Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges...

15
REVIEW Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel, 1 Francesca Palladino, 2 and Monika Jedrusik-Bode 1 * 1 Electron Microscopy Group 3 Epigenetics in C. elegans Group, Max Planck Institute for Biophysical Chemistry, Am Fabberg 11, 37077 Go ¨ ttingen, Germany 2 Laboratory of Molecular and Cellular Biology, University of Lyon, CNRS UMR5239, Ecole Normale Superieure, 69364 Lyon Cedex 07, France Received 15 November 2010; Revised 14 April 2011; Accepted 15 April 2011 Summary: Epigenetics is defined as the study of herit- able changes in gene expression that are not accompa- nied by changes in the DNA sequence. Epigenetic mechanisms include histone post-translational modifi- cations, histone variant incorporation, non-coding RNAs, and nucleosome remodeling and exchange. In addition, the functional compartmentalization of the nu- cleus also contributes to epigenetic regulation of gene expression. Studies on the molecular mechanisms underlying epigenetic phenomena and their biological function have relied on various model systems, includ- ing yeast, plants, flies, and cultured mammalian cells. Here we will expose the reader to the current under- standing of epigenetic regulation in the roundworm C. elegans. We will review recent models of nuclear or- ganization and its impact on gene expression, the bio- logical role of enzymes modifying core histones, and the function of chromatin-associated factors, with spe- cial emphasis on Polycomb (PcG) and Trithorax (Trx-G) group proteins. We will discuss how the C. elegans model has provided novel insight into mechanisms of epigenetic regulation as well as suggest directions for future research. genesis 49:647–661, 2011. V V C 2011 Wiley-Liss, Inc. Key words: core histones; post-translational modifications; chromatin; Hox; trithorax; PcG INTRODUCTION In metazoan development, cellular identity is estab- lished by tissue and lineage-specific transcription fac- tors acting in concert with epigenetic mechanisms. Dif- ferentiated cells also rely on epigenetic mechanisms in order to transmit specific gene expression patterns that distinguish the differentiated state through multiple cell divisions (Hemberger et al., 2009). The basic subunit of chromatin is the nucleosome, composed of the highly conserved core histones H2A, H2B, H3, and H4. Epigenetic mechanisms can modify this structure in several ways. Depending on the chro- mosomal context, core histones can be replaced by histone variants, including H3.3, H2A.Z, or H2A.X, each of which is associated with specialized functions (Jin and Felsenfeld, 2007; Talbert and Henikoff, 2010). The flexible N- and C-terminal tails of core histones are also subjected to a variety of post-translational modifications, including acetylation, methylation, phosphorylation, ubiquitination, sumoylation, ADP- ribosylation, and biotinylation. These modifications, alone or in combination, correlate with transcrip- tional repression or activation (Jenuwein and Allis, 2001). In addition, chromatin-associated factors such as Trithorax (Trx-G) and Polycomb group (PcG) pro- teins function to maintain cellular memory of tran- scriptional states of developmentally important genes through many rounds of cell division. Transcriptional regulation by PcG and Trx-G group proteins is achieved through both histone methyltransferase activities of individual components, as well as specific * Correspondence to: Dr. Monika Jedrusik-Bode, Max Planck Institute for Biophysical Chemistry, Epigenetics in C. elegans Group, Am Fassberg 11, 37077 Go ¨ttingen, Germany. E-mail: [email protected] Contract grant sponsor: German National Funding Agency, Contract grant number: JE 505/1-3, Contract grant sponsor: Max Planck Society Published online 2 May 2011 in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/dvg.20762 ' 2011 Wiley-Liss, Inc. genesis 49:647–661 (2011)

Transcript of Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges...

Page 1: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

REVIEW

Epigenetics in C. elegans: Facts and Challenges

Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1*

1Electron Microscopy Group 3 Epigenetics in C. elegans Group, Max Planck Institute for Biophysical Chemistry,Am Fabberg 11, 37077 Gottingen, Germany

2Laboratory of Molecular and Cellular Biology, University of Lyon, CNRS UMR5239,Ecole Normale Superieure, 69364 Lyon Cedex 07, France

Received 15 November 2010; Revised 14 April 2011; Accepted 15 April 2011

Summary: Epigenetics is defined as the study of herit-able changes in gene expression that are not accompa-nied by changes in the DNA sequence. Epigeneticmechanisms include histone post-translational modifi-cations, histone variant incorporation, non-codingRNAs, and nucleosome remodeling and exchange. Inaddition, the functional compartmentalization of the nu-cleus also contributes to epigenetic regulation of geneexpression. Studies on the molecular mechanismsunderlying epigenetic phenomena and their biologicalfunction have relied on various model systems, includ-ing yeast, plants, flies, and cultured mammalian cells.Here we will expose the reader to the current under-standing of epigenetic regulation in the roundwormC. elegans. We will review recent models of nuclear or-ganization and its impact on gene expression, the bio-logical role of enzymes modifying core histones, andthe function of chromatin-associated factors, with spe-cial emphasis on Polycomb (PcG) and Trithorax (Trx-G)group proteins. We will discuss how the C. elegansmodel has provided novel insight into mechanisms ofepigenetic regulation as well as suggest directions forfuture research. genesis 49:647–661, 2011. VVC 2011

Wiley-Liss, Inc.

Key words: core histones; post-translational modifications;chromatin; Hox; trithorax; PcG

INTRODUCTION

In metazoan development, cellular identity is estab-lished by tissue and lineage-specific transcription fac-tors acting in concert with epigenetic mechanisms. Dif-ferentiated cells also rely on epigenetic mechanisms inorder to transmit specific gene expression patterns that

distinguish the differentiated state through multiple celldivisions (Hemberger et al., 2009).

The basic subunit of chromatin is the nucleosome,composed of the highly conserved core histones H2A,H2B, H3, and H4. Epigenetic mechanisms can modifythis structure in several ways. Depending on the chro-mosomal context, core histones can be replaced byhistone variants, including H3.3, H2A.Z, or H2A.X,each of which is associated with specialized functions(Jin and Felsenfeld, 2007; Talbert and Henikoff, 2010).The flexible N- and C-terminal tails of core histonesare also subjected to a variety of post-translationalmodifications, including acetylation, methylation,phosphorylation, ubiquitination, sumoylation, ADP-ribosylation, and biotinylation. These modifications,alone or in combination, correlate with transcrip-tional repression or activation (Jenuwein and Allis,2001). In addition, chromatin-associated factors suchas Trithorax (Trx-G) and Polycomb group (PcG) pro-teins function to maintain cellular memory of tran-scriptional states of developmentally important genesthrough many rounds of cell division. Transcriptionalregulation by PcG and Trx-G group proteins isachieved through both histone methyltransferaseactivities of individual components, as well as specific

*Correspondence to: Dr. Monika Jedrusik-Bode, Max Planck Institute for

Biophysical Chemistry, Epigenetics in C. elegans Group, Am Fassberg 11,

37077 Gottingen, Germany.

E-mail: [email protected]

Contract grant sponsor: German National Funding Agency, Contract

grant number: JE 505/1-3, Contract grant sponsor: Max Planck Society

Published online 2 May 2011 in

Wiley Online Library (wileyonlinelibrary.com).

DOI: 10.1002/dvg.20762

' 2011 Wiley-Liss, Inc. genesis 49:647–661 (2011)

Page 2: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

binding to and interpretation of these histone marks.PcG and Trx-G group proteins are also able to recruitadditional proteins that greatly expand their reper-toire of covalent histone modifications (Sparmann andvan Lohuizen, 2006).

Histone post-translational modifications are carriedout by a variety of complexes containing histone-modi-fying enzymes. Histone methyltransferases (HMTs), his-tone demethylases (HDMs), histone acetyltransferases(HATs), histone deacetylases (HDACs), and histone ki-nases not only modify histone tails, but also act asrecruiters of protein complexes and nucleosomeremodelers (Jenuwein and Allis, 2001). Histone tailmodifications are in turn associated with specific chro-matin-binding proteins or protein complexes (readers)that modify chromatin structure. Small non-codingRNAs, nucleosome exchange and remodeling by ATP-dependent complexes, and histone variant incorpora-tion, are additional mechanisms that can alter thedegree of chromatin compaction (Jenuwein and Allis,2001). In addition, nuclear organization has recentlyemerged as another important player in epigenetic regu-lation (Luger and Hansen, 2005).

This review summarizes recent models of nuclearcompartmentalization in the nematode C. elegans, anddescribes the different developmental roles attributedto histone-modifying enzymes, focusing on the initiationand maintenance of epigenetic memory. In particular,we will describe the molecular players involved in PcGand Trx-G regulation of Hox gene expression and lifespan determination.

Compartmentalization: from NuclearGenome to Epigenome

Higher-order chromatin organization results frominterplay between the compacted nuclear DNAsequence, DNA methylation, histone modifications,small RNAs, and chromatin remodeling. Nucleararchitecture is dynamically dependent on (1) celltype, (2) developmental timing (embryo/adult), (3)environmental challenges, and (4) changes in tran-scriptional activities correlated with the repositioningof genes from active to repressive nuclear compart-ments. In vivo observations have shown that in thecell nucleus, transcriptionally active DNA sequencesare organized into regions called euchromatin, andtranscriptionally silent DNA sequences in regionsknown as heterochromatin.

In C. elegans, heterochromatin can be identified byelectron microscopy as specific regions enriched in re-pressive marks: di- and tri-methylated core histone H3at lysine 9, and 27 (H3K9me2/3 and H3K27me3) (Fig.1a,b). In contrast, euchromatin is marked by core his-tone H3 di-methylated at lysine 4 (H3K4me2) (Fig. 1c).Therefore, C. elegans possesses an epigenetic landscape

very similar to that of flies and mammals. In C. elegans,transcriptionally silent regions marked by the repressivemark H3K9me3 have been found at the end of chromo-somes, including telomeres (Gerstein et al., 2010; Liuet al., 2011; Wirth et al., 2009). In many organisms,telomeric heterochromatin participates in gene repres-sion and recombination. Actively transcribed genesmoved to telomeric regions are reversibly silenced, aphenomenon termed telomere position effect (TPE)(Aparicio et al., 1991) or position-effect variegation(PEV) (Henikoff, 1990). Curiously, although C. eleganspossesses telomeric heterochromatin, TPE has not beendescribed so far (Wirth et al., 2009). The importance oftelomeric chromatin structure in telomere stability ishighlighted by the fact that its disruption in mammaliancells alters telomere length and increases aberrantrecombination events (Schoeftner and Blasco, 2009).Whether a similar function exists in C. elegans remainsto be established.

Additional findings have suggested a conserved rolefor RNAi in the formation of heterochromatin. In fissionyeast, endogenous siRNAs (small interfering RNAs) arespecifically implicated in RNA-induced transcriptionalsilencing (RITS), and RITS activity is required forH3K9me2 accumulation on repetitive sequences, includ-ing centromeric repeats (Hall et al., 2002; Moazed, 2009;Volpe et al., 2002; Zaratiegui et al., 2007). Mutations ofany component of the RNAi machinery results in defectsin chromosome segregation due to defects in centromerefunction. Although C. elegans chromosomes are holocen-tric and lack pericentromeric repeats, recent studiesidentified CSR-1 (a Piwi/PAZ/Argonaute protein), EKL-1(a Tudor domain protein), and DRH-3 (a DEAH/D-boxhelicase), as regulators of meiotic H3K9me2 accumula-tion on unpaired chromosomes during C. elegans meio-sis, implicating RNAi in the regulation of meiotic chroma-tin (She et al., 2009).

Several genes in the RNAi-pathway have also beenshown to be involved in RNAi-induced Transcrip-tional Gene Silencing (TGS) to influence heterochro-matin formation and silence the expression of repeti-tive transgenes in both the C. elegans germ line andsoma (Grishok et al., 2005a,b; Robert et al., 2005;Sijen and Plasterk, 2003). Repetitive transgenes in theC. elegans germ line are effectively silenced through achromatin-related mechanism dependent on the MES(maternal effect sterility) group proteins, the histonedeacetylase SIR-2.1 (sirtuin-2), the HP1-like proteinHPL-2, and linker histone HIS-24 (see Fig. 2) (Couteauet al., 2002; Jedrusik and Schulze, 2001, 2003; Kellyand Fire, 1998). Interestingly, all of these proteins areassociated with heterochromatin associated marks:MES-2 deposits H3K27me3, SIR-2.1 deacetylatesH3K9, HPL-2 recognizes H3K9me2/3, and HIS-24binds H3K27me3 (Bender et al., 2004; Couteau et al.,2002; Wirth et al., 2009). Although worms do not

648 WENZEL ET AL.

Page 3: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

encode a conventional DNA methyltransferase tosilence DNA repeats, as observed in plants and mam-mals (Bird, 2002; Simpson et al., 1986), the presenceof H3K27me3 and H3K9me2 on repetitive transgenesmay play a function similar to DNA methylation inthis context.

To probe the existence of nuclear compartments dur-ing C. elegans development, the behavior of stably inte-grated fluorescent reporter constructs was recently ana-lyzed through live imaging in embryos and larvae (Mei-ster et al., 2010). It was shown that in embryos, largerepeat-induced heterochromatic arrays are associatedwith the nuclear envelope, while arrays carrying tissue-specific promoters are decompacted and relocated tothe nuclear lumen when activated in specific cell types(Meister et al., 2010). These findings suggest the pres-

ence of a nuclear compartmentalization similar to thatof mammals.

C. elegans Enzymes Modifying Core Histones andTheir Biological Function

Reversible modifications play a key role in definingchromatin states and in regulating transcription in a cell-specific manner. Whether the functions of individualmembers of chromatin-modifying complexes are uniqueor redundant, and to what extent the observed redun-dancy depends on temporal and spatial regulation oftheir expression, remains elusive (Jenuwein and Allis,2001). In C. elegans, many chromatin-modifying enzymes(modifiers) have been identified by genetic approaches,or through a combination of multiple sequence align-ments to visualize evolutionary relationships.

FIG. 1. Electron microscopy of somatic nuclei showing extended regions of heterochromatin (electron-dense and dispersed) as well asless-dense regions corresponding to euchromatin. Electron-dense regions are marked by H3K9me3 (a) and H3K27me3 (b) (labeling with10 nm gold) whereas less-dense regions are labeled with H3K4me2 (c) euchromatic marks. Eu-euchromatin, he- heterochromatin, nu-nucle-olus and env-envelope. Scale bar, 500 nm.

649EPIGENETICS IN C. elegans

Page 4: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

Tables 1 and 2 list the major chromatin-modifyingenzymes identified in C. elegans and summarize their bi-ological function in conserved multiprotein complexes.As shown, most enzymes not only have specificity fortarget residues, but also differ in tissue-specificity anddevelopmental requirements, reflecting the dynamic na-ture of histone modifications. Nonetheless, althoughthe function of histone modifying enzymes in C. elegans

development has become the focus of intense studies,we still know very little about the mechanisms responsi-ble for the establishment and maintenance of specificchromatin marks, and their recognition.

During C. elegans development, important roles forchromatin modifiers have been identified in vulval cell-fate specification, lifespan determination, genome sta-bility, and embryonic, germline and foregut develop-ment (Table 1). The C. elegans genome contains 38genes predicted to encode histone lysine methyltrans-ferase (HMT) activity, based on the presence of a con-served SET domain. The first of these genes to be identi-fied through a genetic screen were mes-2 and mes-4

(Capowski et al., 1991; Garvin et al., 1998; Holdemanet al., 1998). MES-2 is the ortholog of Drosophila Poly-comb group protein Enhancer of zeste E(z), while MES-4 is a nuclear SET domain protein related to mouseNSD1 (nuclear receptor binding SET-Domain 1) (Benderet al., 2004, 2007; Holdeman et al., 1998). MES-2 physi-cally interacts with both MES-6, homologue of the Pcgroup protein Extra sex combs (Esc), and MES-3 (anovel protein), in a repressive complex with H3K27

HMT activity similar to mammalian and fly E(z)/ESCcomplexes (Bender, 2004; Paulsen et al., 1995; Xuet al., 2001). MES deficiency results in maternal-effectsterility, derepression of high copy transgenes in thegermline, and specific loss of the H3K27me3 hetero-chromatin mark from the X chromosome (Bender et al.,2004, 2006; Fong et al., 2002). In addition, the absenceof MES activity suppresses the synthetic multivulva(synMuv) phenotype associated with inactivation of alarge group of histone modifying enzymes which actredundantly in vulval cell fate specification (Cui et al.,2004, 2006b; see below).

The C. elegans vulva represents a simple system inwhich to study how chromatin factors influence spe-cific developmental pathways (Ceol and Horvitz, 2004;Cui et al., 2006a,b; Lu and Horvitz, 1998; Sternebrg andHorvitz, 1991). synMuv genes fall into three classesencoding chromatin associated factors that act redun-dantly for vulval cell fate specification. Simultaneousinactivation of synMuv genes from any two differentclasses leads to ectopic EGF/RTK/Ras signaling and theformation of ectopic vulvae (the Multivulva phenotype).synMuv genes include the homologue of mammalianclass I histone deacetylases (HDA-1), histone methyl-transferases (MET-2; MET-1), and homologues of the evo-lutionarily conserved NuRD/Mi-2 nucleosome remodel-ing and histone deacetylase complex (LET-418/Mi-2,LIN-53/RbAp48, MEP-1, and HDA1/HDAC-1; Andersenand Horvitz, 2007; Dufourcq et al., 2002; Lu and Hor-vitz, 1998; Poulin et al., 2005; Unhavaithaya et al.,

FIG. 2. Difference between heterochromatic and euchromatic domains. Extrachromosomal arrays consisting of tandem DNA repeats aremarked by repressive histone methylation marks (H3K27me3, H3K9me2/3) and silenced by the HP1-like protein HPL-2 and SIR-2.1/SIRT1(sirtuin), whereas euchromatic domains are stained with active histone methylation marks and recognized by transcription factors or remod-eling complexes.

650 WENZEL ET AL.

Page 5: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

Table

1MajorChromatin-M

odifyingEnzymesIdentifiedin

C.elegansandTheirBiologicalF

unction

Classofhistone

modifyingenzymes

Nameofprotein

Mutantphenotype;function

References

HMT

MES-2/EzH

2(H3K27me3)

maternalsterile;requiredforchromatinorganisation

inthegerm

lineandX-chromosomesilencing

Holdemannetal.,

1998;Xuetal.,

2001;

Benderetal.,

2004

MES-4/N

SD1(H3K36me2)

maternalsterile;associateswithautosomalc

hromatin,

functionsin

epigeneticmemory

ofgerm

cells

Fongetal.,

2002;Benderetal.,

2006;

Rechtsteineretal.,

2010

MET-1/Set2pyeast(H3K9me3,

H3K36me3)

synMuv;

inhibitionofvulvalc

ellfate

AndersenandHorvitz,

2007

MET-2/SETDB1(H3K9me3,

H3K36me3)

synMuv;

inhibitionofvulvalc

ellfate

AndersenandHorvitz,

2007;

Bessleretal.,

2010.

BLMP-1/S

ETdomain-containingortholog

small,protrudingvulva(pvl)

Ceronetal.,

2007

SET-1/S

ETdomain-containingortholog

embryonic

andlarvallethal

AndersenandHorvitz,

2007

SET-3/S

MYD4,SET-4/S

UV420H1,SUV420H2,

SET-10,-14,-18,-30/SMYD1,2,3,

SET-11/EHMT1,TTLL-12/TTLL12,

SET-17/PRDM11,SET-27/SETD3,

SET-29/S

ETD4,SET-5,-6,-8,-9,-12,-13,-

15,-19,-20,-21,-24,-26,-28,-31,-32

noobviousphenotypes

AndersenandHorvitz,

2007

SET-2(H3K4me2)

slow

growth,mortalg

erm

line;regulateslifespan

XuandStrome2001;Sim

onetetal.,

2007;

Greeretal.,

2010;Xiaoetal.,

2011

SET-16/MLL(H3K4me3)

vulvald

evelopment;regulatesexpressionof

Hoxgenelin-39

Fisheretal.,2010

SET-23/SETMAR

embryonic

lethal

AndersenandHorvitz,

2007

SET-25/S

ETdomain-containingprotein

requiredfornorm

ala

xonalg

uidance,protects

genomeagainstmutations

Pothofetal.,

2003

WDR-5

(SWD-3

orTA

G-125)/WDR-5

(H3K4me3)

regulateslifespan;mortalg

erm

linephenotype,

antagonizessynMuvtranscriptionalrepressors

Fisheretal.,2010;Sim

onetetal.,

2007;

Greeretal.,

2010

ASH-2/ASH2fly(H3K4me3)

embryonic

lethalitycharacterize

dbydefects

incortical

activityandspindle

rocking;regulateslifespan

Fisheretal.,2010;Greeretal.,

2010

RBBP-5

(F21H12.1)

n/d

Fisheretal.,2010

HDM

JMJD2(forH3K9me3andH3K36me3)

regulateslifespan,increasedgerm

cellapoptosis,

activationofDNA-damagecheckpoint

Whetstineetal.,

2006

UTX-1/U

TX-1/JmjC

(H3K27me3)

vulvald

evelopment,cellcycle

arrest;regulates

expressionofHoxgenelin-39

Fisheretal.,2010;Wangetal.,

2010

TAG-279/JMJD3(H3K27me3)

abnorm

alg

onaddevelopment

Aggeretal.,

2007

C29F7.6/JMJD3

n/d

Aggeretal.,

2007

F29B9.2/JmjC

(H3K9me2and

H3K27me2)

regulateslifespan,dauerform

ation

Lin

etal.,

2010

F23D12.5;C06H2.3;F23D12.5;

F43G6.6;T26A5.5;T28F2.4;

C27F2.5;Y48B6A.11/JmjC

n/d

Aggeretal.,

2007;Klose

etal.,2006

SPR-5/LSD-1

(H3K4me2)

mortalg

erm

linephenotype

Katz

etal.,

2009

RBR-2/JARID1A(H3K4me3)

vulvald

evelopment;regulateslifespan

Christiansenetal.,

2007;Greeretal.,2010

HAT

MYS-1/TIP-60

syntheticslow

growth

phenotype;negativelyregulates

expressionofgenesrequiredforvulvalinduction

Ceola

ndHorvitz,

2004

LSY-12/M

YS-3

vulvald

evelopment,locomotion,egg-laying,fecundity

Sarinetal.,

2007

NuA4/M

YS-4

regulatesRassignalingandspecifiescellfates

Ceola

ndHorvitz,

2004

CBP-1/C

BP/p300

requiredduringembryogenesisfordifferentiation

ofallnon-neuronalsomaticcelltypes

Eastburn

DJ,HanM,2005

651EPIGENETICS IN C. elegans

Page 6: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

2002; von Zalewsky et al., 2000). HDA-1 is sumoylated inC. elegans, and both SUMO and the E2SUMO ligase UBC9are also members of the synMuv group, suggesting thatSUMOylation may regulate the activity of the NuRD/Mi-2complex in vulval cell fate specification (Poulin et al.,2005). Additionally, HDA-1 physically interacts with TRA-4, the C. elegans homolog of the human proto-oncopro-tein and transcriptional repressor PLZF, to promotefemale development by repressing the transcription ofmale-specific genes (Grote and Conradt, 2006). Interest-ingly, in these studies TRA-4 was also found to act as asynMuv gene to influence vulval cell fate specification.

Deletion of many synMuv genes results in tempera-ture sensitive developmental abnormalities, suggestingthat a temperature sensitive process is involved in chro-matin remodeling (Cui et al., 2006b; Petrella et al.,2011). For example, the C. elegans HP1 homolog hpl-2,a synMuv gene at permissive temperatures, shows ahighly penetrant multivulval (Muv) phenotype and ste-rility at 258C, the non-permissive temperature (Cous-tham et al., 2006; Couteau et al., 2002).

Until recently, it was unclear whether enzymes antag-onizing histone methylation are present in C. elegans.

The first histone demethylases (HDM) to be describedwere SPR-5, ortholog of the human LSD1 H3K4 demethyl-ase, and UTX-1, homolog of the mammalian Jumonji-C(JmjC) domain H3K27me2/3 demethylase UTX (Table 1)(Agger et al., 2007; Katz et al., 2009). Histone demethy-lation catalyzed by LSD1 relies on an oxidative reactionin which flavin is a cofactor, while JmjC-domain contain-ing enzymes require Fe(II) and a-ketoglutarate as cofac-tors (Klose et al., 2006; Lee et al., 2005; Metzger et al.,2005; Shi et al., 2004; Tsukada et al., 2006). JmjC-domain enzymes can remove all three histone lysine-methylation states, whereas LSD-1 removes only mono-and dimethyl lysines.

spr-5 loss of function suppresses the egg-laying defec-tive (egl) phenotype of mutations in sel-12/presenilinand derepresses expression of the second C. elegans

presenilin, hop-1. In binding assays and yeast two-hybrid experiments, SPR-5 interacts with SPR-1, the C.

elegans ortholog of CoREST (Eimer et al., 2002; Lakow-ski et al., 2003) (Table 2). A recent study demonstratedthat SPR-5 deficiency results in progressive sterility over20–30 generations (the mortal germline phenotype,mrt), suggesting an important function in the mainte-nance of germline immortality. This progressive sterilitycorrelates with the accumulation of H3K4me2 in germ-cell precursors, and the misregulation of spermatogene-sis genes (Katz et al., 2009). Mutation of another HDM,JMJD-2, causes increased germ cell apoptosis, consistentwith the activation of a DNA-damage checkpoint. Thisphenotype correlates with higher levels of H3K9me3 inthe germline and H3K36me3 on the X chromosome dur-ing meiotic prophase (Whetstine et al., 2006). Defi-ciency of the JMJD3 homolog F18E9.5 leads to defects in

Table

1Continued

Classofhistone

modifyingenzymes

Nameofprotein

Mutantphenotype;function

References

TAF-1-TAFII250(TBP)

embryonic

andlarvald

evelopment

Walkeretal.,

2004

MYS-2/M

OF

n/d

Shibata

etal.,

2010

TAG-235/H

AT-1

n/d

–GCN5-likesubfamily

n/d

Abo-D

alo

Betal.,

2004

HDAC

SIR

-2.1/SIRT1(H3K9)/classIII

requiredforgerm

linesilencing,longevity,

associateswithtelomeres

JedrusikandSchulze,2003;

Tissenbaum

andGuarente,2001;

Wirth

etal.,

2009

HDAC-11/classIV

decreasedgenomestability

Pothofetal.,

2003

HDA-1(G

ON-19)/H

DAC1/

RPD3yeastandfly/classI

embryonic

viability,gonadogenesisand

vulvald

evelopment

Dufourcqetal.,

2002

HDA-2/classI

genomestability

Lansetal.,

2010

HDA-3/H

D1/classI

affects

radiationsensitivity

Lansetal.,

2010

HDA-4/classII

locomotion,bodymorphology,andgrowth

Choie

tal.,

2002

HDA-6/classII

n/d

–F43G6.4/classII

n/d

HMT,histonemethyltransferase;HDM,histonedemethylase

;HAT,histoneacetyltransferase;HDAC,histonedeacetyltransferase.Annotationoftheproteinsfrom

C.elegansandtheir

human,flyoryeasthomologuesare

listed.

652 WENZEL ET AL.

Page 7: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

somatic gonad development (Agger et al., 2007), whileloss of function of rbr-2, encoding an ortholog of humanJARID1A, results in vulval specification defects associatedwith increased global levels of H3K4me3 in late larvalstages and adults (Christensen et al., 2007).

In mammals, the activity of different histone modify-ing enzymes is often functionally linked to reinforce theactivation or repression of gene expression. Severalexamples of this cross talk have been observed in C. ele-

gans, showing that similar mechanisms are implicatedin the dynamic regulation of chromatin structure in ver-tebrates and invertebrates. For example, the UTX-1H3K27 demethylase and the SET-16/MLL H3K4 methyl-transferase act in the same genetic pathway to attenuateLET-60 RAS signalling and regulate egl-5 Hox geneexpression (Fisher et al., 2010). In mammalian cells,UTX physically associates with MLL (Cho et al., 2007;Lee et al., 2007), suggesting that UTX-1 and SET-16 mayphysically interact to modulate LET-60 RAS signalling in

C. elegans. Coordinate regulation of HMT and HDMactivities has also been shown in the case of Hox generegulation during ES cell differentiation (Christensenet al., 2007) and in neuronal development (Jepsenet al., 2007). Whether the activity of histone-modifyingenzymes is also coordinately regulated to influence Hoxgene expression in C. elegans is an area of future inves-tigation. Interestingly, a recent study has shown thatcoordinate regulation can also be mediated by a singleenzyme (Lin et al., 2010). The C. elegans KDM7A his-tone demethylase can demethylate both H3K9me2 andH3K27me2, and activate gene expression by interactionof its PHD domain with H3K4me3.

Collectively, most of the enzymes modifying chroma-tin are evolutionary conserved (Tables 1 and 2). There-fore, novel insight from C. elegans on how histonemodifiers and readers contribute to cellular functionwill help us to better understand the underlying molec-ular mechanisms in mammalian systems and in physio-

Table 2Conservation of NuRD, CoREST, Sin3, DRM, and SET1/COMPASS Components and Their Function in C. elegans and Mammals

Complex C. elegans function Complex components Mammalian function Complex components

Mi2/NuRD vulva specification(Ahringer, 2000,von Zalewski 2000)

HDA-1RBA-1LIN-53LET-418 CHD-3MEP-1

important componentsof developmental signalingpathways, potentially withcancer relevance (Xue et al.,1998; Denslow andWade, 2006)

HDAC1, HDAC2RbAp46, RbAp48RbAp38Mi-2alfa, Mi-2beta––

CoREST repressor of the presenilingene hop-1 (Lakowskiet al., 2003)

HDA-1SPR-1SPR-5SPR-3SPR-4DIN-1 (putative

ortholoque)

regulate neuronal geneexpression and neuronalstem cell fate(Lakowski et al., 2006)

HDAC1HDAC2CoRESTLSD1––SHARP

Sin3 formation of malesensory rays(Choy et al., 2007)

HDA-1RBA-1LIN-53SIN-3MAB-21

epigenetic reprogramming(Farias et al., 2010)

HDAC1, HDAC2RbAp46, RbAp48RbAp38Sin3AMab21L1, Mab21L2

NuA4 vulva specification(Ceol et al., 2004)

MYS-1TRR-1EPC-1

essential for cell cycleprogression, gene- specificregulation and DNArepair (Doyon et al., 2004)

HATTRRAPE(Pc)

DRM vulva specification(Harrison et al., 2006)

EFL-1DPL-1LIN-53LIN-35LIN-37LIN-9LIN-52LIN-54

cell proliferation (van denHeuvel and Dyson 2008 )

E2FDPRbAp38RbMip40Mip130–Mip120

SET-1/COMPASS germline immortality,longevity (Simonet et al.,2007; Greer et al.,2010; Li and Kelly,2011; Xiao et al., 2011)

SET-12SET-16ASH-2RBBP-5WDR-5.1WDR-5.2WDR-5.3CFP-1WDR-82DPY-30

essential for vertebratedevelopment, regulationof Hox gene expression(Wysocka et al., 2005)

SET1MLL1Ash2LRbBP5WDR5––CxxC1/Cfp-1WDR82hDPY30

653EPIGENETICS IN C. elegans

Page 8: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

logical processes leading to disease. Future studiesshould shed light on how histone-modifying enzymesregulate the binding of effector proteins to chromatin ina developmental context. Additionally, it will be inter-esting to determine which non-histone substrates arealso modified by these same enzymes.

Histone Modification Patterns and UnusualReaders in C. elegans

In C. elegans, the distribution of H3K36me3(enriched in gene bodies; exons preferentially markedrelative to introns), H4K4me3 (enriched near transcrip-tion start sites) and H3K27me3 (enriched in transcrip-tionally silent regions) is similar to that observed inother organisms (Gerstein et al., 2010; Kolasinska-Zwierz et al., 2009). In addition, H3K36me3 on exonswas found to be dependent on transcription, and waspresent at lower levels on alternatively spliced exons,suggesting a link to splicing. This link between H3K36methylation and splicing may be conserved in mammals(Luco and Misteli, 2011).

Despite the presence of similarities in histone modifi-cation patterns in different species, recent results suggestthat some patterns may vary from one organism to theother (Allis et al., 2006). In C. elegans H3K9 methylation,a repressive mark, is not concentrated at a single regionon each chromosome, as in flies and mammals, but occu-pies broad domains on chromosome arms (Liu et al.,2011). This distribution is consistent with the absence ofcytological heterochromatin and the holocentric natureof C. elegans chromosomes, and may play a role in nu-clear organization through an interaction with the LEM-2protein (Ikegami et al., 2010). Striking novel patterns ofhistone modifications associated with their respectivebinding proteins (readers) may also exist in C. elegans.For example, the linker histone variant HIS-24 recognizesH3K27me3 and is post-translationally modified at lysine14, in contrast to vertebrate H1 which is modified at ly-sine 26 (Kuzmichev et al., 2004; Wirth et al., 2009; M.J-Bunpublished data). Another example is provided by theMBT domain protein LIN-61. While MBT proteins fromDrosophila and mammals recognize mono- and di-meth-ylated lysines on histone H3 and H4 (Bonasio et al.,2010), LIN-61 prefers H3K9me2/3 residues (Koester-Eiserfunke and Fischle, 2011; M.J-B unpublished data).

Histone-Modifying Enzymes Help to MaintainGermline Continuity

Recent data has begun to shed light on how epige-netic mechanisms contribute to the maintenance of cellu-lar identity in multi-cellular organism (cellular memory)(Fisher, 2002). In C. elegans, the H3K4 demethylase SPR-5/LSD1 and the H3K36 methyltransferase MES-4 are impli-cated in distinct mechanisms of epigenetic programmingbetween generations (Katz et al., 2009; Rechtsteiner

et al., 2010). SPR-5/LSD-1 contributes to germline immor-tality by reprogramming epigenetic memory of primordialgerm cells (PGCs) (Katz et al., 2009). Accumulation ofH3K4me2 in PGCs in the absence of SPR-5 demethylaseactivity correlates with misregulation of genes requiredfor spermatogenesis, and with a mortal germline pheno-type (mrt) characterized by progressive sterility. Theseresults suggest that SPR-5/LSD-1 is required to eraseH3K4me2 in the germline and prevent inappropriatetransmission of transcriptional states (Katz et al., 2009).Interestingly, absence of the SET-2 H3K4 HMT is also asso-ciated with a mortal germline phenotype (Xiao et al.,2011), suggesting that the H3K4 methylation state in gen-eral plays an important role in germline stability.

MES-4 has been shown to transmit memory of geneexpression from one generation of germ cells to the nextby maintaining H3K36 methylation of germline-expressed loci in embryos. Genome-wide analysisshowed that in early embryos, MES-4 binds to the all fiveautosomes, where it is abundantly present on the mosthighly transcribed genes, but is under-represented on theX chromosome. Interestingly, the researchers found thatonly genes previously expressed in the maternal germline are bound by MES-4 in a Pol II independent manner,suggesting a mechanism unrelated to ongoing transcrip-tion (Rechtsteiner et al., 2010). Many of these genes areknown to be involved in meiosis (syp-2, zim-2, him-8)and P-granule assembly (pgl-1, -3, glh-2, -4), consistentwith an important role for MES-4 in imparting epigeneticmemory of the transcriptional state of PGCs.

Chromatin regulators are also involved in the mainte-nance of somatic cell fate. synMuv genes antagonizegermline fate in somatic cells, and in their absence germ-line-specific genes are ectopically expressed in the soma(Ceron et al., 2007, Wang et al., 2005; Petrella et al.,2011). Interestingly, MES-4 is required for somatic cellsto acquire germ cell fate in the absence of synMuv activ-ity (Wang et al., 2005) (see Fig. 3). Somewhat surpris-ingly, recent results show that transformation of germcells into somatic cells only requires CHE-1, a transcrip-tion factor that specifies the fate of somatic cells, andloss of the chromatin regulatory factor LIN-53 (Tursunet al., 2011). Further investigation of the important roleplayed by chromatin factors in regulating the germline-soma distinction will shed light on the processesinvolved in germline/soma identity and how germline/soma specific gene expression patterns are controlled.

The Molecular Complexity of PcG

PcG proteins are involved in many aspects of meta-zoan development, including the regulation of Hoxgene clusters, cell cycle progression, X inactivation, celldifferentiation and genomic imprinting (Schuetten-gruber and Cavalli, 2009; Sparmann and van Lohuizen,2006). Ubiquitination of lysine 119 of histone H2A

654 WENZEL ET AL.

Page 9: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

(H2AK119ub), and trimethylation of histone H3 lysine27 (H3K27me3) are indicative of Polycomb group(PcG) activity and are regulated by two major PolycombRepressive Complexes, PRC1 and PRC2, respectively.H2AK119ub is mediated by the PRC1 componentdRing, and H3K27me3 by the PRC2 component E(z)(Cao and Zhang, 2004; Cao et al., 2002, 2005).

C. elegans possesses two Pc-related protein com-plexes: Polycomb group (MES-2/3/6) (Korf et al., 1998)and Polycomb group like (SOP-2/Suppressors of Pal-1and SOP-2-related protein 1/SOR-1). The SOP-2/SOR-1complex appears to be specific to C. elegans (Zhanget al., 2003). More recently, it has been shown thatMIG-32 (abnormal cell migration) and SPAT-3A (sup-pressor of par-two defect), encode functional homo-logues of the human PRC1 complex and are requiredfor H2A ubiquitylation (Karakuzu et al., 2009).

As already discussed, MES proteins were initially iden-tified as factors important for germline development(Capowski et al., 1991; Fong et al., 2002; Holdemanet al., 1998). Subsequently mes-2, mes-3, and mes-6

were identified in a genetic screen to identify regulatorsof male tail neurogenesis. Functional analysis showedthat mes-2, mes-3, and mes-6 act upstream of the Hoxgenes mab-5 and egl-5, and are required for formationof sensory rays in the male tail as well as migration ofspecific neurons (Ross and Zarkower, 2003). Absenceof another Pc related protein, SOP-2, was also shown toresults in homeotic transformations related to wide-spread misexpression of Hox genes (Zhang et al.,2003). However, sop-2 function appears to be inde-pendent ofmes gene activity.

MES-2 also plays a role in late embryonic develop-ment. mes-2 inactivation at this stage results in inappro-priate expression of developmental regulators as well asglobal changes in chromatin structure and organization.Tissue-specific developmental regulators whose expres-sion is upregulated in the absence of MES-2 activityinclude the MADS-box factor UNC-120/SRF (muscle),the GATA factor END-1 (intestine), and the Tbox pro-teins TBX-37 and TBX-38 (foregut) (Yuzyuk et al.,2009). This is similar to the phenomenon observed inDrosophila and mammalian ES cells, where loss ofPRC2 activity results in upregulation of important devel-opmental regulators required for maintenance of cellfate (Schuettengruber et al., 2007).

In flies, the ESC-E(z) complex binds Hox genes anddeposits H3K27me3. This mark is in turn recognized bythe PRC1 complex, thereby antagonizing Trx-G com-plex activity (Simon and Tamkun, 2002). It is unclearwhether the same interplay between E(z)/ESC andPRC1 activities exists in C. elegans. Lack of MIG-32 andSPAT-3 activity causes neuronal defects similar to thoseobserved in mes mutants, and a reduction in H2A ubiq-uitylation. However, unlike mes mutants, mig-32 andspat-3a mutant worms are fertile (Karakuzu et al.,2009). It has been suggested that in the nervous system,MIG-32 and SPAT-3 may play a subtle role together withMES-proteins, distinct from the more global role playedby MES proteins in the establishment of embryonic cellfate and germline development (Karakuzu et al., 2009).How the diverse PcG complexes are targeted to chro-matin to regulate histone modifications and mediaterepression in C. elegans is a key question whichremains to be answered.

Over the last few years, a number of studies haveimplicated RNA in the process of PcG recruitment, pri-marily in mammalian cells. A novel role for a non-codingRNA, HOTAIR, was identified in the regulation of mam-malian Hox gene transcription (Rinn et al., 2007).HOTAIR interacts with both PRC2 and LSD1 to couplehistone H3K27 methylation and H3K4 demethylation(Tsai et al., 2010). A similar mechanism may exist in theworm, where specific short noncoding RNAs (50–200bases) and lincRNAs (long, intergenic noncoding RNAs)can potentially trigger complex chromatin patterns atspecific genes in a temporally organized manner duringdevelopment (M.J-B, unpublished results).

In Drosophila, specific DNA sequences called Poly-comb response elements (PREs) serve as an assemblyplatform for the repressive function of different PcGcomplexes through DNA–protein and protein–proteininteractions (Muller and Kassis, 2006). Whether suchPREs also exist in C. elegans is still unknown. Additionalcomponents of the fly and mammalian ESC-E(z) com-plex include NURF55 and Su(Z)12 (Muller et al., 2002),which are required for binding of the PRC2 complex tonucleosomes (Nekrasov et al., 2005). C. elegans doesnot possess a Su(Z)12 homologue, but does have twoNURF55 homologues: LIN-53 and RBA-1. However, it isunknown whether either one of these two proteinsinteracts with the MES proteins. It remains possible that

FIG. 3. Cellular identity in C. elegans is the sum of interplay between Polycomb (PcG) and Trithorax (Trx-G) group proteins, as well as his-tone modifying enzymes (MES-4, SPR-5). The soma (grey) influences germline cell fate decisions and conversely the germline (black) cancontrol soma-specific gene expression.

655EPIGENETICS IN C. elegans

Page 10: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

additional PRC2 components and recruitment factorsmay exist in C. elegans. Genome-wide studies and bio-chemical analysis, in addition to forward and reversegenetic approaches, may help to understand the molec-ular basis of PcG repression in C. elegans Hox gene reg-ulation and additional processes.

Trx-G and Its Interplay With PcG

PcG repression is antagonized by the Tritorax group(Trx-G) complex, which activates gene expression (Caoet al., 2002; Kal et al., 2000). Trx-G proteins includetwo H3K4 HMTs, MLL (mixed-lineage leukemia), ASH1,ASH2, and the WD40 protein WDR5. Mutations in lin-

59, encoding the C. elegans homologue of ASH-1, affectexpression of two C. elegans HOM-C genes required forposterior development: egl-5 an mab-5 (Chamberlinand Thomas, 2000). Additionally, LIN-59 and UNC-62 (aTALE-class Hox cofactor) were identified as regulatorsof the programmed cell death of specific cells in thedeveloping ventral nerve cord (Potts et al., 2009). InDrosophila, the antagonistic roles of PcG and Trx-Ggroup proteins in the maintenance of transcriptionalON and OFF states of HOX genes have been well char-acterized. Recent data suggests that this antagonismextends to other processes, including longevity control(Siebold et al., 2010). e(z) and esc mutations in fliesincrease life span, resistance to oxidative stress, andstarvation. Furthermore, mutations in Trx-G genes par-tially suppress the increased longevity and stress resist-ance phenotypes of e(z) mutants. While in C. elegans

ash-2 inactivation extends lifespan (Greer et al., 2010),it is not known if the MES/PcG proteins play a similarrole in lifespan regulation.

Are Alterations in the Epigenome Dependenton Early Experience?

C. elegans has a relatively short life cycle (2–3 weeks)and is therefore an excellent model organism to explorewhether and how different environmental or develop-mental experiences influence the cellular memory ofgenetically identical populations. It has recently beenshown that worms that have gone through the stress-resistant dauer larval stage display distinct gene expres-sion patterns and chromatin profiles compared to adultanimals which never exited the reproductive cycle (Hallet al., 2010). These effects depend on the activity ofchromatin-associated proteins, suggesting that chroma-tin remodeling plays a pivotal role in the establishmentof post-dauer expression profiles (Hall et al., 2010).These genomic alterations may contribute to pheno-typic differences among animals exposed to differentenvironmental or developmental experiences, and there-fore influence the phenotypic variation of geneticallyidentical animals. It would be of great interest to betterunderstand the molecular mechanism involved in the

‘‘epigenetically imprinted experience’’ resulting in globalchanges in the adult organism, as well as to establishwhether these mechanisms are evolutionary conserved.

CONCLUSIONS

It is now well established that histone modifiers, theassociated histone modifications and their readers, of-ten function in a concerted way to contribute to chro-matin function. Nonetheless, we are now only begin-ning to understand how the transition from repressedto active chromatin states is achieved, how normal cellsuse dynamic chromatin modifications to establish epige-netic homeostasis, how environmental cues are readand translated through epigenetic mechanisms, andhow chromatin states are established and maintainedfrom one generation to the next.

Information obtained from genome-wide histonemodification profiling, combined with the power ofgenetic approaches in C. elegans, will undoubtedly pro-vide novel insight into the underlying molecular mecha-nisms and the functional importance of epigenetic regu-lation. Given the high degree of conservation betweenchromatin modifiers across species, the informationderived from these studies will likely contribute to abetter understanding of epigenetic regulation in normaldevelopment and longevity. C. elegans itself can be animportant model organism to test different drug targetsinhibiting the function of chromatin modifyingenzymes, and thereby impact research on cancer thera-peutics as well as contribute important information rel-evant to stem-cell technology and cancer.

ACKNOWLEDGMENTS

The authors thank Maja Studencka and Helena Mileticfor critically reading the manuscript.

LITERATURE CITED

Abo-Dalo B, Ndjonka D, Pinnen F, Liebau E, Luersen K.2004. A novel member of the GCN5-related N-acetyl-transferase superfamily from Caenorhabditis elegans

preferentially catalyses the N-acetylation of thialysine[S-(2-aminoethyl)-L-cysteine]. Biochem J 384:129–137.

Agger K, Cloos PA, Christensen J, Pasini D, Rose S, Rap-psilber J, Issaeva I, Canaani E, Salcini AE, Helin K.2007. UTX and JMJD3 are histone H3K27 demethy-lases involved in HOX gene regulation and develop-ment. Nature 449:731–734.

Ahringer J. 2000. NuRD and SIN3 histone deacetylase com-plexes in development. Trends Genet 16:351–356.

Allis CD, Jenuwein T, Reinberg D, Caparros ML. editors.2006. Epigenetics. Cold Spring Harbor, NY: ColdSpring Harbor Laboratory Press.

Andersen EC, Horvitz HR. 2007. Two C. elegans histonemethyltransferases repress lin-3 EGF transcription to

656 WENZEL ET AL.

Page 11: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

inhibit vulval development. Development 134:2991–2999.

Andersen EC, Lu X, Horvitz HR. 2006. C. elegans ISWIand NURF301 antagonize an Rb-like pathway in thedetermination of multiple cell fates. Development133:2695–2704.

Aparicio OM, Billington BL, Gottschling DE. 1991. Modi-fiers of position effect are shared between telomericand silent mating-type loci in S. cerevisiae. Cell 66:1279–1287.

Bender LB, Cao R, Zhang Y, Strome S. 2004. The MES-2/MES-3/MES-6 complex and regulation of histoneH3 methylation in C. elegans. Curr Biol 14:1639–1643.

Bender LB, Suh J, Carroll CR, Fong Y, Fingerman IM,Briggs SD, Cao R, Zhang Y, Reinke V, Strome S.2006. MES-4: An autosome-associated histone meth-yltransferase that participates in silencing the Xchromosomes in the C. elegans germ line. Develop-ment 133:3907–3917.

Bessler JB, Andersen EC, Villeneuve AM. 2010. Differen-tial localization and independent acquisition of theH3K9me2 and H3K9me3 chromatin modificationsin the Caenorhabditis elegans adult germ line. PLoSGenet 6:e1000830.

Bird A. 2002. DNA methylation patterns and epigeneticmemory. Genes Dev 16:6–21.

Bonasio R, Lecona E, Reinberg D. 2010. MBT domainproteins in development and disease. Semin CellDev Biol 21:221–230.

Cao R, Tsukada Y, Zhang Y. 2005. Role of Bmi-1 andRing1A in H2A ubiquitylation and Hox gene silenc-ing. Mol Cell 20:845–854.

Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H,Tempst P, Jones RS, Zhang Y. 2002. Role of histoneH3 lysine 27 methylation in Polycomb-group silenc-ing. Science 298:1039–1043.

Cao R, Zhang Y. 2004. The functions of E(Z)/EZH2-mediated methylation of lysine 27 in histone H3.Curr Opin Genet Dev 14:155–164.

Capowski EE, Martin P, Garvin C, Strome S. 1991. Identi-fication of grandchildless loci whose products arerequired for normal germ-line development in thenematode Caenorhabditis elegans. Genetics 129:1061–1072.

Ceol CJ, Horvitz HR. 2004. A new class of C. eleganssynMuv genes implicates a Tip60/NuA4-like HATcomplex as a negative regulator of Ras signaling.Dev Cell 6:563–576.

Ceron J, Rual JF, Chandra A, Dupuy D, Vidal M, van denHeuvel S. 2007. Large-scale RNAi screens identifynovel genes that interact with the C. elegans retino-blastoma pathway as well as splicing-related compo-nents with synMuv B activity. BMC Dev Biol 7:30.

Chamberlin HM, Thomas JH. 2000. The bromodomainprotein LIN-49 and trithorax-related protein LIN-59

affect development and gene expression in Caeno-

rhabditis elegans. Development 127:713–723.Cho YW, Hong T, Hong S, Guo H, Yu H, Kim D, Guszc-

zynski T, Dressler GR, Copeland TD, Kalkum M.2007. PTIP associates with MLL3- and MLL4-contain-ing histone H3 lysine 4 methyltransferase complex.J Biol Chem 282:20395–20406.

Choi KY, Ji YJ, Jee C, Ahnn J. 2002. Characterization ofCeHDA-7, a class II histone deacetylase interactingwith MEF-2 in Caenorhabditis elegans. BiochemBiophys Res Commun 293:1295–1300.

Coustham V, Bedet C, Monier K, Schott S, Karali M, Pal-ladino F. 2006. The C. elegans HP1 homologue HPL-2 and the LIN-13 zinc finger protein form a compleximplicated in vulval development. Dev Biol 297:308–322.

Couteau F, Guerry F, Muller F, Palladino F. 2002. A heter-ochromatin protein 1 homologue in Caenorhabdi-

tis elegans acts in germline and vulval development.EMBO Rep 3:235–241.

Christensen J, Agger K, Cloos PA, Pasini D, Rose S, Sen-nels L, Rappsilber J, Hansen KH, Salcini AE, Helin K.2007. RBP2 belongs to a family of demethylases,specific for tri-and dimethylated lysine 4 on histone3. Cell 128:1063–1076.

Cui M, Chen J, Myers TR, Hwang BJ, Sternberg PW, Green-wald I, Han M. 2006a. SynMuv genes redundantly in-hibit lin-3/EGF expression to prevent inappropriatevulval induction in C. elegans. Dev Cell 10:667–672.

Cui M, Fay DS, Han M. 2004. lin-35/Rb cooperates withthe SWI/SNF complex to control Caenorhabditis ele-gans larval development. Genetics 167:1177–1185.

Cui M, Kim EB, Han M. 2006b. Diverse chromatinremodeling genes antagonize the Rb-involved Syn-Muv pathways in C. elegans. PLoS Genet 2:e74.

Denslow SA, Wade J. 2007. The human Mi-2/NuRD com-plex and gene regulation. Oncogene 26:5433–5438.

Doyon Y, Selleck W, Lane WS, Song Tan S, Cote J. 2004.Structural and functional conservation of the NuA4histone acetyltransferase complex from yeast tohumans. Mol Cell Biol 24:1884–1896.

Dufourcq P, Victor M, Gay F, Calvo D, Hodgkin J, Shi Y.2002. Functional requirement for histone deacety-lase 1 in Caenorhabditis elegans gonadogenesis.Mol Cell Biol 22:3024–3034.

Eastburn DJ, Han M. 2005. A gain-of-function allele ofcbp-1, the Caenorhabditis elegans ortholog of themammalian CBP/p300 gene, causes an increase inhistone acetyltransferase activity and antagonism ofactivated Ras. Mol Cell Biol 25:9427–9434.

Eimer S, Lakowski B, Donhauser R, Baumeister R. 2002.Loss of spr-5 bypasses the requirement for the C. ele-gans presenilin sel-12 by derepressing hop-1. EMBOJ 21:5787–5796.

Farias EF, Petrie K, Leibovitch B, Murtagh J, Chornet MB,Schenk T, Zelent A, Waxman S. 2010. Interference

657EPIGENETICS IN C. elegans

Page 12: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

with Sin3 function induces epigenetic reprogram-ming and differentiation in breast cancer cells. ProcNatl Acad Sci USA 26:11811–11816.

Fisher AG. 2002. Cellular identity and lineage choice.Nat Rev Immunol 2:977–982.

Fisher K, Southall SM, Wilson JR, Poulin GB. 2010.Methylation and demethylation activities of a C. ele-

gans MLL-like complex attenuate RAS signalling.Dev Biol 341:142–153.

Fong Y, Bender L, Wang W, Strome S. 2002. Regulationof the different chromatin states of autosomes and Xchromosomes in the germ line of C. elegans. Sci-ence 296:2235–2238.

Garvin C, Holdeman R, Strome S. 1998. The phenotypeof mes-2, mes-3, mes-4, and mes-6, maternal-effectgenes required for survival of the germline in Cae-

norhabditis elegans, is sensitive to chromosomedosage. Genetics 148:167–185.

Gerstein MB, Lu ZJ, Van Nostrand EL, Cheng C, ArshinoffBI, Liu T, Yip KY, Robilotto R, Rechtsteiner A, Ike-gami K, Alves P, Chateigner A, Perry M, Morris M,Auerbach RK, Feng X, Leng J, Vielle A, Niu W, Rhris-sorrakrai K, Agarwal A, Alexander RP, Barber G,Brdlik CM, Brennan J, Brouillet JJ, Carr A, CheungMS, Clawson H, Contrino S, Dannenberg LO, Dern-burg AF, Desai A, Dick L, Dose AC, Du J, Egelhofer T,Ercan S, Euskirchen G, Ewing B, Feingold EA, Gass-mann R, Good PJ, Green P, Gullier F, Gutwein M,Guyer MS, Habegger L, Han T, Henikoff JG, Henz SR,Hinrichs A, Holster H, Hyman T, Iniguez AL, Janette J,Jensen M, Kato M, Kent WJ, Kephart E, Khivansara V,Khurana E, Kim JK, Kolasinska-Zwierz P, Lai EC,Latorre I, Leahey A, Lewis S, Lloyd P, Lochovsky L,Lowdon RF, Lubling Y, Lyne R, MacCoss M, Macko-wiak SD, Mangone M, McKay S, Mecenas D, Merri-hew G, Miller DM 3rd, Muroyama A, Murray JI, OoiSL, Pham H, Phippen T, Preston EA, Rajewsky N,Ratsch G, Rosenbaum H, Rozowsky J, Rutherford K,Ruzanov P, Sarov M, Sasidharan R, Sboner A, Scheid P,Segal E, Shin H, Shou C, Slack FJ, Slightam C, Smith R,Spencer WC, Stinson EO, Taing S, Takasaki T, Vafea-dos D, Voronina K, Wang G, Washington NL, WhittleCM, Wu B, Yan KK, Zeller G, Zha Z, Zhong M, ZhouX; modENCODE Consortium, Ahringer J, Strome S,Gunsalus KC, Micklem G, Liu XS, Reinke V, Kim SK,Hillier LW, Henikoff S, Piano F, Snyder M, Stein L, LiebJD, Waterston RH. 2010. Integrative analysis of theCaenorhabditis elegans genome by the modEN-CODE project. Science 330:1775–1787.

Greer EL, Maures TJ, Hauswirth AG, Green EM, LeemanDS, Maro GS, Han S, Banko MR, Gozani O, Brunet A.2010. Members of the H3K4 trimethylation com-plex regulate lifespan in a germline-dependent man-ner in C. elegans. Nature 466:383–387.

Grishok A. 2005a. RNAi mechanisms in Caenorhabditis

elegans. FEBS Lett 579:5932–5939.

Grishok A, Sinskey JL, Sharp PA. 2005b. Transcriptionalsilencing of a transgene by RNAi in the soma ofC. elegans. Genes Dev 19:683–696.

Grote P, Conradt B. 2006. The PLZF-like protein TRA-4cooperates with the Gli-like transcription factorTRA-1 to promote female development in C. ele-

gans. Dev Cell 11:561–573.Hall IM, Shankaranarayana GD, Noma K, Ayoub N,

Cohen A, Grewal SI. 2002. Establishment and main-tenance of a heterochromatin domain. Science 297:2232–2237.

Hall SE, Beverly M, Russ C, Nusbaum C, Sengupta P.2010. A cellular memory of developmental historygenerates phenotypic diversity in C. elegans. CurrBiol 20:149–155.

Harrison MM, Ceol CJ, Lu X, Horvitz HR. 2006. Some C.

elegans class B synthetic multivulva proteinsencode a conserved LIN-35 Rb-containing complexdistinct from a NuRD-like complex. Proc Natl AcadSci USA 103:16782–16787.

Hemberger M, Dean W, Reik W. 2009. Epigenetic dy-namics of stem cells and cell lineage commitment:Digging Waddington’s canal. Nat Rev Mol Cell Biol10:526–537.

Henikoff S. 1990. Position-effect variegation after 60years. Trends Genet 6:422–426.

Holdeman R, Nehrt S, Strome S. 1998. MES-2, a maternalprotein essential for viability of the germline in Cae-

norhabditis elegans, is homologous to a DrosophilaPolycomb group protein. Development 125:2457–2467.

Ikegami K, Egelhofer T, Strome S, Lieb JD. 2010. C. ele-gans chromosome arms are anchored to the nuclearmembrane via discontinuous association with LEM-2. Genome Biol 11:R120.

Jedrusik MA, Schulze E. 2001. A single histone H1 iso-form (H1.1) is essential for chromatin silencing andgermline development in Caenorhabditis elegans.Development 128:1069–1080.

Jedrusik MA, Schulze E. 2003. Telomeric position effectvariegation in Saccharomyces cerevisiae by Caeno-

rhabditis elegans linker histones suggests a mecha-nistic connection between germ line and telomericsilencing. Mol Cell Biol 23: 3681–3691.

Jenuwein T, Allis CD. 2001. Translating the histonecode. Science 293:1074–1080.

Jepsen K, Solum D, Zhou T, McEvilly RJ, Kim HJ, GlassCK, Hermanson O, Rosenfeld MG. 2007. SMRT-medi-ated repression of an H3K27 demethylase in pro-gression from neural stem cell to neuron. Nature450:415–419.

Jin C, Felsenfeld G. 2007. Nucleosome stability medi-ated by histone variants H3.3 and H2A.Z. Genes Dev21:1519–1529.

Kal AJ, Mahmoudi T, Zak NB, Verrijzer CP. 2000. TheDrosophila brahma complex is an essential coacti-

658 WENZEL ET AL.

Page 13: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

vator for the trithorax group protein zeste. GenesDev 14:1058–1071.

Karakuzu O, Wang DP, Cameron S. 2009. MIG-32 andSPAT-3A are PRC1 homologs that control neuronalmigration in Caenorhabditis elegans. Development136:943–953.

Katz DJ, Edwards TM, Reinke V, Kelly WG. 2009. A C.elegans LSD1 demethylase contributes to germlineimmortality by reprogramming epigenetic memory.Cell 137:308–320.

Kelly WG, Fire A. 1998. Chromatin silencing and themaintenance of a functional germline in Caeno-

rhabditis elegans. Development 125:2451–2456.Klose RJ, Kallin EM, Zhang Y. 2006. JmjC-domain-con-

taining proteins and histone demethylation. Nat RevGenet 7:715–727.

Koester-Eiserfunke N, Fischle W. 2011. H3K9me2/3binding of the MBT domain protein LIN-61 is essen-tial for Caenorhabditis elegans vulva development.PLoS Genet 7:e1002017.

Kolasinska-Zwierz P, Down T, Latorre I, Liu T, Liu XS,Ahringer J. 2009. Differential chromatin marking ofintrons and expressed exons by H3K36me3. NatGenet 41:376–381.

Korf I, Fan Y, Strome S. 1998. The polycomb group in Cae-

norhabditis elegans and maternal control of germlinedevelopment. Development 125:2469–2478.

Kuzmichev A, Jenuwein T, Tempst P, Reinberg D. 2004.Different EZH2-containing complexes target methyl-ation of histone H1 or nucleosomal histone H3. MolCell 14:183–193.

Lakowski B, Eimer S, Gobel C, Bottcher A, Wagler B,Baumeister R. 2003. Two suppressors of sel-12

encode C2H2 zinc-finger proteins that regulate pre-senilin transcription in Caenorhabditis elegans. De-velopment 130:2117–2128.

Lakowski B, Roelens I, Jacob S. 2006. CoREST-like com-plexes regulate chromatin modification and neuro-nal gene expression. J Mol Neurosci 29:227–239.

Lans H, Marteijn JA, Schumacher B, Hoeijmakers JH, JansenG, Vermeulen W. 2010. Involvement of global genomerepair, transcription coupled repair, and chromatinremodeling in UV DNA damage response changes dur-ing development. PLoS Genet 6:e1000941.

Lee MG, Villa R, Trojer P, Norman J, Yan KP, Reinberg D,Croce LD, Shiekhattar R. 2007. Demethylation ofH3K27 regulates polycomb recruitment and H2Aubiquitination. Science 318:447–450.

Lee MG, Wynder C, Cooch N, Shiekhattar R. 2005. Anessential role for CoREST in nucleosomal histone 3lysine 4 demethylation. Nature 437:432–435.

Li T, Kelly WG. 2011. A role for Set1/MLL-related com-ponents in epigenetic regulation of the Caenorhab-

ditis elegans germ line. PLoS Genet 7:e1001349.Lin H, Wang Y, Wang Y, Tian F, Pu P, Yu Y, Mao H, Yang

Y, Wang P, Hu L, Lin Y, Liu Y, Xu Y, Chen CD. 2010.

Coordinated regulation of active and repressive his-tone methylations by a dual-specificity histone de-methylase ceKDM7A from Caenorhabditis elegans.Cell Res 20:899–907.

Liu T, Rechtsteiner A, Egelhofer TA, Vielle A, Latorre I,Cheung MS, Ercan S, Ikegami K, Jensen M, Kolasin-ska-Zwierz P, Rosenbaum H, Shin H, Taing S, Taka-saki T, Iniguez AL, Desai A, Dernburg AF, Kimura H,Lieb JD, Ahringer J, Strome S, Liu XS. 2011. Broadchromosomal domains of histone modification pat-terns in C. elegans. Genome Res 21:227–236.

Lu X, Horvitz HR. 1998. lin-35 and lin-53, two genesthat antagonize a C. elegans Ras pathway, encodeproteins similar to Rb and its binding proteinRbAp48. Cell 95:981–991.

Luco RF, Allo M, Schor IE, Kornblihtt AR, Misteli T.2011. Epigenetics in alternative pre-mRNA splicing.Cell 144:16–26.

Luger K, Hansen JC. 2005. Nucleosome and chromatinfiber dynamics. Curr Opin Struct Biol 15:188–196.

McColl G, Killilea DW, Hubbard AE, Vantipalli MC,Melov S, Lithgow GJ. 2008. Pharmacogenetic analy-sis of lithium-induced delayed aging in Caenorhab-

ditis elegans. J Biol Chem 283:350–357.Meister P, Towbin BD, Pike BL, Ponti A, Gasser SM.

2010. The spatial dynamics of tissue-specific pro-moters during C. elegans development. Genes Dev24:766–782.

Metzger E, Wissmann M, Yin N, Muller JM, Schneider R,Peters AH, Gunther T, Buettner R, Schule R. 2005.LSD1 demethylates repressive histone marks to pro-mote androgen-receptor-dependent transcription.Nature 437:436–439.

Moazed D. 2009. Molecular biology. Rejoice–RNAi foryeast. Science 326:533–534.

Muller J, Hart CM, Francis NJ, Vargas ML, Sengupta A,Wild B, Miller EL, O’Connor MB, Kingston RE,Simon JA. 2002. Histone methyltransferase activityof a Drosophila Polycomb group repressor complex.Cell 111:197–208.

Muller J, Kassis JA. 2006. Polycomb response elementsand targeting of Polycomb group proteins in Dro-

sophila. Curr Opin Genet Dev 16:476–484.Nekrasov M, Wild B, Muller J. 2005. Nucleosome bind-

ing and histone methyltransferase activity of Dro-

sophila PRC2. EMBO Rep 6:348–353.Paulsen JE, Capowski EE, Strome S. 1995. Phenotypic

and molecular analysis of mes-3, a maternal-effectgene required for proliferation and viability of thegerm line in C. elegans. Genetics 141:1383–1398.

Petrella LN, Wang W, Spike CA, Rechtsteiner A, ReinkeV, Strome S. 2011. synMuv B proteins antagonizegermline fate in the intestine and ensure C. elegans

survival. Development 138:1069–1079.Pothof J, van Haaften G, Thijssen K, Kamath RS, Fraser

AG, Ahringer J, Plasterk RH, Tijsterman M. 2003.

659EPIGENETICS IN C. elegans

Page 14: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

Identification of genes that protect the C. elegans

genome against mutations by genome-wide RNAi.Genes Dev 17:443–448.

Potts MB, Wang DP, Cameron S. 2009. Trithorax, Hox,and TALE-class homeodomain proteins ensure cellsurvival through repression of the BH3-only geneegl-1. Dev Biol 329:374–385.

Poulin G, Dong Y, Fraser AG, Hopper NA, Ahringer J.2005. Chromatin regulation and sumoylation inthe inhibition of Ras-induced vulval developmentin Caenorhabditis elegans. EMBO J 24:2613–2623.

Rechtsteiner A, Ercan S, Takasaki T, Phippen TM,Egelhofer TA, Wang W, Kimura H, Lieb JD, Strome S.2010. The histone H3K36 methyltransferase MES-4acts epigenetically to transmit the memory of germ-line gene expression to progeny. PLoS Genet 6:e1001091.

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brug-mann SA, Goodnough LH, Helms JA, Farnham PJ,Segal E, Chang HY. 2007. Functional demarcation ofactive and silent chromatin domains in human HOXloci by noncoding RNAs. Cell 129:1311–1323.

Robert VJP, Sijen T, van Wolfswinkel J, Plasterk RHA.2005. Chromatin and RNAi factors protect the C.

elegans germline against repetitive sequences.Genes Dev 19:782–787.

Ross JM, Zarkower D. 2003. Polycomb group regulationof Hox gene expression in C. elegans. Dev Cell 4:891–901.

Sarin S, O’meara MM, Flowers EB, Antonio C, Poole RJ,Didiano D, Johnston RJ, Chang S, Narula S, HobertO. 2007. Genetic screens for Caenorhabditis ele-

gans mutants defective in left/right asymmetric neu-ronal fate specification. Genetics 176:2109–2130.

Schoeftner S, Blasco MA. 2009. A ‘‘higher order’’ of telo-mere regulation: Telomere heterochromatin andtelomeric RNAs. EMBO J 28:2323–2336.

Schuettengruber B, Cavalli G. 2009. Recruitment of pol-ycomb group complexes and their role in thedynamic regulation of cell fate choice. Development136:3531–3542.

Schuettengruber B, Chourrout D, Vervoort M, LeblancB, Cavalli G. 2007. Genome regulation by polycomband trithorax proteins. Cell 128:735–745.

She X, Xu X, Fedotov A, Kelly WG, Maine EM 2009. Reg-ulation of heterochromatin assembly on unpairedchromosomes during Caenorhabditis elegans meio-sis by components of a small RNA-mediated path-way. PLoS Genet 5:e1000624.

Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, ColePA, Casero RA. 2004. Histone demethylation medi-ated by the nuclear amine oxidase homolog LSD1.Cell 119:941–953.

Shibata Y, Takeshita H, Sasakawa N, Sawa H. 2010. Dou-ble bromodomain protein BET-1 and MYST HATs es-

tablish and maintain stable cell fates in C. elegans.Development 137:1045–1053.

Siebold AP, Banerjee R, Tie F, Kiss DL, Moskowitz J, HartePJ. 2010. Polycomb repressive complex 2 and tri-thorax modulate Drosophila longevity and stress re-sistance. Proc Natl Acad Sci USA 107:169–174.

Sijen T, Plasterk RHA. 2003. Transposon silencing in theCaenorhabditis elegans germ line by natural RNAi.Nature 426:310–314.

Simon JA, Tamkun JW. 2002. Programming off and onstates in chromatin: Mechanisms of Polycomb andtrithorax group complexes. Curr Opin Genet Dev12:210–218.

Simonet T, Dulermo R, Schott S, Palladino F. 2007.Antagonistic functions of SET-2/SET1 and HPL/HP1proteins in C. elegans development. Dev Biol 312:367–383.

Simpson VJ, Johnson TE, Hammen RF. 1986. Caeno-

rhabditis elegans DNA does not contain 5-methylcy-tosine at any time during development or aging.Nucleic Acids Res 14:6711–6719.

Sparmann A, van Lohuizen M. 2006. Polycomb silencerscontrol cell fate, development and cancer. Nat RevCancer 6:846–856.

Sternberg PW, Horvitz HR. 1991. Signal transductionduring C. elegans vulval induction. Trends Genet 7:366–371.

Talbert PB, Henikoff S. 2010. Histone variants—Ancientwrap artists of the epigenome. Nat Rev Mol Cell Biol11:264–275.

Tissenbaum HA, Guarente L. 2001. Increased dosage ofa sir-2 gene extends lifespan in Caenorhabditis ele-

gans. Nature 410:227–230.Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK,

Lan F, Shi Y, Segal E, Chang HY. 2010. Long noncod-ing RNA as modular scaffold of histone modificationcomplexes. Science 329:689–693.

Tsukada Y, Fang J, Erdjument-Bromage H, Warren ME,Borchers CH, Tempst P, Zhang Y. 2006. Histonedemethylation by a family of JmjC domain-contain-ing proteins. Nature 439:811–816.

Tursun B, Patel T, Kratsios P, Hobert O. 2011. Directconversion of C. elegans germ cells into specificneuron types. Science 331:304–308.

Unhavaithaya Y, Shin TH, Miliaras N, Lee J, Oyama T,Mello CC. 2002. MEP-1 and a homolog of the NURDcomplex component Mi-2 act together to maintaingermline-soma distinctions in C. elegans. Cell 111:991–1002.

van den Heuvel S, Dyson NS. 2008. Conserved functionsof the pRB and E2F families. Nat Rev Mol Cell Biol 9:713–724.

Volpe TA, Kidner C, Hall IM, Teng G, Grewal SI, Mar-tienssen RA. 2002. Regulation of heterochromaticsilencing and histone H3 lysine-9 methylation byRNAi. Science 297:1833–1837.

660 WENZEL ET AL.

Page 15: Epigenetics in C. elegans: Facts and Challenges · Epigenetics in C. elegans: Facts and Challenges Dirk Wenzel,1 Francesca Palladino,2 and Monika Jedrusik-Bode1* 1Electron Microscopy

von Zelewsky T, Palladino F, Brunschwig K, Tobler H,Hajnal A, Muller F. 2000. The C. elegans Mi-2chromatin-remodeling proteins function in vulvalcell fate determination. Development 127:5277–5284.

Walker AK, Shi Y, Blackwell TK. 2004. An extensiverequirement for transcription factor IID-specificTAF-1 in Caenorhabditis elegans embryonic tran-scription. J Biol Chem 279:15339–15347.

Wang D, Kennedy S, Conte D Jr., Kim JK, Gabel HW,Kamath RS, Mello CC, Ruvkun G. 2005. Somatic mis-expression of germline P granules and enhancedRNA interference in retinoblastoma pathwaymutants. Nature 436:593–597.

Wang J, Zhang M, Zhang Y, Kou Z, Han Z, Chen DY, SunQY, Gao S. 2010. The histone demethylase JMJD2Cis stage-specifically expressed in preimplantationmouse embryos and is required for embryonic de-velopment. Biol Reprod 82:105–111.

Whetstine JR, Nottke A, Lan F, Huarte M, Smolikov S,Chen Z, Spooner E, Li E, Zhang G, Colaiacovo M, ShiY. 2006. Reversal of histone lysine trimethylation bythe JMJD2 family of histone demethylases. Cell 125:467–481.

Wirth M, Paap F, Fischle W, Wenzel D, Agafonov DE,Samatov TR, Wisniewski JR, Jedrusik-Bode M. 2009.HIS-24 linker histone and SIR-2.1 deacetylase induceH3K27me3 in the Caenorhabditis elegans germline. Mol Cell Biol 29:3700–3709.

Wysocka J, Swigut T, Milne TA, Dou Y, Zhang X, Burlin-game AL, Roeder RG, Brivanlou AH, Allis CD. 2005.WDR5 associates with histone H3 methylated at K4and is essential for H3 K4 methylation and verte-brate development. Cell 121:859–872.

Xiao Y, Bedet C, Robert VJP, Simonet T, Dunkelbarger S,Rakotomalala C, Soete G, Korswagen HC, Strome S,Palladino F. C. elegans SET-2 and ASH-2 are differen-tially required for H3 Lys4 methylation in embryosand adult germ cells. Proc Natl Acad Sci USA, doi:10.1073/pnas.1019290108.

Xu L, Fong Y, Strome S. 2001. The Caenorhabditis ele-

gans maternal-effect sterile proteins. MES-2, MES-3,and MES-6, are associated in a complex in embryos.Proc Natl Acad Sci USA 98:5061–5066.

Xue Y, Wong J, Moreno GT, Young MK, Cote J, Wang W.1998. NURD, a novel complex with both ATP-de-pendent chromatin-remodeling and histone deacety-lase activities. Mol Cell 2:851–861.

Yuzyuk T, Fakhouri TH, Kiefer J, Mango SE. 2009. The poly-comb complex protein mes-2/E(z) promotes the transi-tion from developmental plasticity to differentiation inC. elegans embryos. Dev Cell 16:699–710.

Zaratiegui M, Irvine DV, Martienssen RA. 2007. Noncod-ing RNAs and gene silencing. Cell 128:763–776.

Zhang H, Azevedo RB, Lints R, Doyle C, Teng Y, HaberD, Emmons SW. 2003. Global regulation of Hoxgene expression in C. elegans by a SAM domain pro-tein. Dev Cell 4:903–915.

661EPIGENETICS IN C. elegans