). Using

Page 93: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

73

our TimeChIP strategy, histone dynamics is measured by pulse

biotinylation of SNAP-tagged histones in chromatin. The fraction of

biotinylated histone retained in nucleosomes diminished over time as a

consequence of turnover. First, we tested the dynamic range of the

assay indicating the sensitivity at which we can detect retention of

histones. Equal amounts of in vivo labeled HeLa H3.1-SNAP cells

were mixed at different ratios with unbiotinylated HeLa H3.1-SNAP

cells. These mixtures were then assayed by the TimeChIP protocol

and the genomic DNA recovered was quantified as a measure of the

amount of histone retained. We found a linear response corresponding

to the 2 fold serial dilutions of the biotinylated H3.1 cells indicating

there is good agreement between the degree of biotinylation and the

amount of DNA recovered. Moreover, we found that with the TimeChIP

assay we can detect a minimum of 6.25% of histone retained (Figure

2.6).

Figure 2.6 Proof of principle. In vivo pulse labeled H3.1-SNAP cells are

mixed with unlabeled H3.1-SNAP cells in a 2 fold serial dilution, subsequently

native chromatin is isolated from the 6 different pools of cells. Nucleosomes

are purified by immobilization on Streptavidin. Total DNA purified was

Page 94: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

74

quantified with a PicoGreen Assay. The graph indicates average of five reads

of the amount of genomic DNA that is recovered and indicates the dose-

response of DNA recovery as a function of biotinylation.

Pulse-chase with TimeChIP

SNAP fluorescence-based experiments have shown that

CENP-A is stably localized at centromeres. To test the TimeChIP

procedure in a pulse-chase labeling setting we used CENP-A-SNAP

as a trail, since its in vivo stability has been previously established

(Bodor et al., 2013). We performed pulse-chase experiments in

asynchronous CENP-A-SNAP cells. The primary locus for CENP-A

assembly is on α-satellite repeats, the major DNA component of the

centromeres (Vafa and Sullivan, 1997). We measured the enrichment

of CENP-A nucleosomes at α-satellite I, pericentromeric domain (Sat2)

and 5SrDNA (Figure 2.7 A). As expected, CENP-A is most enriched at

α-satellite, while levels drop to approximately half of that at the

5SrDNA and are reduced to one fourth at the pericentromeric domain.

When normalizing to the initial enrichment for each locus, after 12

hours of chase, approximately 50% retention of CENP-A was seen at

all loci tested. While CENP-A-SNAP levels were eventually reduced to

background levels at all loci, levels remained high (~40%) at α-satellite

I and Sat2 repeat. At 5SrDNA turnover appears to be faster (Figure 2.7

B). These results suggest that CENP-A is stably retained at

centromeres and pericentromeric domains during the cell cycle

compared with the retention at ribosomal 5SrDNA repeats.

Page 95: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

75

Figure 2.7 CENP-A-SNAP Pulse-chase with TimeChIP. A) Quantification by

qPCR of total enrichment of CENP-A at α-satellite I, pericentromeric domains

(Sat2) and 5SrDNA. Average %IP is plotted and SEM is shown for two

independent experiments. B) Time course of CENP-A-SNAP pulse-chase

shows quantification by qPCR of CENP-A retained at α-satellite I,

pericentromeric domains (Sat2) and 5SrDNA. CENP-A is more stably retained

at α-satellite I and Sat2 in comparison to 5SrDNA where the turnover is faster.

Quench-chase-pulse with TimeChIP

The use of the SNAP system combining fluorescent and

nonfluorescent SNAP substrates in a quench-chase-pulse experiment,

allows for determining the dynamics and fate of the nascent protein

pool. Cells are quenched using the unconjugated SNAP substrate BTP

(no dye). Following the quench of the ancestral pool cells are chased

during which a nascent unlabeled pool is synthesized. This pool can

subsequently be labeled using a labeled substrate. This quench-

Page 96: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

76

chase-pulse strategy has been used to show that in contrast to

canonical H3 and H3.3, CENP-A is loaded during G1 phase (Jansen et

al., 2007). To determine the utility of TimeChIP for the performance of

quench-chase-pulse experiments, we tested whether the assembly of

nascent CENP-A into chromatin also occurs in early G1 phase which is

the cell cycle window during which new CENP-A is visibly targeted to

the centromere. Alternatively, incorporation into chromatin occurs at a

later stage in the cell cycle. First, we aimed at detecting newly

synthesized nucleosomal CENP-A by western blot. Asynchronous

CENP-A-SNAP cells were quenched with BTP. After 16 hours of

chase, CENP-A-SNAP cells were pulse labeled with BG-biotin,

followed by another 24 hour chase to allow newly synthesized pulse

labeled histone to be assembled into centromeric chromatin. Following

24 hours of chase, nuclei were isolated, mononucleosomes were

liberated by MNase digestion and biotinylated CENP-A-SNAP was

immobile on streptavidin beads. We show that using this Quench-

Chase-Pulse-Chase TimeChIP procedure, nascent CENP-A assembly

into chromatin can be detected, indicating that CENP-A-SNAP is

incorporated into salt-stable chromatin. As expected, the levels of

nascent nucleosomal CENP-A detected are significantly lower to the

total CENP-A-SNAP levels (Compare lanes 3 with 9 in Figure 2.8).

Nevertheless, biotinylated CENP-A-SNAP protein can be readily

detected. As described before, the in vitro labeling is higher in

comparison with in vivo labeling of SNAP with the BG-biotin substrate

(Figure 2.4A). To test whether newly synthesized CENP-A is

assembled into chromatin at centromeres or elsewhere during early

G1 phase we used TimeChIP with in vitro pulse labeling conditions. A

synchronous population of CENP-A-SNAP cells arrested at the G1-S

boundary by double thymidine block, were quenched with BTP in the

presence of thymidine. The cells were then released into S phase for 3

hours. In order to keep the cells tightly synchronized in G2 phase, the

Page 97: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

77

CDK1 inhibitor RO3306 was added for 8 hours (Vassilev et al., 2006).

This treatment arrest cells in late G2 phase preventing mitotic entry. To

ensure mitotic arrest, cells were released from RO3306 into MG132 for

3 hours which will arrest cells in metaphase of mitosis. Cells were then

release by washout of MG132. Nascent CENP-A-SNAP assembly was

analyzed in cells that remained arrested at the G1/S phase boundary,

in mitosis, 3 and 11 hours after mitotic exit (Figure 2.9 A). Microscopy

analysis showed that nascent CENP-A localizes at centromeres after 3

and 10 hours of mitotic exit (Figure 2.9 C). Strikingly, new CENP-A

was detected in chromatin already in mitosis as well as early and late

G1 by western blot (Figure 2.9 D). This suggests that while CENP-A is

targeted to centromeres only in G1 phase; it may already be

incorporated into general chromatin prior to mitotic exit. To determine

where nucleosomal CENP-A accumulates we analyzed the TimeChIP

precipitates for three families of DNA repeats, α-satellite I,

pericentromeric domains (Sat2) and 5SrDNA genes. New CENP-A is

detected in early G1 phase at centromeres, however, new CENP-A

incorporated at α-satellite I chromatin is almost exclusively detected in

late G1, suggesting that CENP-A loads into centromeric chromatin in

late G1. However, after 3h chase (early G1) there is a significant

proportion of cells still in mitosis making it difficult to determine whether

no assembly into α-satellite I occurs at this early stage. Interestingly,

new CENP-A is not only assembled at α-satellite I but also to a lesser

extent at repeats of pericentromeric domains and 5SrDNA gene (as

observed by pulse labeling experiments above, Figure 2.7). The

accumulation of this non-centromeric signal may indicate a low level of

promiscuity in the CENP-A assembly machinery.

Page 98: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

78

Figure 2.8 Detection of nascent nucleosomal CENP-A. A) In vivo pulse

labeling scheme of newly synthesized nucleosomal CENP-A-SNAP by

quench-chase-pulse-chase TimeChIP. CENP-A-SNAP-HA cells are quenched

using 5µM BTP (unconjugated SNAP substrate), then ancestral CENP-A is

chased for 16 hours during which a nascent unlabeled pool is synthesized.

Nascent CENP-A is pulse labeled with 10µM BG-biotin and chase for 24

hours where is incorporated onto chromatin. B) Western blot analysis of

nascent chromatin incorporated CENP-A. Soluble nucleosomes were

prepared and bound to Streptavidin beads from cells that underwent the

quench-chase-pulse-chase procedure. Samples were separated by SDS-

PAGE, blotted and detected using an anti-HA antibody. Lane 3 shows newly

synthesized CENP-A incorporated into chromatin after 24 hours chase. Lane

6 shows that CENP-A is not detected when no time is allowed for new protein

synthesis between quench and pulse labeling with a BG-biotin pulse. Lane 9

shows labeling of the total CENP-A pool.

Page 99: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

79

Figure 2.9 Incorporation of newly synthesized CENP-A into chromatin

during the cell cycle. A) Schematic of cell synchronization and labeling of

chromatin incorporated nascent CENP-A. B) FACS analysis of DNA content

to monitor cell cycle position of CENP-A-SNAP cells arrested in mitosis,

released and progressed in G1 phase. Estimated fractions of cells in

respective cell cycle positions is indicated. C) Visualization of new CENP-A at

centromeres by TMR-Star labeling of nascent pool. Microscopy analysis of

pulse labeled cells reveals CENP-A targeting to centromeres in early and late

G1 phase. D) Newly synthesized CENP-A pull down in a biotin dependent

manner in mitosis, early and late G1 phase. Nascent nucleosomal (left) and

soluble (right) CENP-A in mitosis, early and late G1 phase were separated by

SDS-PAGE and detected with anti-HA antibody. CENP-A-SNAP is detected in

the input and unbound blots of the chromatin and soluble fraction in all

conditions. Nascent nucleosomal CENP-A is detected in chromatin bound

fractions in mitosis, 3h chase, late G1 (lanes 2, 3 and 4). Soluble CENP-A is

not detected in mitosis, early and late G1 (lanes 7, 8 and 9). As expected,

Page 100: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

80

CENP-A quenched with unconjugated SNAP substrate (BTP) and

subsequently in vitro pulsed with BG-biotin without a chase period (quench-

pulse; lanes 1 and 5) could not be detected. Membranes were stained with

Ponceau to detected histones as a loading control and a measure of purity of

soluble fractions. E) Quantification of CENP-A incorporated in chromatin by

qPCR. Amount of DNA associated to CENP-A nucleosomes quantified by

qPCR (%IP) for α-satellite I, pericentromeric domains (Sat2) and ribosomal

loci in mitosis, early and late G1 phase. Enrichment of CENP-A is observed in

late G1 phase at the three different loci by comparing with quench-pulse

CENP-A.

Page 101: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

81

DISCUSSION AND CONCLUSIONS

The comprehension of histone dynamics is important for

understanding the contribution of chromatin to epigenetic inheritance.

Thus far, chromatin dynamics has been analyzed using bulk pulse

labeling of the amino acids, FRAP of GFP-tagged histones, conditional

expression of tagged histones and self-labeling tags. Labeling of

histones by the use of isotope labeled amino acids or amino acid

analogs in SILAC and CATCH-IT allows for the analysis of protein

complex turnover and, in the case of the latter, provide spatial

resolution during turnover without the need to use of tags (Deal et al.,

2010; Jackson, 1990; Xu et al., 2010). RITE which depends on the

conditional induction and concurrent loss of differentially tagged

histones, allows to measure old and new pools of histones

simultaneously at DNA sequence resolution (Radman-Livaja et al.,

2011; Verzijlbergen et al., 2010). FRAP and SNAP-tag fluorescent

pulse labeling has the power to uncover variation in dynamics within

the cell population or during the cell cycle because the analysis is

performed on single cells. While FRAP provides a high temporal

resolution from seconds to minutes, SNAP-tag based analysis

operates on a longer time scale from hours to days (Bodor et al., 2012,

2013; Jansen et al., 2007; Kimura and Cook, 2001). Although, these

methods are powerful and have provided insight on histone dynamics,

what is lacking is a measure of histone turnover at specific loci. The

RITE assay has the capacity to do this but requires highly efficient Cre-

mediated recombination of the gene locus expressing tagged-histones

which is not feasible in human cells.

Histone H3 variants are of particular interest to study due to

their implication in epigenetic processes. Canonical H3 (H3.1) is

assembled strictly in S phase, CENP-A assembly is restricted to early

G1 phase, while H3.3 assembles throughout the cell cycle (Bodor et

Page 102: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

82

al., 2013; Jansen et al., 2007; Ray-Gallet et al., 2011). Each variant

has a particular localization. H3.1 is widely distributed throughout

chromatin, H3.3 is ubiquitously localized at promoters and gene body

of active genes, enhancers and subtelomeric regions, while CENP-A

uniquely loads at centromeres (Goldberg et al., 2010; Vafa and

Sullivan, 1997).

In this chapter I describe the development of the TimeChIP

strategy for use in analyzing histones dynamics. Critical parameters to

be considered are the SNAP labeling, fragmentation of chromatin by

Micrococcal nuclease and the efficiency of chromatin purification.

Regarding SNAP labeling we refer to two main aspects: the SNAP

substrate and the SNAP tag. We conclude that a limiting factor for the

assay is the cell permeability of SNAP substrates. We observed that

when cell membrane is permeabilized the SNAP substrate significantly

improve the reaction with the tag, an approach that should be

considered for experiments like quench-chase-pulse or pulse. For

labeling of intact cells, CP-biotin provides an improvement of labeling

over BG-biotin. In relation to the tag itself, we saw that the faster

variant of SNAP (SNAPf) exhibits a faster reaction kinetics which

favors the in vivo labeling step as a larger fraction of SNAPf can be

labeled within the same timeframe and substrate concentration. The

second parameter is fragmentation of chromatin. To obtain high spatial

resolution of histone turnover, soluble chromatin should be fragmented

to mononucleosomes. By titration of MNase incubation times we

optimize conditions such as to maximize the amount of

mononucleosomes and limiting the amount of di- tri-nucleosomes or

over digestion of chromatin. We find that this should be re-optimized

for each batch of MNase. This is also important in the context of deep

sequencing of isolated material (see chapter 3) where MNase

digestion can create a bias in detection of specific sequences (Rizzo et

Page 103: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

83

al., 2012). The last parameter that we highlight is the use of blocking

reagents for the purification of the protein. Although we used one of

the strongest non-covalent interactions biotin-streptavidin for the

purification of histones, to find the appropriate buffer conditions for the

binding reaction was important. To avoid non-specific binding we found

that the use of bovine serum albumin, yeast tRNA and non ionic

detergent NP40 were important factors to block unoccupied binding

sites and dislodge loosely bound molecules, respectively. Overall, the

major limitation of the strategy is the inefficient labeling of intact cells

by SNAP substrates that due to cell impermeability. While this may be

a limiting factor for the analysis of CENP-A which is low in abundance,

highly abundant proteins such as H3.1 and H3.3 can still successfully

be analyzed (see chapter 3).

CENP-A nucleosome dynamics along the cell cycle

Centromeres are composed of α-satellite repeats that constitute

5% of total human DNA but the number of CENP-A nucleosomes per

centromere is likely much smaller than the total number of

nucleosomes occupying α-satellite arrays (D. Bodor, personal

communication). Because CENP-A is relatively low in abundance

compared to other histones and is broadly distributed across large

alphoid arrays it served to test the detection limit of the TimeChIP

method. We show that CENP-A-SNAP is stably retained as has been

shown by FRAP and SNAP fluorescent pulse labeling (Hemmerich et

al., 2008; Jansen et al., 2007). We take this observation beyond the

general stability of CENP-A by demonstrating CENP-A dynamics at

different loci. Specifically, we find that CENP-A is stable at α-satellite I

repeats. Furthermore, our TimeChIP analysis suggests that CENP-A

turnover is not only slow at centromeres but also at pericentromeric

domains, even though CENP-A levels appear to be lower at these loci.

In contrast, the enrichment of CENP-A at 5SrDNA repeats that can be

Page 104: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

84

detected during the first 12 hours is rapidly lost in subsequent time

points indicating that outside the centromere domain, CENP-A is not

only lower in abundance but also turns over faster. Quench-chase-

pulse analysis suggests that CENP-A is incorporated into

nucleosomes in G1 phase. Interestingly, assembly of CENP-A is not

restricted to alphoid DNA but is also observed at major satellites and at

the rDNA repeats. In addition, we detect nascent CENP-A-SNAP

protein in chromatin before detection of specific DNA loci suggesting

that low levels of CENP-A assembly throughout chromatin may occur

as early as in mitosis. Taken together, we speculate that CENP-A not

only is targeted at centromeres in G1 phase where it is stably

maintained but that there is also a subpopulation of CENP-A that

assembles distal to centromeres and turns over faster. An explanation

is that like the labile histone H3.3-H2A.Z variant nucleosome shown in

human cells (Jin et al., 2009), the population of CENP-A with fast

turnover constitute a CENP-A-H2A.Z double variant nucleosome

where H2A.Z is less stable as has been described in S. cerevisiae

(Meneghini et al., 2003). Interestingly, purification of the CENP-A

nucleosome complex indicates that CENP-A containing nucleosomes

are likely to contain the histone H2A.Z variant (Foltz et al., 2006).

Alternatively, the same study identified a proportion of CENP-A-H3

hybrid nucleosomes which may confer a different stability.

Our ability to detect CENP-A by TimeChIP led us to question

whether assembly into chromatin of newly synthesis CENP-A-SNAP

occurs in early G1 as microscopy analysis has indicated or whether,

nucleosome assembly is delayed following initial targeting. Our results

indicate that at present we do not have the resolution to determine

CENP-A turnover dynamics. Clear enrichment of CENP-A into

centromeric chromatin is observed only in samples enriched in late G1

phase. However, the fact that we are unable to obtain a homogenous

Page 105: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

85

population of early G1 cells does not allow us to draw conclusion on

the precise timing of CENP-A assembly into centromeric chromatin. In

addition, although our PCR probes detect an abundant class of alphoid

repeats only a small subset of these are likely occupied by CENP-A at

any time thereby reducing our sensitivity. The development of SNAP

substrates with higher capability to cross the cell membrane will

significantly contribute to the performance of in vivo pulse labeling

experiments and may help the analysis of CENP-A dynamics in

chromatin in future efforts.

A comprehensive understanding of protein function includes the

elucidation of its dynamics. The role of chromatin in epigenetic process

has been assessed through histone modifications and their regulation.

We show here that TimeChIP as a method for the study of protein

dynamics provides: 1) the ability to asses protein dynamics at long

time scales, 2) analysis in living cells, 3) distinction between ancestral

from newly synthesized pools of proteins, 4) high spatial resolution.

Thus, TimeChIP is potentially a versatile method for the analysis of site

specific histone dynamics such as turnover in living cells with which we

may gain insight in the relationship between histone dynamics and

epigenetic memory as well as the molecular mechanisms involved.

Page 106: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

86

ACKNOWLEDGEMENTS

I would like to acknowledge Ivan Correa from New England

Biolabs, Ipswich, MA, for the synthesis of the CP-biotin substrate and

SNAP-tag variants. I thank Dani Bodor for developing the CRaQ macro

we used for fluorescence intensity quantifications.

Page 107: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

87

REFERENCES

Ahmad, K., and Henikoff, S. (2002). The Histone Variant H3.3 Marks Active Chromatin by Replication-Independent Nucleosome Assembly. Mol. Cell 9, 1191–1200.

Aida, M., Hamad, N., Stanlie, A., Begum, N.A., and Honjo, T. (2013). Accumulation of the FACT complex, as well as histone H3.3, serves as a target marker for somatic hypermutation. Proc. Natl. Acad. Sci. 110, 7784–7789.

Allshire, R.C., and Karpen, G.H. (2008). Epigenetic regulation of centromeric chromatin: old dogs, new tricks? Nat. Rev. Genet. 9, 923–937.

Annunziato, A.T. (2005). Split Decision: What Happens to Nucleosomes during DNA Replication? J. Biol. Chem. 280, 12065–12068.

Axel, R. (1975). Cleavage of DNA in nuclei and chromatin with staphylococcal nuclease. Biochemistry (Mosc.) 14, 2921–2925.

Bannister, A.J., and Kouzarides, T. (2011). Regulation of chromatin by histone modifications. Cell Res. 21, 381–395.

Bannister, A.J., Zegerman, P., Partridge, J.F., Miska, E.A., Thomas, J.O., Allshire, R.C., and Kouzarides, T. (2001). Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain. Nature 410, 120–124.

Barth, T.K., and Imhof, A. (2010). Fast signals and slow marks: the dynamics of histone modifications. Trends Biochem. Sci. 35, 618–626.

Belotserkovskaya, R., Oh, S., Bondarenko, V.A., Orphanides, G., Studitsky, V.M., and Reinberg, D. (2003). FACT facilitates transcription-dependent nucleosome alteration. Science 301, 1090–1093.

Bergmann, J.H., Rodríguez, M.G., Martins, N.M.C., Kimura, H., Kelly, D.A., Masumoto, H., Larionov, V., Jansen, L.E.T., and Earnshaw, W.C. (2011). Epigenetic engineering shows H3K4me2 is required for HJURP targeting and CENP-A assembly on a synthetic human kinetochore. EMBO J. 30, 328–340.

Page 108: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

88

Bickmore, W.A., and van Steensel, B. (2013). Genome architecture: domain organization of interphase chromosomes. Cell 152, 1270–1284.

Bintu, L., Kopaczynska, M., Hodges, C., Lubkowska, L., Kashlev, M., and Bustamante, C. (2011). The elongation rate of RNA polymerase determines the fate of transcribed nucleosomes. Nat. Struct. Mol. Biol. 18, 1394–1399.

Bird, A. (2002). DNA methylation patterns and epigenetic memory. Genes Dev. 16, 6–21.

Biswas, M., Voltz, K., Smith, J.C., and Langowski, J. (2011). Role of histone tails in structural stability of the nucleosome. PLoS Comput. Biol. 7, e1002279.

Black, B.E., Foltz, D.R., Chakravarthy, S., Luger, K., Woods, V.L., Jr, and Cleveland, D.W. (2004). Structural determinants for generating centromeric chromatin. Nature 430, 578–582.

Blower, M.D., and Karpen, G.H. (2001). The role of Drosophila CID in kinetochore formation, cell-cycle progression and heterochromatin interactions. Nat. Cell Biol. 3, 730–739.

Bodor, D.L., Rodríguez, M.G., Moreno, N., and Jansen, L.E.T. (2012). Analysis of protein turnover by quantitative SNAP-based pulse-chase imaging. Curr. Protoc. Cell Biol. Editor. Board Juan S Bonifacino Al Chapter 8, Unit8.8.

Bodor, D.L., Valente, L.P., Mata, J.F., Black, B.E., and Jansen, L.E.T. (2013). Assembly in G1 phase and long-term stability are unique intrinsic features of CENP-A nucleosomes. Mol. Biol. Cell 24, 923–932.

Bönisch, C., and Hake, S.B. (2012). Histone H2A variants in nucleosomes and chromatin: more or less stable? Nucleic Acids Res.

Brennecke, J., Aravin, A.A., Stark, A., Dus, M., Kellis, M., Sachidanandam, R., and Hannon, G.J. (2007). Discrete Small RNA-Generating Loci as Master Regulators of Transposon Activity in Drosophila. Cell 128, 1089–1103.

Calabrese, J.M., Sun, W., Song, L., Mugford, J.W., Williams, L., Yee, D., Starmer, J., Mieczkowski, P., Crawford, G.E., and Magnuson, T. (2012). Site-specific silencing of regulatory elements as a mechanism of X inactivation. Cell 151, 951–963.

Page 109: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

89

Cassé, C., Giannoni, F., Nguyen, V.T., Dubois, M.F., and Bensaude, O. (1999). The transcriptional inhibitors, actinomycin D and alpha-amanitin, activate the HIV-1 promoter and favor phosphorylation of the RNA polymerase II C-terminal domain. J. Biol. Chem. 274, 16097–16106.

Chanoine, C., Della Gaspera, B., and Charbonnier, F. (2004). Myogenic regulatory factors: Redundant or specific functions? Lessons from Xenopus. Dev. Dyn. 231, 662–670.

Chen, S., Bohrer, L.R., Rai, A.N., Pan, Y., Gan, L., Zhou, X., Bagchi, A., Simon, J.A., and Huang, H. (2010). Cyclin-dependent kinases regulate epigenetic gene silencing through phosphorylation of EZH2. Nat. Cell Biol. 12, 1108–1114.

Cleveland, D.W., Mao, Y., and Sullivan, K.F. (2003). Centromeres and Kinetochores: From Epigenetics to Mitotic Checkpoint Signaling. Cell 112, 407–421.

Deal, R.B., Henikoff, J.G., and Henikoff, S. (2010). Genome-Wide Kinetics of Nucleosome Turnover Determined by Metabolic Labeling of Histones. Science 328, 1161–1164.

Dodd, I.B., Micheelsen, M.A., Sneppen, K., and Thon, G. (2007). Theoretical Analysis of Epigenetic Cell Memory by Nucleosome Modification. Cell 129, 813–822.

Drané, P., Ouararhni, K., Depaux, A., Shuaib, M., and Hamiche, A. (2010). The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev. 24, 1253–1265.

Dunleavy, E.M., Almouzni, G., and Karpen, G.H. (2011). H3.3 is deposited at centromeres in S phase as a placeholder for newly assembled CENP-A in G₁ phase. Nucl. Austin Tex 2, 146–157.

Easwaran, H.P., Schermelleh, L., Leonhardt, H., and Cardoso, M.C. (2004). Replication-independent chromatin loading of Dnmt1 during G2 and M phases. EMBO Rep. 5, 1181–1186.

Eickbush, T.H., and Moudrianakis, E.N. (1978). The histone core complex: an octamer assembled by two sets of protein-protein interactions. Biochemistry (Mosc.) 17, 4955–4964.

ENCODE Project Consortium (2011). A user’s guide to the encyclopedia of DNA elements (ENCODE). PLoS Biol. 9, e1001046.

Page 110: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

90

Estève, P.-O., Chin, H.G., Smallwood, A., Feehery, G.R., Gangisetty, O., Karpf, A.R., Carey, M.F., and Pradhan, S. (2006). Direct interaction between DNMT1 and G9a coordinates DNA and histone methylation during replication. Genes Dev. 20, 3089–3103.

Fazzio, T.G., and Tsukiyama, T. (2003). Chromatin remodeling in vivo: evidence for a nucleosome sliding mechanism. Mol. Cell 12, 1333–1340.

Felsenfeld, G., and Groudine, M. (2003). Controlling the double helix. Nature 421, 448–453.

Felsenfeld, G., Clark, D., and Studitsky, V. (2000). Transcription through nucleosomes. Biophys. Chem. 86, 231–237.

Ficz, G., Heintzmann, R., and Arndt-Jovin, D.J. (2005). Polycomb group protein complexes exchange rapidly in living Drosophila. Development 132, 3963–3976.

Foltz, D.R., Jansen, L.E.T., Black, B.E., Bailey, A.O., Yates, J.R., 3rd, and Cleveland, D.W. (2006). The human CENP-A centromeric nucleosome-associated complex. Nat. Cell Biol. 8, 458–469.

Fong, A.P., Yao, Z., Zhong, J.W., Cao, Y., Ruzzo, W.L., Gentleman, R.C., and Tapscott, S.J. (2012). Genetic and epigenetic determinants of neurogenesis and myogenesis. Dev. Cell 22, 721–735.

Fonseca, J.P., Steffen, P.A., Müller, S., Lu, J., Sawicka, A., Seiser, C., and Ringrose, L. (2012). In vivo Polycomb kinetics and mitotic chromatin binding distinguish stem cells from differentiated cells. Genes Dev. 26, 857–871.

Francis, N.J., Follmer, N.E., Simon, M.D., Aghia, G., and Butler, J.D. (2009). Polycomb Proteins Remain Bound to Chromatin and DNA during DNA Replication In Vitro. Cell 137, 110–122.

Gaszner, M., and Felsenfeld, G. (2006). Insulators: exploiting transcriptional and epigenetic mechanisms. Nat. Rev. Genet. 7, 703–713.

Goetze, S., Mateos-Langerak, J., Gierman, H.J., de Leeuw, W., Giromus, O., Indemans, M.H.G., Koster, J., Ondrej, V., Versteeg, R., and van Driel, R. (2007). The three-dimensional structure of human interphase chromosomes is related to the transcriptome map. Mol. Cell. Biol. 27, 4475–4487.

Page 111: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

91

Goldberg, A.D., Banaszynski, L.A., Noh, K.-M., Lewis, P.W., Elsaesser, S.J., Stadler, S., Dewell, S., Law, M., Guo, X., Li, X., et al. (2010). Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 140, 678–691.

Gómez-Rodríguez, M., and Jansen, L.E. (2013). Basic properties of epigenetic systems: lessons from the centromere. Curr. Opin. Genet. Dev. 23, 219–227.

Goutte-Gattat, D., Shuaib, M., Ouararhni, K., Gautier, T., Skoufias, D.A., Hamiche, A., and Dimitrov, S. (2013). Phosphorylation of the CENP-A amino-terminus in mitotic centromeric chromatin is required for kinetochore function. Proc. Natl. Acad. Sci. U. S. A. 110, 8579–8584.

De Graaf, C.A., and van Steensel, B. (2013). Chromatin organization: form to function. Curr. Opin. Genet. Dev. 23, 185–190.

Groth, A., Corpet, A., Cook, A.J.L., Roche, D., Bartek, J., Lukas, J., and Almouzni, G. (2007). Regulation of Replication Fork Progression Through Histone Supply and Demand. Science 318, 1928–1931.

Haig, D. (2004). The (Dual) Origin of Epigenetics. Cold Spring Harb. Symp. Quant. Biol. 69, 67–70.

Hansen, K.H., Bracken, A.P., Pasini, D., Dietrich, N., Gehani, S.S., Monrad, A., Rappsilber, J., Lerdrup, M., and Helin, K. (2008). A model for transmission of the H3K27me3 epigenetic mark. Nat. Cell Biol. 10, 1291–1300.

Hardy, S., and Robert, F. (2010). Random deposition of histone variants: A cellular mistake or a novel regulatory mechanism? Epigenetics Off. J. DNA Methylation Soc. 5, 368–372.

Hathaway, N.A., Bell, O., Hodges, C., Miller, E.L., Neel, D.S., and Crabtree, G.R. (2012). Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460.

Heard, E., and Disteche, C.M. (2006). Dosage compensation in mammals: fine-tuning the expression of the X chromosome. Genes Dev. 20, 1848–1867.

Hemmerich, P., Weidtkamp-Peters, S., Hoischen, C., Schmiedeberg, L., Erliandri, I., and Diekmann, S. (2008). Dynamics of inner kinetochore assembly and maintenance in living cells. J. Cell Biol. 180, 1101–1114.

Page 112: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

92

Henikoff, S., Ahmad, K., and Malik, H.S. (2001). The Centromere Paradox: Stable Inheritance with Rapidly Evolving DNA. Science 293, 1098–1102.

Hess, S.T., Girirajan, T.P.K., and Mason, M.D. (2006). Ultra-high resolution imaging by fluorescence photoactivation localization microscopy. Biophys. J. 91, 4258–4272.

Heun, P., Erhardt, S., Blower, M.D., Weiss, S., Skora, A.D., and Karpen, G.H. (2006). Mislocalization of the Drosophila Centromere-Specific Histone CID Promotes Formation of Functional Ectopic Kinetochores. Dev. Cell 10, 303–315.

Hodges, C., and Crabtree, G.R. (2012). Dynamics of inherently bounded histone modification domains. Proc. Natl. Acad. Sci. 109, 13296–13301.

Hübner, M.R., Eckersley-Maslin, M.A., and Spector, D.L. (2013). Chromatin organization and transcriptional regulation. Curr. Opin. Genet. Dev. 23, 89–95.

Ishikawa-Ankerhold, H.C., Ankerhold, R., and Drummen, G.P.C. (2012). Advanced fluorescence microscopy techniques--FRAP, FLIP, FLAP, FRET and FLIM. Mol. Basel Switz. 17, 4047–4132.

Ishimi, Y., Sugasawa, K., Hanaoka, F., and Kikuchi, A. (1991). Replication of the simian virus 40 chromosome with purified proteins. J. Biol. Chem. 266, 16141–16148.

Jackson, V. (1990). In vivo studies on the dynamics of histone-DNA interaction: evidence for nucleosome dissolution during replication and transcription and a low level of dissolution independent of both. Biochemistry (Mosc.) 29, 719–731.

Jackson, V., and Chalkley, R. (1974). Separation of newly synthesized nucleohistone by equilibrium centrifugation in cesium chloride. Biochemistry (Mosc.) 13, 3952–3956.

Jackson, V., Granner, D.K., and Chalkley, R. (1975). Deposition of histones onto replicating chromosomes. Proc. Natl. Acad. Sci. U. S. A. 72, 4440–4444.

Jamai, A., Puglisi, A., and Strubin, M. (2009). Histone chaperone spt16 promotes redeposition of the original h3-h4 histones evicted by elongating RNA polymerase. Mol. Cell 35, 377–383.

Page 113: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

93

Jansen, L.E.T., Black, B.E., Foltz, D.R., and Cleveland, D.W. (2007). Propagation of centromeric chromatin requires exit from mitosis. J. Cell Biol. 176, 795–805.

Jb, A., K, M.-M., M, Z., M, G., and Ba, H. (2009). Ribosomal protein mRNAs are primary targets of regulation in RNase-L-induced senescence., Ribosomal protein mRNAs are primary targets of regulation in RNase-L-induced senescence. RNA Biol. RNA Biol. 6, 6, 305, 305–315.

Jin, C., Zang, C., Wei, G., Cui, K., Peng, W., Zhao, K., and Felsenfeld, G. (2009). H3.3/H2A.Z double variant-containing nucleosomes mark “nucleosome-free regions” of active promoters and other regulatory regions. Nat. Genet. 41, 941–945.

Jin, J., Bai, L., Johnson, D.S., Fulbright, R.M., Kireeva, M.L., Kashlev, M., and Wang, M.D. (2010). Synergistic action of RNA polymerases in overcoming the nucleosomal barrier. Nat. Struct. Mol. Biol. 17, 745–752.

Johnson, A., Meyer, B.J., and Ptashne, M. (1978). Mechanism of action of the cro protein of bacteriophage lambda. Proc. Natl. Acad. Sci. U. S. A. 75, 1783–1787.

Johnson, A.D., Poteete, A.R., Lauer, G., Sauer, R.T., Ackers, G.K., and Ptashne, M. (1981). λ Repressor and cro—components of an efficient molecular switch. Nature 294, 217–223.

Kalocsay, M., Hiller, N.J., and Jentsch, S. (2009). Chromosome-wide Rad51 spreading and SUMO-H2A.Z-dependent chromosome fixation in response to a persistent DNA double-strand break. Mol. Cell 33, 335–343.

Katan-Khaykovich, Y., and Struhl, K. (2011). Splitting of H3-H4 tetramers at transcriptionally active genes undergoing dynamic histone exchange. Proc. Natl. Acad. Sci. U. S. A. 108, 1296–1301.

Keppler, A., Kindermann, M., Gendreizig, S., Pick, H., Vogel, H., and Johnsson, K. (2004). Labeling of fusion proteins of O6-alkylguanine-DNA alkyltransferase with small molecules in vivo and in vitro. Methods San Diego Calif 32, 437–444.

Kimura, H., and Cook, P.R. (2001). Kinetics of Core Histones in Living Human Cells Little Exchange of H3 and H4 and Some Rapid Exchange of H2b. J. Cell Biol. 153, 1341–1354.

Page 114: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

94

Kouskouti, A., and Talianidis, I. (2005). Histone modifications defining active genes persist after transcriptional and mitotic inactivation. EMBO J. 24, 347–357.

Kouzarides, T. (2007). Chromatin Modifications and Their Function. Cell 128, 693–705.

Kuo, A.J., Song, J., Cheung, P., Ishibe-Murakami, S., Yamazoe, S., Chen, J.K., Patel, D.J., and Gozani, O. (2012). The BAH domain of ORC1 links H4K20me2 to DNA replication licensing and Meier-Gorlin syndrome. Nature 484, 115–119.

Kurumizaka, H., Horikoshi, N., Tachiwana, H., and Kagawa, W. (2013). Current progress on structural studies of nucleosomes containing histone H3 variants. Curr. Opin. Struct. Biol. 23, 109–115.

Lachner, M., O’Carroll, D., Rea, S., Mechtler, K., and Jenuwein, T. (2001). Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410, 116–120.

Lam, L.T., Pickeral, O.K., Peng, A.C., Rosenwald, A., Hurt, E.M., Giltnane, J.M., Averett, L.M., Zhao, H., Davis, R.E., Sathyamoorthy, M., et al. (2001). Genomic-scale measurement of mRNA turnover and the mechanisms of action of the anti-cancer drug flavopiridol. Genome Biol. 2, RESEARCH0041.

Lassar, A.B., Buskin, J.N., Lockshon, D., Davis, R.L., Apone, S., Hauschka, S.D., and Weintraub, H. (1989). MyoD is a sequence-specific DNA binding protein requiring a region of myc homology to bind to the muscle creatine kinase enhancer. Cell 58, 823–831.

Lavoie, G., and St-Pierre, Y. (2011). Phosphorylation of human DNMT1: Implication of cyclin-dependent kinases. Biochem. Biophys. Res. Commun. 409, 187–192.

Law, J.A., and Jacobsen, S.E. (2010). Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat. Rev. Genet. 11, 204–220.

Lee, J.T. (2012). Epigenetic Regulation by Long Noncoding RNAs. Science 338, 1435–1439.

Lee, T.-F., Zhai, J., and Meyers, B.C. (2010). Conservation and divergence in eukaryotic DNA methylation. Proc. Natl. Acad. Sci. U. S. A. 107, 9027–9028.

Page 115: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

95

Lewis, P.W., Elsaesser, S.J., Noh, K.-M., Stadler, S.C., and Allis, C.D. (2010). 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. 107, 14075–14080.

Livak, K.J., and Schmittgen, T.D. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods San Diego Calif 25, 402–408.

Lo, S.M., Follmer, N.E., Lengsfeld, B.M., Madamba, E.V., Seong, S., Grau, D.J., and Francis, N.J. (2012). A Bridging Model for Persistence of a Polycomb Group Protein Complex through DNA Replication In Vitro. Mol. Cell 46, 784–796.

Loyola, A., Tagami, H., Bonaldi, T., Roche, D., Quivy, J.P., Imhof, A., Nakatani, Y., Dent, S.Y.R., and Almouzni, G. (2009). The HP1alpha-CAF1-SetDB1-containing complex provides H3K9me1 for Suv39-mediated K9me3 in pericentric heterochromatin. EMBO Rep. 10, 769–775.

Lucchini, R., Wellinger, R.E., and Sogo, J.M. (2001). Nucleosome positioning at the replication fork. EMBO J. 20, 7294–7302.

Luger, K., Mäder, A.W., Richmond, R.K., Sargent, D.F., and Richmond, T.J. (1997). Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389, 251–260.

Luger, K., Dechassa, M.L., and Tremethick, D.J. (2012). New insights into nucleosome and chromatin structure: an ordered state or a disordered affair? Nat. Rev. Mol. Cell Biol. 13, 436–447.

Mahoney, D.J., Cheung, H.H., Mrad, R.L., Plenchette, S., Simard, C., Enwere, E., Arora, V., Mak, T.W., Lacasse, E.C., Waring, J., et al. (2008). Both cIAP1 and cIAP2 regulate TNFalpha-mediated NF-kappaB activation. Proc. Natl. Acad. Sci. U. S. A. 105, 11778–11783.

Margueron, R., Justin, N., Ohno, K., Sharpe, M.L., Son, J., Drury, W.J., 3rd, Voigt, P., Martin, S.R., Taylor, W.R., De Marco, V., et al. (2009). Role of the polycomb protein EED in the propagation of repressive histone marks. Nature 461, 762–767.

Mendiburo, M.J., Padeken, J., Fülöp, S., Schepers, A., and Heun, P. (2011). Drosophila CENH3 Is Sufficient for Centromere Formation. Science 334, 686–690.

Page 116: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

96

Meneghini, M.D., Wu, M., and Madhani, H.D. (2003). Conserved histone variant H2A.Z protects euchromatin from the ectopic spread of silent heterochromatin. Cell 112, 725–736.

Mitchell, J.A., and Fraser, P. (2008). Transcription factories are nuclear subcompartments that remain in the absence of transcription. Genes Dev. 22, 20–25.

Molnar, A., Melnyk, C.W., Bassett, A., Hardcastle, T.J., Dunn, R., and Baulcombe, D.C. (2010). Small Silencing RNAs in Plants Are Mobile and Direct Epigenetic Modification in Recipient Cells. Science 328, 872–875.

Nakayama, T., Nishioka, K., Dong, Y.-X., Shimojima, T., and Hirose, S. (2007). Drosophila GAGA factor directs histone H3.3 replacement that prevents the heterochromatin spreading. Genes Dev. 21, 552–561.

Nekrasov, M., Amrichova, J., Parker, B.J., Soboleva, T.A., Jack, C., Williams, R., Huttley, G.A., and Tremethick, D.J. (2012). Histone H2A.Z inheritance during the cell cycle and its impact on promoter organization and dynamics. Nat Struct Mol Biol 19, 1076–1083.

Ng, R.K., and Gurdon, J.B. (2008). Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription. Nat. Cell Biol. 10, 102–109.

Nobrega, M.A., Ovcharenko, I., Afzal, V., and Rubin, E.M. (2003). Scanning human gene deserts for long-range enhancers. Science 302, 413.

Ong, S.-E., Blagoev, B., Kratchmarova, I., Kristensen, D.B., Steen, H., Pandey, A., and Mann, M. (2002). Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics MCP 1, 376–386.

Orphanides, G., and Reinberg, D. (2000). RNA polymerase II elongation through chromatin. Nature 407, 471–475.

Panchenko, T., Sorensen, T.C., Woodcock, C.L., Kan, Z.-Y., Wood, S., Resch, M.G., Luger, K., Englander, S.W., Hansen, J.C., and Black, B.E. (2011). Replacement of histone H3 with CENP-A directs global nucleosome array condensation and loosening of nucleosome superhelical termini. Proc. Natl. Acad. Sci. U. S. A. 108, 16588–16593.

Panse, S.L., Masson, C., Héliot, L., Chassery, J.M., Junéra, H.R., and Hernandez-Verdun, D. (1999). 3-D organization of ribosomal

Page 117: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

97

transcription units after DRB inhibition of RNA polymerase II transcription. J. Cell Sci. 112 ( Pt 13), 2145–2154.

Pellett, P.A., Sun, X., Gould, T.J., Rothman, J.E., Xu, M.-Q., Corrêa, I.R., Jr, and Bewersdorf, J. (2011). Two-color STED microscopy in living cells. Biomed. Opt. Express 2, 2364–2371.

Peric-Hupkes, D., Meuleman, W., Pagie, L., Bruggeman, S.W.M., Solovei, I., Brugman, W., Gräf, S., Flicek, P., Kerkhoven, R.M., van Lohuizen, M., et al. (2010). Molecular maps of the reorganization of genome-nuclear lamina interactions during differentiation. Mol. Cell 38, 603–613.

Petruk, S., Sedkov, Y., Johnston, D.M., Hodgson, J.W., Black, K.L., Kovermann, S.K., Beck, S., Canaani, E., Brock, H.W., and Mazo, A. (2012). TrxG and PcG Proteins but Not Methylated Histones Remain Associated with DNA through Replication. Cell 150, 922–933.

Price, R., and Penman, S. (1972). A distinct RNA polymerase activity, synthesizing 5.5 s, 5 s and 4 s RNA in nuclei from adenovirus 2-infected HeLa cells. J. Mol. Biol. 70, 435–450.

Ptashne, M., Backman, K., Humayun, M.Z., Jeffrey, A., Maurer, R., Meyer, B., and Sauer, R.T. (1976). Autoregulation and function of a repressor in bacteriophage lambda. Science 194, 156–161.

Radman-Livaja, M., Verzijlbergen, K.F., Weiner, A., van Welsem, T., Friedman, N., Rando, O.J., and van Leeuwen, F. (2011). Patterns and Mechanisms of Ancestral Histone Protein Inheritance in Budding Yeast. PLoS Biol 9, e1001075.

Ray-Gallet, D., Quivy, J.-P., Scamps, C., Martini, E.M.-D., Lipinski, M., and Almouzni, G. (2002). HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol. Cell 9, 1091–1100.

Ray-Gallet, D., Woolfe, A., Vassias, I., Pellentz, C., Lacoste, N., Puri, A., Schultz, D.C., Pchelintsev, N.A., Adams, P.D., Jansen, L.E.T., et al. (2011). Dynamics of histone H3 deposition in vivo reveal a nucleosome gap-filling mechanism for H3.3 to maintain chromatin integrity. Mol. Cell 44, 928–941.

Rizzo, J.M., Bard, J.E., and Buck, M.J. (2012). Standardized collection of MNase-seq experiments enables unbiased dataset comparisons. BMC Mol. Biol. 13, 15.

Page 118: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

98

Schermelleh, L., Haemmer, A., Spada, F., Rösing, N., Meilinger, D., Rothbauer, U., Cardoso, M.C., and Leonhardt, H. (2007). Dynamics of Dnmt1 interaction with the replication machinery and its role in postreplicative maintenance of DNA methylation. Nucleic Acids Res. 35, 4301–4312.

Sexton, T., Umlauf, D., Kurukuti, S., and Fraser, P. (2007). The role of transcription factories in large-scale structure and dynamics of interphase chromatin. Semin. Cell Dev. Biol. 18, 691–697.

Shibahara, K., and Stillman, B. (1999). Replication-dependent marking of DNA by PCNA facilitates CAF-1-coupled inheritance of chromatin. Cell 96, 575–585.

Simon, J.A., and Kingston, R.E. (2009). Mechanisms of polycomb gene silencing: knowns and unknowns. Nat. Rev. Mol. Cell Biol. 10, 697–708.

Simon, J.A., and Kingston, R.E. (2013). Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol. Cell 49, 808–824.

Smith, S., and Stillman, B. (1989). Purification and characterization of CAF-I, a human cell factor required for chromatin assembly during DNA replication in vitro. Cell 58, 15–25.

Spada, F., Haemmer, A., Kuch, D., Rothbauer, U., Schermelleh, L., Kremmer, E., Carell, T., Längst, G., and Leonhardt, H. (2007). DNMT1 but not its interaction with the replication machinery is required for maintenance of DNA methylation in human cells. J. Cell Biol. 176, 565–571.

Studitsky, V.M., Clark, D.J., and Felsenfeld, G. (1994). A histone octamer can step around a transcribing polymerase without leaving the template. Cell 76, 371–382.

Studitsky, V.M., Kassavetis, G.A., Geiduschek, E.P., and Felsenfeld, G. (1997). Mechanism of transcription through the nucleosome by eukaryotic RNA polymerase. Science 278, 1960–1963.

Sun, X., Zhang, A., Baker, B., Sun, L., Howard, A., Buswell, J., Maurel, D., Masharina, A., Johnsson, K., Noren, C.J., et al. (2011). Development of SNAP-tag fluorogenic probes for wash-free fluorescence imaging. Chembiochem Eur. J. Chem. Biol. 12, 2217–2226.

Page 119: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

99

Suto, R.K., Clarkson, M.J., Tremethick, D.J., and Luger, K. (2000). Crystal structure of a nucleosome core particle containing the variant histone H2A.Z. Nat. Struct. Biol. 7, 1121–1124.

Tachiwana, H., Kagawa, W., Shiga, T., Osakabe, A., Miya, Y., Saito, K., Hayashi-Takanaka, Y., Oda, T., Sato, M., Park, S.-Y., et al. (2011). Crystal structure of the human centromeric nucleosome containing CENP-A. Nature 476, 232–235.

Talbert, P.B., and Henikoff, S. (2010). Histone variants — ancient wrap artists of the epigenome. Nat. Rev. Mol. Cell Biol. 11, 264–275.

Thayer, M.J., Tapscott, S.J., Davis, R.L., Wright, W.E., Lassar, A.B., and Weintraub, H. (1989). Positive autoregulation of the myogenic determination gene MyoD1. Cell 58, 241–248.

Trojer, P., and Reinberg, D. (2006). Histone Lysine Demethylases and Their Impact on Epigenetics. Cell 125, 213–217.

Tyler-Smith, C., Gimelli, G., Giglio, S., Floridia, G., Pandya, A., Terzoli, G., Warburton, P.E., Earnshaw, W.C., and Zuffardi, O. (1999). Transmission of a fully functional human neocentromere through three generations. Am. J. Hum. Genet. 64, 1440–1444.

Vafa, O., and Sullivan, K.F. (1997). Chromatin containing CENP-A and α-satellite DNA is a major component of the inner kinetochore plate. Curr. Biol. 7, 897–900.

Vassilev, L.T., Tovar, C., Chen, S., Knezevic, D., Zhao, X., Sun, H., Heimbrook, D.C., and Chen, L. (2006). Selective small-molecule inhibitor reveals critical mitotic functions of human CDK1. Proc. Natl. Acad. Sci. 103, 10660–10665.

Verdun, R.E., and Karlseder, J. (2007). Replication and protection of telomeres. Nature 447, 924–931.

Verzijlbergen, K.F., Menendez-Benito, V., van Welsem, T., van Deventer, S.J., Lindstrom, D.L., Ovaa, H., Neefjes, J., Gottschling, D.E., and van Leeuwen, F. (2010). Recombination-induced tag exchange to track old and new proteins. Proc. Natl. Acad. Sci. U. S. A. 107, 64–68.

Waddington, C.H. (2011). The Epigenotype. Int. J. Epidemiol.

Warburton, P.E., Greig, G.M., Haaf, T., and Willard, H.F. (1991). PCR amplification of chromosome-specific alpha satellite DNA: definition of

Page 120: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 2 - TimeChIP

100

centromeric STS markers and polymorphic analysis. Genomics 11, 324–333.

Warburton, P.E., Cooke, C.A., Bourassa, S., Vafa, O., Sullivan, B.A., Stetten, G., Gimelli, G., Warburton, D., Tyler-Smith, C., Sullivan, K.F., et al. (1997). Immunolocalization of CENP-A suggests a distinct nucleosome structure at the inner kinetochore plate of active centromeres. Curr. Biol. CB 7, 901–904.

Waye, J.S., and Willard, H.F. (1986). Structure, organization, and sequence of alpha satellite DNA from human chromosome 17: evidence for evolution by unequal crossing-over and an ancestral pentamer repeat shared with the human X chromosome. Mol. Cell. Biol. 6, 3156–3165.

Weber, C.M., Henikoff, J.G., and Henikoff, S. (2010). H2A.Z nucleosomes enriched over active genes are homotypic. Nat Struct Mol Biol 17, 1500–1507.

Xu, C., Bian, C., Yang, W., Galka, M., Ouyang, H., Chen, C., Qiu, W., Liu, H., Jones, A.E., MacKenzie, F., et al. (2010a). Binding of different histone marks differentially regulates the activity and specificity of polycomb repressive complex 2 (PRC2). Proc. Natl. Acad. Sci. U. S. A. 107, 19266–19271.

Xu, M., Long, C., Chen, X., Huang, C., Chen, S., and Zhu, B. (2010b). Partitioning of Histone H3-H4 Tetramers During DNA Replication–Dependent Chromatin Assembly. Science 328, 94–98.

Yankulov, K., Yamashita, K., Roy, R., Egly, J.-M., and Bentley, D.L. (1995). The Transcriptional Elongation Inhibitor 5,6-Dichloro-1-β-D-ribofuranosylbenzimidazole Inhibits Transcription Factor IIH-associated Protein Kinase. J. Biol. Chem. 270, 23922–23925.

Zee, B.M., Levin, R.S., Xu, B., LeRoy, G., Wingreen, N.S., and Garcia, B.A. (2010). In vivo residue-specific histone methylation dynamics. J. Biol. Chem. 285, 3341–3350.

Zentner, G.E., and Henikoff, S. (2013). Regulation of nucleosome dynamics by histone modifications. Nat. Struct. Mol. Biol. 20, 259–266.

Page 121: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

101

Page 122: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

102

Page 123: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

103

Chapter 3 – H3.1 and H3.3 Turnover

Page 124: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

104

Page 125: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

105

AUTHOR CONTRIBUTION

All the experiments were planned by the author and the

supervisor Lars E.T. Jansen. All experiments were executed by the

author, except those depicted in figure 3.6 which were performed in

collaboration with Daniel Sobral from the Bioinformatics Unit at IGC.

SUMMARY

Epigenetic forms of inheritance are implicated in the

transmission of cellular states independent on the DNA sequence.

Chromatin is a pivotal component of carriers of epigenetic information.

The steady-state maintenance of chromatin components dependents

on the rates of nucleosome assembly and disassembly, as well as the

rate of reestablishment of histone modifications which have been

implicated in epigenetic memory. There is evidence for rapid histone

turnover at specific loci, however the proportion of histone stably

retained at specific loci is unknown. By using a TimeChIP strategy,

based on the self-labeling SNAP-tag for the isolation of pulse labeled

histones of different ages, we show that a significant pool of H3.1 and

H3.3 is retained at genes and non-genes for the duration of the cell

cycle. Turnover of H3.1 and H3.3 does not increase when transcription

is activated, whereas inhibition of RNA polymerase leads to retention

of H3.1 and H3.3. Our results support a model where nucleosomes

contribute to inheritance of non genetic information.

Page 126: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

106

INTRODUCTION

Inheritance of gene expression profiles and associated

phenotypes that cannot be explained by changes in the DNA

sequence is known as epigenetic inheritance. The maintenance of

epigenetic information depends on the ability to persist through cell

divisions (Waddington, 2011). Many factors may contribute to the

epigenetic state. These include positive feedback loops that maintain a

network of transcription factors that in this way perpetuate the

transcriptional program. However, maintenance also requires the

existence of molecules that propagate specific chromatin structures to

either activate or repress gene expression, especially when identical

sequences within the same cell are controlled differently in cis. This

implies that chromatin may be a pivotal carrier of epigenetic

information. There is a plethora of chromatin regulators involved in

epigenetic phenomena, including DNA or histone modifications, DNA

or histone binding proteins and histone variants (Gómez-Rodríguez

and Jansen, 2013). The most extensively studied among these are

histone modifications and the enzymes that regulate them, although

whether modifications are heritable is a controversial issue and

possible mechanisms are unclear. To consider histone modifications

as epigenetic carriers it is necessary to understand the dynamic

equilibrium between histone turnover and the dynamics of the

modifications of those histones.

The basic unit of chromatin is the nucleosome. The

nucleosome is formed by an octamer of four core histones (H2A, H2B,

H3 and H4) wrapped by 147bp of DNA (Luger et al., 1997).

Nucleosome assembly occurs in two steps: deposition of (H3-H4)2

tetramer followed by the addition of two H2A-H2B dimers (Jackson

1990). With the exception of H4 all histones have variants that are not

linked to DNA replication. The specific enrichment of histone variants

Page 127: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

107

at different genomic loci indicate that they serves different roles in

preserving epigenetic identity. In mammals, the major H3 variants are

H3.3 and centromere protein A (CENP-A) (Talbert and Henikoff 2010).

H3.3 differs from canonical H3.1 by only four amino acids and its

incorporation into chromatin occurs throughout the cell cycle and is

mainly localized at sites of active transcription as well as enhancers

and telomeres (Goldberg et al., 2010; Ray-Gallet et al., 2011). CENP-

A localization is restricted to the centromere, its deposition occurs in

G1 phase after which the protein is stably maintained (Bodor et al.,

2013; Jansen et al., 2007; Vafa and Sullivan, 1997).

The inheritance of nucleosomes across cell division cycles

cannot be understood intuitively as is in the case of DNA. The

semiconservative nature of DNA replication and segregation is given

by the complementary base-pairing of its sequences. Propagation of a

chromatin state requires an orchestration between nucleosome

assembly, positioning, disassembly and stability (Luger et al., 2012;

Simon and Kingston, 2013). Moreover, in the case of nucleosomes,

there appears to be little opportunity for direct templating new

molecules onto older ones indicating that maintenance of a “state”

requires self-templating in an indirect manner (Gómez-Rodríguez and

Jansen, 2013).

Classical radioactive pulse-chase analysis on bulk chromatin

and FRAP experiments on GFP-tagged histones have shown that, in

general, H3/H4 and H2A/H2B are stable molecules. However,

H2A/H2B dimers exchange into chromatin faster and part of the

H2A/H2B dynamics is suppressed when RNA polymerase II is

inhibited (Jackson, 1990; Kimura and Cook, 2001). This indicates that

transcription is a driver of H2A/H2B exchange. Indeed, genome wide

analysis of pulse labeled histones indicates that nucleosomes undergo

fast turnover at active genes, epigenetic regulatory elements and

Page 128: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

108

origins of replication in Drosophila cells (Deal et al., 2010). However, in

this study a method is used (CATCH-IT) in which histones are labeled

during synthesis using a methionine analogue that is subsequently

coupled to biotin. Therefore, the analysis is biased towards the

dynamics of newly incorporated nucleosomes which may not represent

the overall histone pool. By using an inducible recombination tagging

system in budding yeast, Radman-Livaja et al., followed the fate of

parental histones and compared their distribution with nascent

histones genome wide through multiple cell divisions. They found that

ancestral nucleosomes distribute to adjacent locations after replication

(Radman-Livaja et al., 2011). Nevertheless, there is no evidence of

retention of ancestral histones at specific loci in mammals.

To understand the mechanisms of chromatin-based epigenetic

inheritance it is necessary to know at what rate histones turnover and

at which loci in living cells. We know that CENP-A exhibits a strikingly

slow turnover using SNAP-based pulse labeling experiments (Bodor et

al., 2013; Jansen et al., 2007). We have developed a methodology

based on the SNAP system (which we call TimeChIP) to determine the

turnover of histone pools of different ages at specific genomic loci. We

find that both H3.1 and H3.3 are turned over faster at active genes

versus non-gene loci, with H3.3 displaying faster dynamics than H3.1,

particularly at active genes. Moreover, we find that H3.1 and H3.3

turnover is slowed upon inhibition of transcription. For both histones a

variable but significant pool is retained for the duration of the cell cycle.

These findings are therefore consistent with the idea of nucleosomes

as candidate carriers of epigenetic information.

Page 129: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

109

MATERIALS AND METHODS

SNAP labeling and drug treatments

Pulse labeling of intact cells was performed with 10µM CP-

Biotin (provided by Ivan Correa, New England Biolabs), in growth

medium for 1 hour at 37ºC. After labeling, cells were washed twice with

medium and reincubated at 37ºC to allow excess SNAP substrate to

be released from cells. After an additional 30 minutes, cells were

washed again once with medium and reincubated with growth medium

and further treated for analysis as indicated. TNF-α was used at

50ng/mL (R&D Systems), Thymidine at 2mM (Sigma), DRB (5,6-

Dichloro-1-β-D-ribofuranosylbenzimidazole) at 100µM (Sigma) and α–

Amanitin at 10µg/mL (Sigma).

Soluble nucleosome preparation, SNAP nucleosomes

purification and quantification of bound nucleosomes and qPCR

analysis were performed as described in chapter 2.

Page 130: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

110

*(Warburton et al., 1991), **(Bergmann et al., 2011).

Immunofluorescence

HeLa cells were grown on glass coverslips coated with poly-L-

Lysine (Sigma) and fixed with 4% formaldehyde (Thermo Scientific) for

10 minutes. Cells were extracted after fixation and processed for

immunofluorescence using standard procedures. Cells were stained

with anti-NF-ƙB (1:25; sc8414, Santa Cruz Biotechnology). For BrdU

staining, HeLa cells were fixed in Methanol/Acetone (1:1; Sigma) for 3

minutes, acid treated with 2M HCl during 30 minutes at RT followed by

3 x 10 minute washes with 100 mM Borax (Sigma). HeLa cells were

extracted with 0.1% Triton X-100 and stained with anti-BrdU (1:100;

MoBU-1, Santa Cruz Biotechnology). Secondary antibodies used were

FITC-conjugated anti-mouse (Jackson Immunoresearch Laboratories)

and Cy3-conjugated anti mouse (Sigma). Cells were stained with DAPI

(4’,6-diamidino-2-phenylindole; Sigma) before mounting in Mowiol

Description Target Sequence (5’ -3’)

ACTBe3F exon 3 GCCCGTGCTCAGGGCTTCTTACTBe3R exon 3 TGGGCCTCGTCGCCCACATAGAPDHF exon 1 AATTGAGCCCGCAGCCTCCCGAPDHR exon 1 CGTCTTCACCTGGCGACGCAAATBPF exon 1 TTATCAACGCGCGCCAGGGGTBPR exon 1 TTAAACAGCCGGCGGCCCAGRPL13AF promoter-exon join GCCGCCCCTGTTTCAAGGGATARPL13AR promoter-exon join CTGCACCTCCGCCATCTTCGG13/21-3A * α-satellite I Chr 13 & 21 CTTCTGTCTAGATTTTAGA13/21-1B α-satellite I Chr 13 & 21 CATAGAGATGAACATGGSat2_F ** pericentromeric satellite 2 TCGCATAGAATCGAATGGAA

Sat2_R pericentromeric satellite 2 GCATTCGAGTCCGTGGA

desertpDACHF2 0.8Mb DACH1 gene TTCAGGTTCACTAAATGCAGGCCG

desertpDACHR2 0.8Mb DACH1 gene AGCATGAACAAGAAGGGCCACTGT

pNFkBF2 promoter TCAGTGGGAATTTCCAGCCAG

pNFkBR2 promoter GGCGAAACCTCCTCTTCCTG

NFkBF exon 2 TTTGCAGAGAGGATTTCGTTTCCGTNFkBR exon 2 AGAGGCACCAGGTAGTCCACCA

Table 1. List of primers used for TimeChIP

Page 131: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

111

(Calbiochem). Cells treated with TNF-α were stained with Rhodamine

Phalloidin (Invitrogen) before DAPI staining.

Microscopy

Digital images were captured using a DeltaVision Core System

(Applied Precision) that controls an inverted microscope (Olympus, IX-

71), coupled to a Cascade 2 EMCCD camera (Photometrics). Images

(512x512) were collected at 1x binning using a 100x oil objective

(NA1.40, UPIanSApo) with 0.2µm axial sections spanning the entire

nucleus. Images were subsequently deconvolved and maximum

signals were projected as 2D images using softWoRx (Applied

Precision). Fluorescence quantification was performed on

nondeconvolved images. The BrdU fluorescence signal intensity was

quantified using ImageJ (NIH). The sum of the mean and two times the

standard deviation of cells for which BrdU treatment was omitted was

used as background cutoff. BrdU treated cells that displayed a signal

higher than this cutoff were considered positive.

Flow cytometry

HeLa cells (106) were harvested and fixed 1 hour at 4ºC with

70% ethanol. Cells were washed twice in PBS containing 3% BSA

(Sigma) and incubated for 30 minutes at room temperature with

5µg/mL propidium iodide (Sigma) and 200µg/mL of RNaseA in PBS

containing 3% BSA. Subsequently flow cytometry analysis was

performed on FACScan (Becton Dickinson) using CellQuest software

and for quantification FlowJo software.

Next-generation sequencing

10ng of purified TimeChIPed DNA quantified by PicoGreen

assay were submitted. Sequencing libraries were generated and

barcoded for multiplexing according to Illumina recommendations.

Resulting libraries were submitted for Illumina sequencing on a 4x

Page 132: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

112

Illumina HiSeq at the Gene Core Facility at EMBL, Heidelberg

Germany.

RNA extraction and qRT-PCR

RNA was isolated from 106 HeLa cells with TRizol (Life

Sciences), according to manufacturer´s instructions. Prior to cDNA

synthesis, RNA was treated with RNase-Free DNase (Promega)

following the provided protocol. cDNA synthesis of 1µg of DNAse-

treated RNA was performed using SuperScript II Reverse

Transcriptase (Invitrogen). qRT-PCR analyses were performed on a

7900HT Fast Real-Time PCR System (Applied Biosystems), using the

iTaq Universal SYBR Green Supermix (BioRad), and 2-ΔΔCt method for

relative quantification (Livak and Schmittgen, 2001). Expression values

were normalized using threshold cycle (CT) values obtained for

RPLP13A and GAPDH genes.

Description Sequence (5’ -3’)

rcIAP2F TGCCAAGTGGTTTCCAAGGTGTGAGTrcIAP2R CTGTTCAAGAAGATGAGGrCCNA2F TCCTCGTGGACTGGTTAGTTGrCCNA2R ACAGCCAAATGCAGGGTCTCrGAPDHF GGACTCATGACCACAGTCCATGCCrGAPDHR GCGGCCATCACGCCACAGTTrH31LF CGAGAAATCGCCCAAGACTTCrH31LR TGTGTCCTCAAAGAGCCCTACCrMYCF CCACAGCAAACCTCCTCACAGrMYCR GCAGGATAGTCCTTCCGAGTGrNFkBF ATGTTTCATTTGGATCCTTrNFkBR GAAACGAAATCCTCTCTGTTTAGrRPLP0F TGGCTCCTCTGGCTTGTTTTrRPLP0R CCACATTGTCTGCTCCCACArRPL13AF CGTGCGAGGTATGCTGCCCCrRPL13AR GGCGGTGGGATGCCGTCAAA

Table 2. List of primers used for qRT-PCR

Page 133: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

113

RESULTS

Quantitative retention of ancestral H3.1 and H3.3

HeLa cells stably expressing H3.1-SNAP-3XHA or H3.3-SNAP-

3XHA were in vivo pulse labeled with CP-biotin and chased for 3, 6

and 12 hours. Chromatin was extracted and fragmented into

mononucleosomes by MNase treatment. H3.1-SNAP or H3.3-SNAP

biotinylated nucleosomes were immobilized on Streptavidin magnetic

beads. Co-purified DNA from recovered H3.1 or H3.3-SNAP

nucleosomes was quantified by spectrophotometry. The occupancy of

pulse and pulse-chase H3.1 and H3.3 nucleosomes at a particular

locus was quantified by qPCR. The loci analyzed were exon 1 of TATA

binding protein (TBP) a lowly transcribed gene as estimated from HeLa

RNA-seq data (3.2-3.5 reads per kilobase per million mapped reads

RPKM), exon 3 of the β-actin gene (ACTB), the promoter-exon 1

junction of the ribosomal protein L13a (RPL13A) and exon 1 of the

glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene which

transcripts levels correspond to 90-93, 110-220 and 600-900 RPKM,

respectively (ENCODE Project Consortium, 2011). To measure the

turnover rate of H3.1 and H3.3 outside genes, we selected the α-

satellite I repeat, pericentromeric satellite 2 repeats (Sat2) and a

conserved non-coding element that is not detectably transcribed (gene

desert) (Nobrega et al., 2003). Initially, cells expressing SNAP-tagged

canonical H3.1 and H3.3 were chased for 3, 6 and 12 hours after pulse

labeling. We observed that canonical H3.1 exhibits slow turnover at the

α-satellite I and pericentromeric satellite 2 where ~50% of ancestral

histone is retained after 12 hours chase. In comparison, H3.1

nucleosomes are turned over faster at the active genes ACTB,

GAPDH and RPL13A. Surprisingly, the extent of turnover is limited and

after 12 hours ~20-30% of nucleosomes are still retained at these

genes. Interestingly, the gene desert locus that is not transcribed,

Page 134: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

114

displays a similar kinetics to active genes (Figure 3.1). H3.3 turnover is

slightly faster than canonical H3.1 at all loci analyzed (Figure 3.1 A &

B). Statistical analysis indicates that H3.3 turnover is significantly

faster after 3 and 6 hours chase at genes and after 6 and 12 hours

chase at non-genes (Figure 3.3). Interestingly, nucleosome turnover

appears to be biphasic in nature at active genes, particularly for H3.3.

Only half of the parental H3.3-SNAP population is retained after 3

hours chase at all loci except Sat2 and the gene desert locus. After

this initial rapid turnover, the rate of loss of nucleosomes slows down

and significant retention is observed up to 12 hours chase at the four

genes. This suggests that nucleosome turnover is not homogenous

and that qualitatively different populations of nucleosomes exist at

active genes (Figure 3.1 A).

Page 135: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

115

Figure 3.1 TimeChIP analysis of H3.1-SNAP and H3.3-SNAP. A) Cells

expressing histones H3.1-SNAP and H3.3-SNAP were pulse-biotinylated and

chased for indicated times. Chromatin was isolated and fragment followed by

biotin-dependent pull down and qPCR analysis for quantification of the

following DNA loci: beta actin (ACTB), glyceraldehyde 3-phosphate

dehydrogenase (GAPDH), TATA binding protein (TBP) and ribosomal protein

L13a (RPL13A) genes at indicated time points. B) Analysis as in A but

processed for detection of α-satellite I repeat, pericentromeric satellite 2

repeats (Sat2) and a conserved non-coding element (gene desert) at

indicated time points. Fraction of biotin retained values are derived from fitting

of the %IP measurements of chased samples on a linear regression of a

TimeChIP standard curve that was included in each experiment (see Chapter

2). Data shown represents an average of six independent experiments, error

Page 136: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

116

bars indicate standard error of the mean (S.E.M). Statistically significant

difference in the comparison is indicated by asterisk (*) p<0.05, (**) p<0.01,

(***) p<0.001 (Kruskall-Wallis test).

Since significant levels of ancestral histones could be detected

after 12 hours we next tested whether canonical histone H3.1 and

H3.3 are retained at the time scale of the cell cycle of HeLa cells. To

this end cells were chased for 12, 24 and 48 hours following pulse

labeling. In general, we observed that number of cells was maintained

from 0h (5x107 cells) to 24h (6-7x107cells) and duplicated from 24 to

48 hours (1-1.2x108 cells), indicating that during the course of the

experiment cells underwent at least one cell cycle. We found that

~20% of ancestral H3.1 is retained after 24 hours chase at ACTB,

GAPDH, TBP, RPL13A genes and the gene desert locus. After 48

hours levels are further reduced to ~10% but are still detectable above

background levels (Figure 3.2). In contrast, canonical H3.1 shows a

slower turnover at the α-satellite I repeats and the pericentromeric

domain, but is eventually also reduced to 10% after 48 hours of chase

at Sat2 but not at α-satellite I repeats where retention is maintained at

~35% (Figure 3.2 B). Turnover rates of H3.3 at this timescale are

similar to H3.1 although H3.3 appears slightly faster at most loci with

the exception of TBP (Figure 3.3). Nevertheless, these differences

were found not be significant. We observed that 20% of H3.3 is

retained after 24 and 48 hours chase at α-satellite and pericentromeric

satellite 2 (Figure 3.2 B). In sum, these results suggest both H3.1 and

H3.3 are retained stronger at the α-satellite and pericentromeric

satellite 2 in comparison to genes. However, despite these possible

trends we were unable to establish whether these differences between

loci are significant, given the current experimental variance in the data

(Figure 3.3). Both nucleosome types turnover in a biphasic pattern

where most nucleosomes are lost in the first 12 hours followed by a

much slower turnover at longer time scales (Figure 3.2).

Page 137: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

117

Figure 3.2 TimeChIP analysis of H3.1-SNAP and H3.3-SNAP. A) TimeChIP

analysis as described in Figure 3.1 of H3.1-SNAP and H3.3-SNAP cells at

ACTB, GAPDH, TBP and RPL13A genes for indicated time points. B)

Histones H3.1-SNAP and H3.3-SNAP were biotinylated and chase at α-

satellite I repeat, pericentromeric satellite 2 repeats (Sat2) and a conserved

non-coding element (gene desert) at indicated time points. Fraction of biotin

retained values are derived from fitting of the %IP measurements of chased

samples on a linear regression of a TimeChIP standard curve that was

included in each experiment (see Chapter 2). Data shown represent average

of five independent experiments, error bars indicate standard error of the

mean (SEM). Statistically significant difference in the comparison is indicated

by asterisk (*) p<0.05, (**) p<0.01 (Kruskall-Wallis test).

Page 138: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

118

Figure 3.3 Comparison of the rate of H3.1 and H3.3-SNAP retention at

genes and non-gene loci. Fraction of H3.1-SNAP and H3.3-SNAP retained

at genes (top) and non-genes (bottom) at indicated time points. Fraction of

biotin retained values are derived from fitting of the %IP measurements of

chased samples on a linear regression of a TimeChIP standard curve that

was included in each experiment (see Chapter 2). Data shown represent

average of six (left) and five independent experiments (right), error bars

indicate standard error of the mean (SEM). Statistically significant difference

in the comparison is indicated by asterisk (**) p<0.01, (***) p<0.001, no

statistically significant difference in the comparison is indicated by ns (Two-

way ANOVA, Bonferroni correction)

Local retention of H3.1

The finding that ancestral H3.1 and H3.3 are retained at the

different loci raises the question whether such retention occurs locally.

Three different scenarios could be considered: 1) histones are locally

retained; 2) histones turnover and 3) alternative to turnover, ancestral

histones could be turned over in another location, released into the

soluble pool and “recycled” by re-incorporation at the locus under

Page 139: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

119

investigation. In this later case, ancestral histones are detected but

they are dynamically retained rather than locally (Figure 3.4). There is

however no precedent for histones that are released from chromatin

into the soluble pool to be reincorporated since chromatin based

histones differ in their post translational modification from the soluble

histone pool (Groth et al., 2007). Nevertheless, to address this

possibility we took advantage of the assembly dynamics of H3.1.

Deposition of nascent H3.1 occurs during S phase by the histone

chaperone CAF-1 in strictly DNA replication dependent manner

(Ahmad and Henikoff, 2002; Ray-Gallet et al., 2011; Shibahara and

Stillman, 1999). In contrast, incorporation of newly synthesized H3.3

occurs throughout the cell cycle including S phase but independent of

DNA synthesis by the HIRA complex, ATRX and DAXX chaperones

(Ahmad and Henikoff, 2002; Drané et al., 2010; Lewis et al., 2010).

Because H3.1 assembly is S phase dependent there will be no

deposition of either nascent or ancestral H3.1 outside of S phase. This

creates an opportunity to determine whether the retention of ancestral

canonical H3.1 is due to local or dynamic retention. We treated cells

with Thymidine for 14 hours to prevent DNA replication and

incorporation of both labeled new and labeled old H3.1-SNAP while

ancestral H3.1 was chased (Figure 3.5 A). The efficiency of the arrest

was assessed with BrdU labeling which was included during the

duration of the experiment, and showed that 5% of H3.1-SNAP cells

thymidine treated do not incorporated BrdU while non-arrested H3.1-

SNAP cells incorporated BrdU in 88.4% (Figure 3.5 C). FACS analysis

shows that when H3.1-SNAP cells are arrested with Thymidine there is

an enrichment of cells in G1 phase and a concurrent loss of G2/M cells

in comparison with untreated cells. This indicates that Thymidine arrest

does not allow cells to passage through S phase cells as expected

(Figure 3.5 D). TimeChIP analysis shows that following Thymidine

Page 140: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

120

arrest levels of ancestral H3.1 are maintained at GAPDH and Sat2,

with a small increase at the actin gene (Figure 3.5 B). If the apparent

retention of H3.1 at these loci would be the result of dynamic

reincorporation of old H3.1-SNAP than one would expect that upon

blocking of assembly histone loss would become apparent and no

signal would be retained. The fact that H3.1 levels are maintained, or

even increases, strongly indicates that H3.1-SNAP histones are

retained locally. Although the levels of H3.3 did not change at the

different loci when cells were arrested with Thymidine, we cannot

determine conclusively whether reassembly of labeled H3.3 has

occurred as this may continue during the S phase arrest (Figure 3.5

B). From these results, we conclude that ancestral canonical H3.1 is

locally retained at both active and silent loci.

Figure 3.4 Outline of possible modes of H3.1-SNAP retention. A) Local

retention. The amount of labeled H3.1-SNAP in a given time (time n, tn) is

equal to the number of the initially labeled H3.1-SNAP pool (t0). B) Turnover.

The number of labeled H3.1-SNAP at time n is lower than in time 0. C)

Dynamic retention. The number of labeled H3.1-SNAP at time 0 is equal than

at time n, but there is an intermediate stage during the chase where H3.1 is

lost after which labeled H3.1-SNAP from a different locus is incorporated.

Page 141: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

121

Figure 3.5 H3.1-SNAP is locally retained. A) Schematic of H3.1-SNAP and

H3.3-SNAP pulse-chase and blocking of DNA replication. B) TimeChIP

analysis of H3.1-SNAP and H3.3-SNAP at Actin and GAPDH genes and

major satellite 2 (Sat2) during normal cell proliferation and during Thymidine

arrest for 12 hours. Mean +/- S.E.M. are shown, n=3. No statistically

significant difference in the comparison is indicated by ns (paired, t-test

p<0.05). C) Thymidine arrested and asynchronous H3.1-SNAP cell

populations were pulse labeled with BrdU in the presence or absence of

Thymidine to determine efficiency of arrest. BrdU labeling was visualized by

immunofluorescence (Cy5). BrdU staining is absent from Thymidine arrested,

contrary untreated to cells. Below: Quantification of the number of BrdU+

cells. Number of cells analyzed (n) is indicated. D) FACS analysis of DNA

Page 142: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

122

content of H3.1-SNAP cells either pulse labeled (pulse), released (12h chase)

and released in Thymidine. H3.1-SNAP cells arrested with Thymidine exhibit

enrichment in G1 phase and lack a G2/M population indicating an S phase

arrest. Green line represents an estimation of the G0/G1, S and G2/M

population sizes.

Genomic distribution and turnover of H3.3

We have shown that ~10-20% of parental H3.3 is present after

48 hours chase at different loci (Figure 3.2). However, H3.3 is

incorporated at virtually all expressed gene loci as well as at enhancer

elements (Goldberg et al., 2010; Jin et al., 2009; Ray-Gallet et al.,

2011). Therefore, to determine whether our results for the loci we

analyzed are indicative for what happens across the genome we

carried out TimeChIP-seq. In TimeChIP-seq, H3.3-SNAP cells were in

vivo pulse labeled with CP-biotin and chased for 12 and 24 hours. Salt-

extracted chromatin was fragmented into mononucleosomes by

MNase treatment. H3.3-SNAP biotinylated nucleosomes were

captured on Streptavidin magnetic beads. 10ng of co-purified DNA

from recovered H3.3-SNAP nucleosomes were submitted for

generation of sequencing libraries followed by Illumina sequencing at

high coverage. TimeChIP allows for the analysis of a limited number of

loci while with TimeChIP-seq the coverage is genome wide. However,

we cannot estimate the absolute retention rate of H3.3-SNAP with

TimeChIP-seq. H3.3 enrichment is measured as the number of

sequence reads for each locus normalized against the total number of

reads. Due to the wide distribution of H3.3 across the genome, both

local read counts as well as global read counts change significantly

during the chase period, making it difficult to make an absolute

quantitative statement on local histone retention. However, the

approach does allow for relative measurements between loci over

time. In agreement with our observations based on qPCR, we find that

H3.3-SNAP turnover is faster at the most transcribed genes compared

Page 143: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

123

to unexpressed or lowly expressed genes (Figure 3.6 C). Turnover is

more rapid near the transcription termination site (TTS) compared to

the transcription start site (TSS) (Figure 3.6 B & C). Furthermore,

H3.3-SNAP exhibits to be highly enriched at enhancers and displays a

turnover rate exceeding that of active genes (Figure 3.6 C). At loci

enriched in H3.3 we find that turnover is more rapid in the first 12

hours compared to the 2nd 12 hours indicating a discontinues turnover

rate, similar to what we have observed for specific loci by qPCR

(Figure 3.2). Interestingly, in this experiment we find that, overtime,

ancestral H3.3 accumulates just downstream of the TSS, reminiscent

of the distribution of ancestral H3 observed in budding yeast (Figure

3.6 A) (Radman-Livaja et al., 2011).

Figure 3.6 TimeChIP analysis of H3.3-SNAP at the genome-wide level. A)

Coverage of H3.3-SNAP at the top 1000 expressed genes at 0 (dark blue), 12

(blue) and 24 hours (light blue). Enrichment of H3.3-SNAP (y axis) was

estimated based on the number of reads per total number of reads relative to

the number of read per total number of reads in the input. X axis represents

distribution of H3.3-SNAP at 5 kb upstream of TSS, across the gene body and

Page 144: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

124

5kb downstream of the TTS of most expressed genes (GEO code

GSM958735)(ENCODE Project Consortium, 2011). B) H3.3-SNAP turnover

for highly expressed genes. The average turnover index of H3.3-SNAP was

calculated for regions described for A) for the top 1000 genes according to

RNA-seq data from ENCODE. The turnover rate (y axis) for H3.3 was plotted

as the ratio of enrichment at 12h relative to 0h (dark blue) and as the ratio of

enrichment at 24h relative to 12h (light purple) at top 1000 expressed genes.

1= no change; <1 = histone loss; >1 = histone enrichment. C) Relative H3.3-

SNAP turnover at transcription start site (TSS) and transcription terminal site

(TTS) of top and bottom 1000 expressed genes, as well as enhancers.

Distribution is shown by box plots that show median (black bar), 25th and 75th

percentile (box) and Min-Max (whiskers) H3.3-SNAP turnover of the first 12

hours (dark blue) and H3.3-SNAP turnover the following 12 hours (12-24

hours) (light blue).

Differential histone retention within active gene

Previous reports have indicated that nucleosomes are unstable

just upstream of the transcription start site (Jin et al., 2009). To

determine whether this correlates with fast histone turnover we sought

to quantify the retention of H3.3-SNAP by TimeChIP at the promoter

region and gene body of the basally expressed nuclear transcription

factor-ƙβ (NF-ƙβ). We find H3.3 turnover to be much slower at exon 2

of NF-ƙβ gene body compared to the transcription start site where only

~20-30% of H3.3 is retained in the first 3 hours of chase with a further

reduction to <10% after 12 hours (Figure 3.7). Interestingly, H3.1 is

retained longer at the TSS in comparison to H3.3 while H3.1 turnover

in the gene body is comparable to that of H3.3 (Figure 3.7). We can

conclude that at least at of NF-ƙβ, nucleosome turnover is faster at the

TSS and that H3.1 and H3.3 are impacted differently at the promoter of

this gene.

Page 145: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

125

Figure 3.7 Histones turnover within NF-ƙB gene. TimeChIP experiment as

in Figure 3.1 A) Levels of H3.1 and H3.3-SNAP retained at transcription start

site of NF-ƙB gene. B) Levels of H3.1-SNAP and H3.3-SNAP retained at gene

body (exon 2) of NF-ƙB at indicated time points. Mean +/- S.E.M. are shown,

n=3. Statistically significant difference in the comparison is indicated by

asterisk (*) p<0.05, (***) p<0.001 (One way ANOVA Bonferroni correction). C)

Schematic of NF-ƙB gene indicating regions flanked by PCR probes used to

quantify H3.1 and H3.3 occupancy at transcription start site (TSS) and gene

body (exon 2).

Dynamics of H3.1 and H3.3 during transcription activation

Our assessment of different gene loci by genome wide analysis

indicates that nucleosome retention correlates negatively with

transcription. i.e. higher transcription rates drive more histone loss.

Next, we sought to directly test whether gene activity correlates with

higher rates of histone turnover. We used the activation of the nuclear

transcription factor-ƙβ (NF-ƙβ) gene by the cytokine TNF-α as a means

of conditionally activating transcription. The rapid transcriptional

activation of NF-ƙβ by TNF-α is well characterized (Mahoney et al.,

2008). We measured the rate of H3.1 and H3.3 retention following

gene activation by TNF-α addition. A 3h treatment with TNF-α resulted

Page 146: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

126

in an increase of NF-ƙβ mRNA by ~2 fold and a 3 fold increase in the

transcript levels of the downstream gene cIAP2 (Figure 3.8 C). To

confirm that the activation of NF-ƙβ pathway occurs in the majority of

the cells, we determined the localization of NF-ƙβ by

immunofluorescence (Figure 3.8 D). ~94% of cells exhibited NF-ƙβ

translocation to the nucleus upon TNF-α treatment. Interestingly,

despite the NF-ƙβ activation in the vast majority of cells and the

concurrent transcriptional activation of this gene, retention of H3.1 did

not change during transcription activation of NF-ƙβ, both at the gene

body and transcription start site (TSS) (Figure 3.8 B). H3.3 turns over

faster at the TSS of NF-ƙβ in comparison with canonical histone H3.1,

and this rapid rate is not further accelerated by TNF-α. There is a

minor but not significant increase in retention of H3.3 in the gene body

of NF-ƙβ following induction (Figure 3.8 B). These results suggest that

activation of a gene does not lead to a measurable increase in the

turnover of histones H3.1 and H3.3.

Page 147: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

127

Figure 3.8 Histone turnover during transcription activation. A) Schematic

of pulse-chase labeling of cells expressing SNAP-tagged histones and gene

induction by TNF-α. B) Levels of ancestral H3.1-SNAP and H3.3-SNAP at the

transcription start site and exon 2 of the TNF-α induced NF-ƙβ gene.

Schematic representation of NF-ƙβ gene, indicating regions flanked by PCR

probes. No statistically significant difference in the comparison is indicated by

ns (paired, t-test p<0.05). C) Quantification of TNF-α mediated induction of

NF-ƙB mRNA and the downstream cIAP2 gene in H3.3-SNAP cells by qRT-

PCR. D) Subcellular localization of NF-ƙβ in untreated and TNF-α treated

cells. 94% of cell population displayed nuclear staining following TNF-α

addition. Results of TimeChIP and levels of mRNA represent average of three

independent experiments, error bars indicate standard error of the mean

(SEM).

Page 148: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

128

Dynamics of H3.1 and H3.3 during transcription inhibition

Our analysis of histone turnover at an active gene during

transcription induction did not reveal a detectable increase in histone

loss from chromatin. We next examined whether the turnover of H3.1

and H3.3 can be slowed down by directly interfering with transcription.

We treated the cells with DRB and α-Amanitin. DRB prevents

activating phosphorylation of the RNA polymerase II C-terminal

domain (CTD), which results in repression of transcription elongation

(Yankulov et al., 1995). α-Amanitin binds to the largest subunits of

RNA polymerase II and III (Cassé et al., 1999). To assess the

inhibitory effect of DRB and α-Amanitin on genes transcribed by RNA

polymerase II, we selected H3.1L, cyclin A (CCNA), Myc proto–

oncogen protein (MYC) and ribosomal protein P0 (RPLP0) genes.

H3.1L, CCNA and MYC genes have in common that the half life of

mRNA is relatively short (less than 4 hours) due to their cell cycle

regulated expression (Lam et al., 2001). RPLP0 has a half life of 8.5

hours (Jb et al., 2009). These half-lives allows us to detect the

inhibitory effect of DRB and α-Amanitin by analysis of steady-state

mRNA levels. In a pilot experiment to determine optimal drug

concentration and incubation times to be included in the TimeChIP

experiment we observed that transcript levels decrease with 100µM

DRB for 3 hours and 10µg/mL α-Amanitin for 12 hours (Figure 3.9).

Following treatment with these inhibitors we measured histone

turnover by TimeChIP. The retention of ancestral H3.1 increased ~2-

2.5 fold at ACTB, GAPDH, TBP, the gene desert and Sat2, and 5 fold

at RPL13A when cells are treated with α-Amanitin for 12 hours (Figure

3.10 B, C). DRB treatment slowed H3.1 turnover at the Sat2 and gene

desert loci. At the active RPL13A locus a trend of slow turnover was

observed although not significant given the error in measurement

(Figure 3.10 B, C). Similarly, slow turnover of H3.3 during transcription

inhibition with DRB is observed at TBP, RPL13A and gene desert,

Page 149: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

129

although again not significant from the experimental variance (Figure

3.10 B, C). When transcription is blocked with α-Amanitin, retention of

H3.3 increases 5 fold at GAPDH and at the conserved non-coding

gene desert locus, ~2.5-3 fold at ACTB, TBP, Sat2, α-satellite I repeats

and RPL13A (Figure 3.10 B, C). Although increased retention is not

statistically significant for some loci, all loci follow the same trend.

Because inhibition of RNA polymerase II decreases the turnover of

H3.1 and H3.3 at the conserved non-coding element, we postulated

that this locus might, in fact, be transcribed and as a result respond to

DRB and α-Amanitin treatment. However, we could not detect any

mature transcripts at this locus by qRT-PCR analysis (data not shown).

Moreover, RNA-seq analysis of HeLa cells reports no read counts at

this locus (GEO code GSM958735)(ENCODE Project Consortium,

2011). Instead, it has been reported to be a lamina associated domain

in Tig3 cells and proximal to an enrichment of H3K27Ac mark

(ENCODE Project Consortium, 2011).

Figure 3.9 DRB and α-Amanitin inhibit transcription. Relative mRNA

levels of H3.1L, cyclin A (CCNA), Myc proto–oncogen protein (MYC) and

ribosomal protein P0 (RPLP0) normalized to an untreated control following

treatment with RNA polymerase II inhibitors DRB and α-Amanitin. Mean +/-

S.E.M. are shown, n=3.

Page 150: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

130

Figure 3.10 Histone turnover during transcription inhibition. A)

Schematic of H3.1-SNAP and H3.3-SNAP pulse-chase experiments in

combination with transcription inhibitors. B) TimeChIP analysis of H3.1-SNAP

and H3.3-SNAP at ACTB, GAPDH, TBP and RPL13A genes after 100µM

DRB and 10µg/mL α-Amanitin treatment for 3 hours and 12 hours,

respectively. Untreated samples were analyzed for the same time frame. C)

TimeChIP analysis of H3.1-SNAP and H3.3-SNAP at α-satellite I repeat,

pericentromeric satellite 2 repeats (Sat2) and a conserved non-coding

element (gene desert). Mean +/- S.E.M. are shown, n=3. Statistically

significant difference in the comparison is indicated by asterisk (*) (paired t-

test p<0.05).

Page 151: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

131

The retention of H3.1 and H3.3 increases in most of the loci

analyzed when RNA polymerase II transcription is inhibited with DRB

and α-Amanitin, including surprisingly, a non-transcribed locus. To

examine whether this effect extends to other loci that are not

transcribed by RNA polymerase II, we measured the retention of H3.1

and H3.3 at the 5SrDNA locus, which is transcribed by RNA

polymerase I and insensitive to α-Amanitin and DRB (Price and

Penman, 1972; Yankulov et al., 1995) (Figure 3.11 B). Despite the

prediction that RNA polymerase II does not impact transcription of

rDNA loci we found that retention of ancestral H3.1 and H3.3 increases

by two fold after α-Amanitin treatment, and ~1.5 fold when cells are

treated with DRB (Figure 3.11 A).

Figure 3.11 Histone turnover at 5SrDNA locus. A) TimeChIP analysis of

H3.1-SNAP and H3.3-SNAP at 5SrDNA during treatment with 100µM DRB

and 10µg/mL α-Amanitin. B) qRT-PCR results show that levels of 5SrDNA do

not decrease by treatment with 10µg/mL α-Amanitin and 100µM DRB. Mean

+/- S.E.M. are shown, n=3. Statistically significant difference in the

comparison is indicated by asterisk (*) (paired t-test p<0.05).

Page 152: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

132

DISCUSSION

In this chapter we applied our newly developed Time-ChIP

methodology to the analysis of histone retention in mitotically dividing

HeLa cells in culture. We find that while histones are turned over, a

significant fraction of canonical H3.1 is retained across the cell cycle at

non-genes and genes loci. Importantly, we show that retention of H3.1

at different loci is maintained in the absence of DNA synthesis during

which no deposition of H3.1 occurs. This shows for the first time that

H3.1 is retained locally at active loci in mammalian cells. H3.1 is

generally more stable at centromeric and pericentromeric repeats in

comparison to active genes. It is not clear from our results whether this

stability is as high as has been observed for CENP-A at centromeres,

although CENP-A appears to be more strongly retained at early time

points (Chapter 2, Figure 2.7). Consistently, quantification of histone

turnover at centromeres in a cell by cell analysis at optical resolution,

indicates that H3.1 and H3.3 turn over faster than CENP-A even when

present at the centromere locus (Bodor et al., 2013). Ectopic targeting

of CENP-A is sufficient to lead to self-replication of centromeric

chromatin and heritable kinetochores formation for several cell

divisions (Mendiburo et al., 2011). This indicates that CENP-A has the

capacity to induce and sustain a self-maintaining feedback

mechanism. The existence of such a mechanism of propagation for

H3.1 which is distributed throughout chromatin is unlikely.

We find that a proportion of H3.1 is locally retained even at

active genes. Early in vivo experiments in budding yeast have shown

that nucleosomes are quickly and efficiently positioned on nascent

chains after the passage of the replication fork (Lucchini et al., 2001).

Recently, a quantitative model based on measurements of ancestral

H3 in budding yeast considering the total histone turnover, lateral

movement of histones and dissociation/re-association during

Page 153: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

133

replication; indicates that ancestral histone are reincorporated within

400bp of the original site (Radman-Livaja et al., 2011). We now

provide direct evidence for such local retention of nucleosomes also

during transcription in human somatic cells. We find that during

Thymidine arrest, during which histone H3.1 cannot be assembled, the

previously incorporated pool is retained at non genes and genes.

Similarly, we see that H3.3 levels are unaffected by Thymidine

addition. However, since H3.3 assembly is not cell cycle restricted, we

cannot conclude whether the retention is local or whether dynamic

reassembly occurs.

We see that both H3.1 and H3.3 turnover in a biphasic manner.

Particularly, we find that retention of ancestral H3.3 at active genes

decreases by half after the first 3 hours of chase while the levels drop

at a slower pace after 6 and 12 hours. Like H3.3, H2A.Z is deposited

into chromatin throughout the cell cycle. Unlike H3.3, H2A.Z is

enriched at promoters and sites of DNA double strand breaks

(Kalocsay et al., 2009). Nucleosomes carrying both the H3.3 and

H2A.Z variants have been shown to be highly enriched at transcription

start sites (TSS) and CTCF binding sites. In addition, such double

variant nucleosomes are highly unstable as inferred by their low salt

stability (Jin 2009). In contrast, Nekrasov et al., suggest that the

instability at CTCF binding site is given by the heterotypic histones

H2A.Z-H2A nucleosome (Nekrasov et al., 2012). High-resolution maps

of homotypic and heterotypic Drosophila H2A.Z nucleosomes show

that homotypic nucleosomes are enriched downstream of active

promoters and intron-exon junctions, while heterotypic nucleosomes

are depleted from these regions (Weber et al., 2010). The enrichment

of H2A.Z at active genes may provide an explanation for the rapid

initial loss of ~50% of H3.3 at genes during the first 3 hours chase.

Possibly the ancestral H3.3 that is associated with H2A.Z is lost rapidly

Page 154: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

134

while the pool that is complexed with canonical H2A is more stably

retained.

We show that ancestral H3.3 turnover is fast at promoters while

is stronger retention is observed at active gene bodies and non-gene

loci. In contrast, a previous study in Drosophila cells indicates that H3

has a turnover rate of ~1-1.5 hours at active genes bodies, promoters

and regulatory elements suggesting a rapid nucleosome turnover in

manner dependent on gene expression (Deal et al., 2010). However,

the approach used by Deal et al., where histones are metabolically

labeled, measures the turnover rate exclusively of recently assembled

newly synthesized histones. A possible explanation to reconcile our

results with the Deal et al study is that there are qualitatively different

populations of nucleosomes with different turnover rates, a dynamic

population that is replaced several times during one cell cycle and a

second population that is retained at longer timescales part of which

can be retained throughout across the cell cycle. While we have

obtained evidence that H3.1 is maintained locally in chromatin, we

cannot determine whether H3.3 is retained locally despite the fact that

some retention of parental H3.3 is observed after a 48 hours chase at

active genes. Deposition of H3.3 is facilitated by HIRA, ATRX and

DAXX throughout the cell cycle; which make it difficult to determine

whether retention is local or whether ancestral pools of H3.3 are

dynamically reassembled overtime (Drané et al., 2010; Goldberg et al.,

2010; Lewis et al., 2010; Ray-Gallet et al., 2002). In agreement with

previous studies, genome-wide analysis shows that H3.3 is enrichment

at highly transcribed genes, promoters and enhancers (Goldberg et al.,

2010). Interestingly, we see that turnover rates are different for these

different loci with enhancers sporting the highest H3.3 dynamics.

Moreover, we found that similar to dynamics of H3 in budding yeast

(Radman-Livaja et al., 2011), H3.3 nucleosomes accumulate toward

Page 155: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

135

the 5’ end of genes over time, although this result is preliminary at this

stage. An explanation could be that nucleosomes slide during

disassembly/reassembly process while RNA polymerase II acts in

conjunction with chromatin remodeling (Fazzio and Tsukiyama, 2003;

Radman-Livaja et al., 2011).

Nucleosomes form barriers for transcription. By inducing

transcription of the stress response gene NF-ƙB, we found that,

contrary to our expectation, turnover of H3.1 and H3.3 was not further

increased significantly. There are two alternatives that histones have

during transcription, either histones are evicted or transferred behind

the polymerase. In vitro studies with phage SP6 RNA polymerase and

yeast RNA polymerase III proposed the formation of an

intranucleosomal DNA loop that leads to an intermediate pause of the

RNA polymerase while histones are transferred to the same DNA

molecule (Bintu et al., 2011; Felsenfeld et al., 2000; Studitsky et al.,

1994, 1997). An increase of transcription activity could lead to an

increase of the exposure time of accessible nucleosomal DNA which

implies that histones will be prone to eviction during elevated

transcription. Possibly in our human cell experiments nucleosome

retention at active genes is achieved through similar intranucleosomal

DNA transfer mechanisms. An increase in transcription may be

achieved either by a faster transcription rate or by a higher density of

RNA polymerases. In the latter case the dynamics of transitional

intranucleosomal DNA looping during elongation of RNA polymerase

may not need to change, thereby maintaining a similar degree of

nucleosome turnover as we observe at the NF-ƙB gene upon

activation by TNF-α.

Jin et al., have proposed that an initial group of RNA

polymerases during transcription elongation, acting as a pioneer, will

evict histones to facilitate transcription by the trailing RNA polymerases

Page 156: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

136

(Jin et al., 2010; Orphanides and Reinberg, 2000). This may be a

mechanism that gives rise to differential nucleosome stabilities.

Nucleosome retention during transcription requires the participation of

histone chaperones and chromatin remodelers. Evidence comes from

Jamai et al., who show that inactivation of the nucleosome assembly

factor Spt16 leads to loss of H2B and H3 on active and inactive genes.

Histone loss is rescued when transcription is inhibited, indicating that

Spt16 contributes in the reassembly of histones during transcription

(Jamai et al., 2009). A similar role has been proposed for the FACT

complex that facilitates chromatin transcription by RNA polymerase II,

by reassembly of H2A-H2B dimers after enzyme passage. However, in

this study FACT did not impact on H3-H4 tetramers (Belotserkovskaya

et al., 2003). In future efforts it will be of interest to determine the role

of these histone chaperones in the retention of nucleosomes during

transcription.

We find that histone turnover decreases significantly at all loci

tested when transcription by RNA polymerase II is inhibited. This is

contrary to our expectation of reduced histone loss only at sites of

transcription by RNA polymerase II. Panse et al., have described that

transcription of 45S RNAs is maintained even if RNA polymerase II

transcription is arrested by DRB (Panse et al., 1999). In agreement, we

see that 5SrDNA is transcribed under DRB and α-Amanitin arrest but

histone turnover is nevertheless significantly diminished.

At this point we have no adequate explanation of this

phenomenon. Possible models include: 1) all genomic sequences are

transcribed at some rate by RNA polymerase II leading to Pol II driven

nucleosome turnover, irrespective of transcription rate (if so, this would

mean that transcription occurs below the detection limit of our mRNA

measurements). 2) The chromatin response is systemic in nature

rather than local. Either the drug treatments induce a global stress

Page 157: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

137

response that leads to an overall reduction in nucleosome turnover or,

more interestingly, transcription inhibition leads to changes in local

chromatin domains. In light of this, it has been proposed that the

transcriptional program of a cell may be dependent of the spatial

organization of the genome. Genes that are actively transcribed share

transcription factories, described as sub-nuclear foci with high local

concentrations of RNA polymerase II that are sensitive to α-Amanitin.

These clusters are specialized in particular transcriptional pathways

and driven by discrete transcription factors (Bickmore and van

Steensel, 2013; Sexton et al., 2007). Moreover, 3D analysis of

chromatin domains identified two structures: ridges and antiridges; that

differ in transcriptional activity, the presence of specific proteins and

noncoding RNAs and have a distinct chromatin folding. Although

evidence for the molecular mechanisms is lacking, it seems that

specific sub-nuclear microenvironments are formed that promote high

transcription rates (ridges) or allow only low to moderate transcriptional

activities (antidridges) (Goetze et al., 2007). One speculative

explanation for our results could be that transcription inhibition leads to

histone retention in a genomic region much broader than individual

genes. This may involve changes in histone modifications, compaction

of chromatin, and formation of clusters that drive changes in nuclear

organization and dynamics. Hypothetically, the global inhibition of

transcription could stimulate the mobilization of chromatin to the

nuclear lamina after chromatin has been disengaged of transcription

factories. The results of histone turnover during transcription inhibition

are intriguing. Further experiments in the context of genome

architecture are needed.

In conclusion, our results show that a population of parental

H3.1 and H3.3 histones is stably retained at the timescale of the cell

Page 158: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

138

cycle. Consequently such subpopulation could be responsible to carry

PTM contributing to epigenetic maintenance of gene expression.

Page 159: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

139

ACKNOWLEDGEMENTS

I would like to acknowledge Ivan Correa from New England

Biolabs, Ipswich, MA, for the synthesis of the CP-biotin substrate. I

also thank Daniel Sobral for analysis and discussions of high-through

sequencing experiments, as well as Raffaella Gozzelino and Miguel

Soares for providing reagents.

Page 160: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

140

REFERENCES

Ahmad, K., and Henikoff, S. (2002). The Histone Variant H3.3 Marks Active Chromatin by Replication-Independent Nucleosome Assembly. Mol. Cell 9, 1191–1200.

Belotserkovskaya, R., Oh, S., Bondarenko, V.A., Orphanides, G., Studitsky, V.M., and Reinberg, D. (2003). FACT facilitates transcription-dependent nucleosome alteration. Science 301, 1090–1093.

Bergmann, J.H., Rodríguez, M.G., Martins, N.M.C., Kimura, H., Kelly, D.A., Masumoto, H., Larionov, V., Jansen, L.E.T., and Earnshaw, W.C. (2011). Epigenetic engineering shows H3K4me2 is required for HJURP targeting and CENP-A assembly on a synthetic human kinetochore. EMBO J. 30, 328–340.

Bickmore, W.A., and van Steensel, B. (2013). Genome architecture: domain organization of interphase chromosomes. Cell 152, 1270–1284.

Bintu, L., Kopaczynska, M., Hodges, C., Lubkowska, L., Kashlev, M., and Bustamante, C. (2011). The elongation rate of RNA polymerase determines the fate of transcribed nucleosomes. Nat. Struct. Mol. Biol. 18, 1394–1399.

Bodor, D.L., Valente, L.P., Mata, J.F., Black, B.E., and Jansen, L.E.T. (2013). Assembly in G1 phase and long-term stability are unique intrinsic features of CENP-A nucleosomes. Mol. Biol. Cell 24, 923–932.

Cassé, C., Giannoni, F., Nguyen, V.T., Dubois, M.F., and Bensaude, O. (1999). The transcriptional inhibitors, actinomycin D and alpha-amanitin, activate the HIV-1 promoter and favor phosphorylation of the RNA polymerase II C-terminal domain. J. Biol. Chem. 274, 16097–16106.

Deal, R.B., Henikoff, J.G., and Henikoff, S. (2010). Genome-Wide Kinetics of Nucleosome Turnover Determined by Metabolic Labeling of Histones. Science 328, 1161–1164.

Drané, P., Ouararhni, K., Depaux, A., Shuaib, M., and Hamiche, A. (2010). The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev. 24, 1253–1265.

Page 161: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

141

ENCODE Project Consortium (2011). A user’s guide to the encyclopedia of DNA elements (ENCODE). PLoS Biol. 9, e1001046.

Fazzio, T.G., and Tsukiyama, T. (2003). Chromatin remodeling in vivo: evidence for a nucleosome sliding mechanism. Mol. Cell 12, 1333–1340.

Felsenfeld, G., Clark, D., and Studitsky, V. (2000). Transcription through nucleosomes. Biophys. Chem. 86, 231–237.

Goetze, S., Mateos-Langerak, J., Gierman, H.J., de Leeuw, W., Giromus, O., Indemans, M.H.G., Koster, J., Ondrej, V., Versteeg, R., and van Driel, R. (2007). The three-dimensional structure of human interphase chromosomes is related to the transcriptome map. Mol. Cell. Biol. 27, 4475–4487.

Goldberg, A.D., Banaszynski, L.A., Noh, K.-M., Lewis, P.W., Elsaesser, S.J., Stadler, S., Dewell, S., Law, M., Guo, X., Li, X., et al. (2010). Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 140, 678–691.

Gómez-Rodríguez, M., and Jansen, L.E. (2013). Basic properties of epigenetic systems: lessons from the centromere. Curr. Opin. Genet. Dev. 23, 219–227.

Groth, A., Corpet, A., Cook, A.J.L., Roche, D., Bartek, J., Lukas, J., and Almouzni, G. (2007). Regulation of Replication Fork Progression Through Histone Supply and Demand. Science 318, 1928–1931.

Jackson, V. (1990). In vivo studies on the dynamics of histone-DNA interaction: evidence for nucleosome dissolution during replication and transcription and a low level of dissolution independent of both. Biochemistry (Mosc.) 29, 719–731.

Jamai, A., Puglisi, A., and Strubin, M. (2009). Histone chaperone spt16 promotes redeposition of the original h3-h4 histones evicted by elongating RNA polymerase. Mol. Cell 35, 377–383.

Jansen, L.E.T., Black, B.E., Foltz, D.R., and Cleveland, D.W. (2007). Propagation of centromeric chromatin requires exit from mitosis. J. Cell Biol. 176, 795–805.

Jb, A., K, M.-M., M, Z., M, G., and Ba, H. (2009). Ribosomal protein mRNAs are primary targets of regulation in RNase-L-induced senescence., Ribosomal protein mRNAs are primary targets of

Page 162: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

142

regulation in RNase-L-induced senescence. RNA Biol. RNA Biol. 6, 6, 305, 305–315.

Jin, C., Zang, C., Wei, G., Cui, K., Peng, W., Zhao, K., and Felsenfeld, G. (2009). H3.3/H2A.Z double variant-containing nucleosomes mark “nucleosome-free regions” of active promoters and other regulatory regions. Nat. Genet. 41, 941–945.

Jin, J., Bai, L., Johnson, D.S., Fulbright, R.M., Kireeva, M.L., Kashlev, M., and Wang, M.D. (2010). Synergistic action of RNA polymerases in overcoming the nucleosomal barrier. Nat. Struct. Mol. Biol. 17, 745–752.

Kalocsay, M., Hiller, N.J., and Jentsch, S. (2009). Chromosome-wide Rad51 spreading and SUMO-H2A.Z-dependent chromosome fixation in response to a persistent DNA double-strand break. Mol. Cell 33, 335–343.

Kimura, H., and Cook, P.R. (2001). Kinetics of Core Histones in Living Human Cells Little Exchange of H3 and H4 and Some Rapid Exchange of H2b. J. Cell Biol. 153, 1341–1354.

Lam, L.T., Pickeral, O.K., Peng, A.C., Rosenwald, A., Hurt, E.M., Giltnane, J.M., Averett, L.M., Zhao, H., Davis, R.E., Sathyamoorthy, M., et al. (2001). Genomic-scale measurement of mRNA turnover and the mechanisms of action of the anti-cancer drug flavopiridol. Genome Biol. 2, RESEARCH0041.

Lewis, P.W., Elsaesser, S.J., Noh, K.-M., Stadler, S.C., and Allis, C.D. (2010). 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. 107, 14075–14080.

Livak, K.J., and Schmittgen, T.D. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods San Diego Calif 25, 402–408.

Lucchini, R., Wellinger, R.E., and Sogo, J.M. (2001). Nucleosome positioning at the replication fork. EMBO J. 20, 7294–7302.

Luger, K., Mäder, A.W., Richmond, R.K., Sargent, D.F., and Richmond, T.J. (1997). Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389, 251–260.

Page 163: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

143

Luger, K., Dechassa, M.L., and Tremethick, D.J. (2012). New insights into nucleosome and chromatin structure: an ordered state or a disordered affair? Nat. Rev. Mol. Cell Biol. 13, 436–447.

Mahoney, D.J., Cheung, H.H., Mrad, R.L., Plenchette, S., Simard, C., Enwere, E., Arora, V., Mak, T.W., Lacasse, E.C., Waring, J., et al. (2008). Both cIAP1 and cIAP2 regulate TNFalpha-mediated NF-kappaB activation. Proc. Natl. Acad. Sci. U. S. A. 105, 11778–11783.

Mendiburo, M.J., Padeken, J., Fülöp, S., Schepers, A., and Heun, P. (2011). Drosophila CENH3 Is Sufficient for Centromere Formation. Science 334, 686–690.

Nekrasov, M., Amrichova, J., Parker, B.J., Soboleva, T.A., Jack, C., Williams, R., Huttley, G.A., and Tremethick, D.J. (2012). Histone H2A.Z inheritance during the cell cycle and its impact on promoter organization and dynamics. Nat Struct Mol Biol 19, 1076–1083.

Nobrega, M.A., Ovcharenko, I., Afzal, V., and Rubin, E.M. (2003). Scanning human gene deserts for long-range enhancers. Science 302, 413.

Orphanides, G., and Reinberg, D. (2000). RNA polymerase II elongation through chromatin. Nature 407, 471–475.

Panse, S.L., Masson, C., Héliot, L., Chassery, J.M., Junéra, H.R., and Hernandez-Verdun, D. (1999). 3-D organization of ribosomal transcription units after DRB inhibition of RNA polymerase II transcription. J. Cell Sci. 112 ( Pt 13), 2145–2154.

Price, R., and Penman, S. (1972). A distinct RNA polymerase activity, synthesizing 5.5 s, 5 s and 4 s RNA in nuclei from adenovirus 2-infected HeLa cells. J. Mol. Biol. 70, 435–450.

Radman-Livaja, M., Verzijlbergen, K.F., Weiner, A., van Welsem, T., Friedman, N., Rando, O.J., and van Leeuwen, F. (2011). Patterns and Mechanisms of Ancestral Histone Protein Inheritance in Budding Yeast. PLoS Biol 9, e1001075.

Ray-Gallet, D., Quivy, J.-P., Scamps, C., Martini, E.M.-D., Lipinski, M., and Almouzni, G. (2002). HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol. Cell 9, 1091–1100.

Ray-Gallet, D., Woolfe, A., Vassias, I., Pellentz, C., Lacoste, N., Puri, A., Schultz, D.C., Pchelintsev, N.A., Adams, P.D., Jansen, L.E.T., et al. (2011). Dynamics of histone H3 deposition in vivo reveal a

Page 164: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 3 – H3.1 and H3.3 Turnover

144

nucleosome gap-filling mechanism for H3.3 to maintain chromatin integrity. Mol. Cell 44, 928–941.

Sexton, T., Umlauf, D., Kurukuti, S., and Fraser, P. (2007). The role of transcription factories in large-scale structure and dynamics of interphase chromatin. Semin. Cell Dev. Biol. 18, 691–697.

Shibahara, K., and Stillman, B. (1999). Replication-dependent marking of DNA by PCNA facilitates CAF-1-coupled inheritance of chromatin. Cell 96, 575–585.

Simon, J.A., and Kingston, R.E. (2013). Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol. Cell 49, 808–824.

Studitsky, V.M., Clark, D.J., and Felsenfeld, G. (1994). A histone octamer can step around a transcribing polymerase without leaving the template. Cell 76, 371–382.

Studitsky, V.M., Kassavetis, G.A., Geiduschek, E.P., and Felsenfeld, G. (1997). Mechanism of transcription through the nucleosome by eukaryotic RNA polymerase. Science 278, 1960–1963.

Vafa, O., and Sullivan, K.F. (1997). Chromatin containing CENP-A and α-satellite DNA is a major component of the inner kinetochore plate. Curr. Biol. 7, 897–900.

Waddington, C.H. (2011). The Epigenotype. Int. J. Epidemiol.

Warburton, P.E., Greig, G.M., Haaf, T., and Willard, H.F. (1991). PCR amplification of chromosome-specific alpha satellite DNA: definition of centromeric STS markers and polymorphic analysis. Genomics 11, 324–333.

Weber, C.M., Henikoff, J.G., and Henikoff, S. (2010). H2A.Z nucleosomes enriched over active genes are homotypic. Nat Struct Mol Biol 17, 1500–1507.

Yankulov, K., Yamashita, K., Roy, R., Egly, J.-M., and Bentley, D.L. (1995). The Transcriptional Elongation Inhibitor 5,6-Dichloro-1-β-D-ribofuranosylbenzimidazole Inhibits Transcription Factor IIH-associated Protein Kinase. J. Biol. Chem. 270, 23922–23925.

Page 165: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

145

Page 166: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

146

Page 167: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

147

Chapter 4 – General Discussion

Page 168: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

148

Page 169: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

149

The inheritance of cellular gene expression profiles and

associated phenotypes is critical for the generation and stability of

different cell types. This epigenetic inheritance is carried by non-DNA

sequence based factors across mitotic divisions. Although several

factors have been identified that play a role in this process, their

mechanism to provide memory is in many cases unknown. We have

proposed that mechanisms of epigenetic inheritance should, in the

most parsimonious view, fulfill with the following aspects: stability, self-

templated duplication and cell cycle coupling (Gómez-Rodríguez and

Jansen, 2013). Several epigenetic markers are chromatin based,

implicating nucleosomes, that are central to chromatin, as important

players in epigenetic inheritance but the mechanistic basis is not clear.

Histone H3 variants have been involved in the transmission of active

and repressive gene states (Bannister et al., 2001; Hathaway et al.,

2012; Lachner et al., 2001; Ng and Gurdon, 2008) which raises an

important question: Are histone H3 variants inherited across mitotic

cell divisions? The work presented here provides evidence to answer

this question.

In chapter 2, we introduced TimeChIP, an assay that allows

capturing histone dynamics (Figure 2.3) by covalent pulse labeling of

uniquely tagged histone variants. The main advantages of TimeChIP

are the distinction of old (ancestral) and new pools of histones, the

ability to determine dynamics at a timescale of hours to days, and the

high spatial resolution. Importantly, this allows for the determination of

histone turnover at specific loci. In chapter 3, we show for the first time

that a population of ancestral H3.1 and H3.3 histones is stably retained

across the cell cycle in a human cell line. In this chapter, I will discuss

several hypotheses and predictions that follow from the results

presented in this work.

Page 170: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

150

Can qualitative differences between nucleosomes

result in differential stability?

Our initial results in chapter 2 show that a subpopulation of

CENP-A assembles outside centromeres that displays a faster

turnover in comparison to centromeric pools (Figure 2.7 & 2.9). One

explanation for differential turnover of CENP-A at different loci may be

differential rates of transcription rates. rDNA loci are transcribed at a

much higher rate compared to centromeric loci.

Another hypothesis is that CENP-A nucleosomes are

intrinsically different between different loci. Purification of CENP-A

nucleosomes from human cells indicated that CENP-A containing

nucleosomes are enriched in the histone H2A.Z variant (Foltz 2006).

H2A.Z is involved in transcription regulation, DNA repair,

heterochromatin formation, chromosome segregation and mitosis, but

it does not exhibit a uniform localization within the genome

(Guillemette and Gaudreau, 2006; Kalocsay et al., 2009; Meneghini et

al., 2003; Zlatanova et al., 2009) A particular feature of H2A.Z is that it

is broadly but non-uniformly distributed throughout the chromosomes

and can be actively and selectively removed at specific loci (Hardy and

Robert, 2010). Bönisch et al., suggest that H2A.Z localization is due to

difference of the stability between homo and heterotypic H2A.Z

nucleosomes (Bönisch and Hake, 2012).

Based on these findings, a hypothesis that could explain the

incorporation of unstable CENP-A nucleosomes outside the

centromere locus is the formation of CENP-A/H2A.Z double variant

nucleosomes that may be unstable in a manner analogous to the

previously reported destabilization of H3.3/H2A.Z double variant

nucleosomes (Jin et al., 2009). As an alternative, Foltz et al, also found

CENP-A/H3 hybrid nucleosomes. Possibly, these heterotypic

Page 171: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

151

nucleosomes provide a different stability. However it is not known

whether the CENP-A/H2A.Z or CENP-A/H3 hybrids represent the non

centromeric pool. To explore these hypotheses a TimeChIP

experiment could be carried out to isolate CENP-A (chapter 2) followed

by a reChIP for H2A.Z and/or H3. This will determine where such

hybrid nucleosomes are located and whether they correlate with high

CENP-A turnover, e.g. at the 5SrDNA loci.

In addition to CENP-A, our results indicate that H3.1 and H3.3

also turnover at different rates. However, in this case differential

turnover occurs at the same locus as we observe turnover in a

biphasic manner (Figure 3.1 & 3.3). As discussed above, nucleosomes

carrying both H3.3 and H2A.Z variants have been shown to be highly

unstable (Jin et al., 2009). The crystal structure of H2A.Z nucleosomes

indicates a subtle destabilization of the interaction interface between

the H2A.Z docking domain and H3 (Suto et al., 2000).

One possible explanation of nucleosomes with different

stabilities at specific loci (e.g. at promoter versus the gene body) may

be the presence or absence of the H2A.Z variant histone. To

determine this I would propose to conduct a H2A-SNAP and H2A.Z-

SNAP TimeChIP Pulse-Chase experiment. If rapid nucleosome

turnover is a consequence of H2A.Z presence we would expect H2A.Z

turnover to be fast and largely completed during the first 3 hours. In

contrast H2A-SNAP would be expected to turnover at a slower but

constant rate reflecting the slow phase we observe in our experiments

in Figures 3.1 and 3.3. In addition, one could directly determine the

presence of H2A.Z or H2A in young (Pulsed) versus old (chased)

SNAP-tagged H3.1 and H3.3 by a re-ChIP experiment of H3.1/H3.3-

SNAP followed by H2A.Z ChIP or by mass spectrometry in young

(Pulse) versus old (chased) SNAP-tagged H3.1 and H3.3.

Page 172: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

152

Transcription drives histone turnover

During transcription elongation RNA polymerase II must

overcome nucleosome barriers. Consequently, displacement of

nucleosomes is expected to occur to some extend during transcription.

Surprisingly however, upon induction of transcription the turnover of

H3.1 and H3.3 did not increase (Figure 3.7). Histone chaperones and

chromatin remodelers are involved in the disassembly/reassembly of

nucleosomes during transcription. FACT is proposed to be involved in

exchanging histones during transcription elongation, specifically

H2A/H2B dimers (Belotserkovskaya et al., 2003). In addition,

deposition of H3.3 by FACT has been reported in Drosophila

(Nakayama et al., 2007). Highly transcribed loci show an enrichment of

FACT and H3.3 indicating a high degree of histone turnover (Aida et

al., 2013).

How can we reconcile transcription dependent histone

replacement with the stable nucleosome retention we observe for a

subset of nucleosomes upon transcription induction? As outlined

above, one model would involve different retention rates of histones

depending on intrinsic nucleosome differences. Possibly, chromatin

remodelers turn over selected dynamic nucleosomes leaving intact a

more stably subset. However, while chromatin remodelers have a clear

role in histone dynamics during transcription elongation, it is unknown

whether they selectively discriminate between ancestral and newly

synthesized histones to be deposited. Using a conditional reporter to

induce or repress transcription in cells expressing H3.3-SNAP

combined with RNAi, one might be able to determine the contribution

of specific chaperones to the retention of recently assembled

nucleosomes versus older ones. This may establish whether, for

instance, FACT plays a role in disassembly/reassembly of ancestral

histones to support maintenance of epigenetic information.

Page 173: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

153

Importantly, the same conditional shut off reporter system in

combination with TimeChIP, will allow to test our prediction that

histone turnover diminishes when transcription does not occur. Instead

of using global inhibitors of transcription (as discussed below) such an

experiment would allow for a more subtle manipulation of local

transcription.

Our experiment thus far using small molecule inhibitors of

transcription have produced puzzling results. While a slowing down of

histone turnover can be observed upon transcription inhibition, this

increase in retention appears to be global and not correlated with sites

of transcription (Figure 3.9 & 3.10). One possibility explanation would

be that these inhibitors of RNA polymerase II induce a non-specific

cellular stress that indirectly affect chromatin dynamics.

Alternatively, the inhibition of transcription by DRB and α-

Amanitin leads to the migration of large scale chromatin loops distal of

transcription factories. Chromatin hub structures are formed prior to

entering a transcription factory (Mitchell and Fraser, 2008). Likely,

those chromatin hubs are compacted or migrate to the nuclear lamina

upon transcription inhibition as has been proposed to occur as a

mechanism of gene regulation during development (Peric-Hupkes et

al., 2010). This in turn may lead to a change in nucleosome dynamics

even at sites distal to active genes. An approach to define whether

transcription inhibition leads to high retention of histones by formation

of chromatin hubs will be to obtain high coverage TimeChIP-seq data

to determine whether changes in histone dynamics upon transcription

inhibition occur in large genome segments. Another, perhaps more

challenging approach would be to combine the use of 3C

(chromosome conformation capture) technology (de Wit and de Laat,

2012) in parallel with TimeChIP to dissect this relationship.

Page 174: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

154

Figure 4.1 Model for differential turnover of histones at active

genes. We propose that during transcription, a population of histones

(either H3.1 or H3.3) is locally retained (green) whereas a proportion of

histones is lost and replaced by nascent histones that are selectively

turned over at higher rates (grey)

The role of histones as epigenetic markers

How does histone turnover affect maintenance of histone

modifications? It has been proposed that histone modification patterns

of newly synthesized histones are loaded based on the template of

histone modifications present on the neighboring parental nucleosome

(Annunziato, 2005; Bannister et al., 2001; Felsenfeld and Groudine,

2003; Lachner et al., 2001). This proposal is based, in part, on the

studies that have shown global stability of nucleosomes e.g. during

DNA replication (Ishimi et al., 1991; Jackson, 1990; Katan-Khaykovich

and Struhl, 2011; Xu et al., 2010). Now, we are providing evidence for

stable retention of a population H3.1 and H3.3 at specific loci

Page 175: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

155

throughout a cell cycle, and at least in the case of H3.1, the retention is

local. In support of histone retention during DNA replication that we

described, Groth et al. have shown that the histone chaperone Asf1 in

association with MCM2-7 helicase, through the Asf1-(H3-H4)-MCM

intermediate, functions as a histone acceptor and donor of both old

and new nucleosomes to assemble on daughter DNA strands. They

proposed that the Asf1-(H3-H4)-MCM complex transfers parental

nucleosomes. Part of this argument is based on the fact that when

DNA synthesis is stalled, histone modifications H4K16Ac and

H3K9me3, that are not found on nascent histones, become enriched in

the complex, suggesting that during normal progression of the DNA

replication fork these parental nucleosomes transiently associate with

this complex (Groth et al., 2007). Recently, artificial induction of

H3K9me3 in vivo has been achieved with the so-called chromatin in

vivo assay CiA that involves local tethering of HP1 (Hathaway et al.,

2012). Using CiA Hathaway and colleges, have measured the kinetics

and maintenance of H3K9me3 through cell division following transient

HP1 tethering. They suggest that maintenance of H3K9me3 occurs by

symmetric propagation, where a neighboring nucleosome is marked on

average every 5-7 hours depending on the cell type and propose that

the inheritance of noncentromeric H3K9me3 domains involves a

combination of nucleation, local propagation and histone turnover

(Hathaway et al., 2012; Hodges and Crabtree, 2012). Similarly, earlier

work has proposed the transmission of H3K27me3 using a

heterologous reporter system (Hansen et al., 2008). In a cell line

expressing GAL4-EED under the control of a tetracycline regulated

promoter, EED-mediated inhibition of transcription was induced,

leading to the formation and recruitment of the PRC2 (EED-EZH2-

SUZ12) complex to the promoter. Upon removal of tetracycline,

propagation of H3K27me3 at the TSS and downstream sequences as

well as endogenous PRC2 was detected up to four cell divisions

Page 176: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

156

(Hansen et al., 2008). While transmission of the silent state was

observed these studies do not proof that maintenance of the chromatin

state is mediated solely through histone stability and methyltransferase

activities. Nevertheless, these relatively slow propagation rates are

consistent with the multi-hour retention we observe making it possible

that slow turnover and slow propagation of marks may be sufficient to

heritably maintain the epigenetic state mediated by the mark.

Recent analysis of nucleosome dynamics questioned the role

of histones as players to transmit epigenetic information, because half-

lives of histones at active genes were measured to be ~1-1.5 hours,

(Deal et al., 2010). We describe for the first time a sub-population of

H3.1 that is locally retained for many hours at active genes. An

intriguing question, to be addressed in future efforts, will be whether

the fraction of H3.1 retained carries ancestral histone modifications.

Does the fraction of stable H3.1 correspond to the nucleation factor for

the subsequent propagation of histone modifications like H3K9me3

and H3K27me3? One approach to define whether parental histones

preferentially carry specific histone modifications will be to quantify

parental histones by TimeChIP followed by the measurement of

histone modifications with stable isotope labeling by amino acid in cell

culture (SILAC). Additionally, site specific analysis would allow

determining whether histones turn over more slowly at a locus targeted

for silencing of gene expression. Using a conditional shut-off reporter

e.g. the one developed by Hansen et al., (Hansen et al., 2008) one

could determine the enrichment of old histones at the silenced locus

followed by re-ChIP for H3K27me3 to determine whether this

modification is enriched in old (chased) SNAP-tagged histones H3.1 or

H3.3.

In closing, in the future it will be important to integrate histone

dynamics (retention/replenishment) with the direct transmission of

Page 177: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

157

histone modifications during transcription and silencing. Understanding

the relative contributions of turnover and propagation of both histones

and their modification is key to establish the contribution of histone

modifications to epigenetic memory. Finally, according to

Waddington’s epigenetic landscape, repressed markers are more

stable than active ones (Ferrell, 2012). In light of this, one may have to

consider how a histone modification that accompanies transcription is

propagated and turned over (Clayton et al., 2006). An example is

acetylation, where instead of a stable steady state, an equilibrium of

acetylation/deacetylation cycles define the chromatin state

(Verzijlbergen et al., 2011). This represents a challenge that requires

the development of techniques with high temporal and spatial

resolution to obtain a more complete understanding of inheritance of

active chromatin states.

Page 178: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

158

References

Aida, M., Hamad, N., Stanlie, A., Begum, N.A., and Honjo, T. (2013). Accumulation of the FACT complex, as well as histone H3.3, serves as a target marker for somatic hypermutation. Proc. Natl. Acad. Sci. 110, 7784–7789.

Annunziato, A.T. (2005). Split Decision: What Happens to Nucleosomes during DNA Replication? J. Biol. Chem. 280, 12065–12068.

Bannister, A.J., Zegerman, P., Partridge, J.F., Miska, E.A., Thomas, J.O., Allshire, R.C., and Kouzarides, T. (2001). Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain. Nature 410, 120–124.

Belotserkovskaya, R., Oh, S., Bondarenko, V.A., Orphanides, G., Studitsky, V.M., and Reinberg, D. (2003). FACT facilitates transcription-dependent nucleosome alteration. Science 301, 1090–1093.

Bönisch, C., and Hake, S.B. (2012). Histone H2A variants in nucleosomes and chromatin: more or less stable? Nucleic Acids Res.

Clayton, A.L., Hazzalin, C.A., and Mahadevan, L.C. (2006). Enhanced Histone Acetylation and Transcription: A Dynamic Perspective. Mol. Cell 23, 289–296.

Deal, R.B., Henikoff, J.G., and Henikoff, S. (2010). Genome-Wide Kinetics of Nucleosome Turnover Determined by Metabolic Labeling of Histones. Science 328, 1161–1164.

Felsenfeld, G., and Groudine, M. (2003). Controlling the double helix. Nature 421, 448–453.

Ferrell, J.E., Jr (2012). Bistability, bifurcations, and Waddington’s epigenetic landscape. Curr. Biol. CB 22, R458–466.

Gómez-Rodríguez, M., and Jansen, L.E. (2013). Basic properties of epigenetic systems: lessons from the centromere. Curr. Opin. Genet. Dev. 23, 219–227.

Groth, A., Corpet, A., Cook, A.J.L., Roche, D., Bartek, J., Lukas, J., and Almouzni, G. (2007). Regulation of Replication Fork Progression Through Histone Supply and Demand. Science 318, 1928–1931.

Page 179: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

159

Guillemette, B., and Gaudreau, L. (2006). Reuniting the contrasting functions of H2A.Z. Biochem. Cell Biol. Biochim. Biol. Cell. 84, 528–535.

Hansen, K.H., Bracken, A.P., Pasini, D., Dietrich, N., Gehani, S.S., Monrad, A., Rappsilber, J., Lerdrup, M., and Helin, K. (2008). A model for transmission of the H3K27me3 epigenetic mark. Nat. Cell Biol. 10, 1291–1300.

Hardy, S., and Robert, F. (2010). Random deposition of histone variants: A cellular mistake or a novel regulatory mechanism? Epigenetics Off. J. DNA Methylation Soc. 5, 368–372.

Hathaway, N.A., Bell, O., Hodges, C., Miller, E.L., Neel, D.S., and Crabtree, G.R. (2012). Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460.

Hodges, C., and Crabtree, G.R. (2012). Dynamics of inherently bounded histone modification domains. Proc. Natl. Acad. Sci. 109, 13296–13301.

Ishimi, Y., Sugasawa, K., Hanaoka, F., and Kikuchi, A. (1991). Replication of the simian virus 40 chromosome with purified proteins. J. Biol. Chem. 266, 16141–16148.

Jackson, V. (1990). In vivo studies on the dynamics of histone-DNA interaction: evidence for nucleosome dissolution during replication and transcription and a low level of dissolution independent of both. Biochemistry (Mosc.) 29, 719–731.

Jin, C., Zang, C., Wei, G., Cui, K., Peng, W., Zhao, K., and Felsenfeld, G. (2009). H3.3/H2A.Z double variant-containing nucleosomes mark “nucleosome-free regions” of active promoters and other regulatory regions. Nat. Genet. 41, 941–945.

Kalocsay, M., Hiller, N.J., and Jentsch, S. (2009). Chromosome-wide Rad51 spreading and SUMO-H2A.Z-dependent chromosome fixation in response to a persistent DNA double-strand break. Mol. Cell 33, 335–343.

Katan-Khaykovich, Y., and Struhl, K. (2011). Splitting of H3-H4 tetramers at transcriptionally active genes undergoing dynamic histone exchange. Proc. Natl. Acad. Sci. U. S. A. 108, 1296–1301.

Page 180: This dissertation was sponsored by Fundação para a Ciência ... Thesis Mariluz... · trabalho original por mim desenvolvido entre Fevereiro de 2009 e Abril de 2013 no laboratório

Chapter 4 – General Discussion

160

Lachner, M., O’Carroll, D., Rea, S., Mechtler, K., and Jenuwein, T. (2001). Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410, 116–120.

Meneghini, M.D., Wu, M., and Madhani, H.D. (2003). Conserved histone variant H2A.Z protects euchromatin from the ectopic spread of silent heterochromatin. Cell 112, 725–736.

Mitchell, J.A., and Fraser, P. (2008). Transcription factories are nuclear subcompartments that remain in the absence of transcription. Genes Dev. 22, 20–25.

Nakayama, T., Nishioka, K., Dong, Y.-X., Shimojima, T., and Hirose, S. (2007). Drosophila GAGA factor directs histone H3.3 replacement that prevents the heterochromatin spreading. Genes Dev. 21, 552–561.

Ng, R.K., and Gurdon, J.B. (2008). Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription. Nat. Cell Biol. 10, 102–109.

Peric-Hupkes, D., Meuleman, W., Pagie, L., Bruggeman, S.W.M., Solovei, I., Brugman, W., Gräf, S., Flicek, P., Kerkhoven, R.M., van Lohuizen, M., et al. (2010). Molecular maps of the reorganization of genome-nuclear lamina interactions during differentiation. Mol. Cell 38, 603–613.

Suto, R.K., Clarkson, M.J., Tremethick, D.J., and Luger, K. (2000). Crystal structure of a nucleosome core particle containing the variant histone H2A.Z. Nat. Struct. Biol. 7, 1121–1124.

Verzijlbergen, K.F., van Welsem, T., Sie, D., Lenstra, T.L., Turner, D.J., Holstege, F.C.P., Kerkhoven, R.M., and van Leeuwen, F. (2011). A barcode screen for epigenetic regulators reveals a role for the NuB4/HAT-B histone acetyltransferase complex in histone turnover. PLoS Genet. 7, e1002284.

De Wit, E., and de Laat, W. (2012). A decade of 3C technologies: insights into nuclear organization. Genes Dev. 26, 11–24.

Xu, M., Long, C., Chen, X., Huang, C., Chen, S., and Zhu, B. (2010). Partitioning of Histone H3-H4 Tetramers During DNA Replication–Dependent Chromatin Assembly. Science 328, 94–98.

Zlatanova, J., Bishop, T.C., Victor, J.-M., Jackson, V., and van Holde, K. (2009). The nucleosome family: dynamic and growing. Struct. Lond. Engl. 1993 17, 160–171.