Bracken 2009

download Bracken 2009

of 12

Transcript of Bracken 2009

  • 7/30/2019 Bracken 2009

    1/12

    Considrab attntion is crrnty focsd on idntifyingth nts that ad to th dopmnt of so-cadtmor-initiating cs, as ndrstanding this mightfaciitat th dsign of mor ffcti cancr thrapis13.It is bcoming incrasingy idnt that, in addition togntic atrations, tmor dopmnt inos thatration of gn xprssion pattrns owing to epigeneticchangs4. Rcnt stdis ha impicatd th Polycombgroup proteins (PcG protins) as ky contribtors to thschangs59. Th PcG protins form mtiprotin rprs-si compxs, cad Poycomb rprssi compxs(PRCs), which rprss transcription by a mchanism thatprobaby inos th modification of chromatin (FIG. 1).

    Sra gntic stdis in diffrnt organisms hafirmy stabishd th ita and consrd ros for PcGprotins in mbryonic dopmnt and adt somaticc diffrntiation10. Moror, rcnt stdis ha dm-onstratd that th PcG protins ar rqird for main-taining th corrct idntitis of stm, prognitor anddiffrntiatd cs11. Th gnom-wid mapping of PcGtargt gns in mammaian cs has offrd scintists thopportnity to start to nra th mocar mchanisms

    of PcG protin action1214. Th PcG protins ha bnfond to bind and rprss th promotrs of gns thatncod protins with ky ros in c fat dtrminationin many diffrnt car inags. Athogh ths dataspport th arg body of idnc that points to crciaros for th PcG protins in both dopmnt and adthomostasis, w ar ony bginning to ndrstand howth PcG protins actay rgat thir targt gns.

    Initia stdis ha stabishd that th PcG protinsar dispacd from crtain targt gns, for xamp thhomeobox (Hox) genes, on thir transcriptiona actiationdring diffrntiation1315. Howr, sbsqnt stdisdmonstratd that th binding of PcG protins is mch

    mor dynamic than anticipatd, showing that th PcGprotins ar aso rcritd to th promotrs of crtaingns in rspons to diffrntiation signas and, impor-tanty, that this rcritmnt is rqird for thir sincingdring diffrntiation1519. On th basis of ths rsts,w and othrs ha proposd a mod in which th PcGprotins fnction dynamicay dring dopmnt anddiffrntiation to ock off th xprssion of atrnatic fat rgators in any particar inag (FIG. 2). Inthis Riw w propos that th drgation of thsmchanisms is cntra to tmor initiation.

    PG pro r

    Th PcG protins ar ssntia for th maintnanc ofboth norma and cancr stm c popations2022. Thisis party attribtd to thir abiity to bind to and rprssth CDKN2B and CDKN2A loci, which ncod th tumoursuppressors INK4B (ncodd byCDKN2B), INK4A andARF (both ncodd byCDKN2A)21,2331. INK4A andINK4B fnction pstram in th RB pathway, and ARFfnctions pstram in th p53 pathway25,32. In addition tofrqnt gntic atrations, this ocs is oftn pignti-

    cay sincd by DNA mthyation in cancr, and th PcGprotins ha bn proposd to contribt to this26. Manyadditiona PcG targt gns accmat DNA mthyationon thir promotrs in cancr, sch as Wims tmor 1(WT1),rtinoic acid rcptor-(RARB),krpp-ikfactor 4(KLF4),inhibitor of DNA binding 4 (ID4), GATAbinding protin 3 (GATA3)chromodomain hicasDNA binding protin 5(CHD5) and PU.1 (aso known asSPI1)13. Th rports that nhancr of zst homoog 2(EZH2)33 and chromobox homoog 7 (CBX7)34 canphysicay associat with DNA methyltransferases (DNMTs)sggst a mchanism whrby th PcG protins dirctycontribt to th atrd DNA mthyation profis that

    *The Smurfit Institute ofGenetics, Trinity College

    Dublin and The Adelaide &

    Meath Hospital, including the

    National Childrens Hospital,

    Dublin, Ireland.Biotech Research and

    Innovation Centre (BRIC) and

    Centre for Epigenetics,

    University of Copenhagen,

    Copenhagen 22009,

    Denmark.

    emails:

    [email protected];

    [email protected]

    doi:10.1038/nrc2736

    Epigenetic

    Relating to effects on patterns

    of gene expression that are

    heritable through cell division

    and caused by mechanisms

    other than changes to the

    underlying DNA sequence.

    Polycomb group proteins

    A group of proteins first

    described in D. melanogasterthat are required for normal

    development. They work in

    multiprotein complexes, called

    Polycomb repressive

    complexes, that establish

    regions of chromatin in which

    gene expression is repressed.

    Polycomb group proteins: navigators oflineage pathways led astray in cancer

    Adrian P. Bracken* and Kristian Helin

    Abstract | The Polycomb group (PcG) proteins are transcriptional repressors that regulate

    lineage choices during development and differentiation. Recent studies have advanced our

    understanding of how the PcG proteins regulate cell fate decisions and how their

    deregulation potentially contributes to cancer. In this Review we discuss the emerging roles

    of long non-coding RNAs (ncRNAs) and a subset of transcription factors, which we call cellfate transcription factors, in the regulation of PcG association with target genes. We also

    speculate about how their deregulation contributes to tumorigenesis.

    e P i G e n e t i c s a n d G e n e t i c s

    REVIEWS

    NATuRe RevIeWS |CanCer vOluMe 9 | NOveMBeR 2009 |773

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.ncbi.nlm.nih.gov/gene/2625?ordinalpos=4&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSummailto:[email protected]:[email protected]:[email protected]:[email protected]://www.ncbi.nlm.nih.gov/gene/2625?ordinalpos=4&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSum
  • 7/30/2019 Bracken 2009

    2/12

    Homeobox (Hox) genes

    Genes encoding a group of

    transcription factors that

    specify the anteriorposterior

    axis and segment identity of

    metazoan organisms during

    early embryonic development.

    CDKN2B and CDKN2A loci

    A stretch of 45 kb of

    chromosome 9 (in humans),

    which is frequently the target of

    mutation, deletion and

    epigenetic silencing in cancer.

    They encode three tumour

    suppressor proteins: INK4A

    and INK4B, which function in

    the RB pathway, and ARF, which

    functions in the p53 pathway.

    Tumour suppressor

    The loss of function of a tumour

    suppressor (by mutation,

    deletion or epigenetic silencing)

    contributes to cancer

    progression.

    EZH2

    A PcG protein and a histone

    methyltransferase that,

    together with EED and SUZ12

    in the PRC2 complex, catalyses

    the trimethylation of histoneH3 at lysine K27 (H3K27me3).

    DNA methyltransferase

    An enzyme that catalyses

    the transfer of a methyl group

    to DNA.

    BMI1

    BMI1 was initially identified as

    a common site of viral

    integration in Moloney

    virus-induced B cell

    lymphomas. Subsequently,

    BMI1 was shown to be a

    member of the PcG family.

    ar obsrd in mtip cancr typs. In fact, PcG targtgns ar as mch as 12 tims as iky to b abrrantysincd by DNA mthyation in cancr as non-PcG targt

    gns79, and poory diffrntiatd and aggrssi hmantmors show prfrntia rprssion of PcG targt gns6.Takn togthr, ths rsts sggst a possib scnario inwhich PcG protins and DNA mthyating nzyms (schas DNMTs) cooprat to abrranty sinc pro-diffrn-tiation and anti-proifrati gns, which ads to thaccmation of a popation of cs nab to rspondto diffrntiation signas. It is thoght that th consqntbock of diffrntiation may aow ths tmor-initiatingcs to ingr and accmat th additiona pignticand/or gntic atrations ncssary to dop intoa tmor.

    Howr, a ky qstion rmains nanswrd: whattriggrs th abrrant sincing of PcG targt gns that isobsrd in many cancr typs? On potntia scnariois that PcG protins, sch as eZH2 and BMI1, bcomabrranty prgatd, ading to th progrssircritmnt of DNMTs to PcG targt gns, a switch toa mor prmannt transcriptiona sincing and th gn-ration of tmor-initiating cs. Spporting idncfor this hypothsis incds th fact that sra PcGprotins ar highy xprssd in cancr10. For xamp,BMI1 is ampifid and orxprssd in B c ymphomaand fnctions as an oncogene that cooprats withMycin a mos mod of ymphoma3538. Simiary, spprs-sor of zst 12 homoog (SUZ12) is transocatd inndomtria cancr39, and EZH2 is ampifid and highy

    xprssd in many tmor typs4047. Potntiay con-tribting to ths incrasd eZH2 s, th microRNAmiR-101 has rcnty bn rportd to dircty targtEZH2 and is itsf dtd in som cancrs48,49. Howr,dspit th fnctiona idnc for a ro of PcG pro-tins, particary BMI1, in th dopmnt of cancr,th highr s of ths protins frqnty obsrdin tmors cod party b a consqnc of th highproportion of proifrating and/or stm-ik csin tmors. For xamp, BMI1 has bn rportd tob highy xprssd in norma stm cs50, and EZH2xprssion corrats with proifration rat as it is con-trod by th RBe2F pathway41. Thrfor, in this

    Riw w discss an atrnati and compmntaryhypothsis in which PcG protins ar d astray in cancrby th drgation of factors that ar rqird for thirassociation to targt gns. W propos that th drg-ation of ths factors dircty contribts to th abrrantmodation of transcriptiona programms obsrd inmany cancrs.

    PG rrum o rg g

    PcG protins do not ha th abiity to bind spcificDNA motifs. Thrfor, a ky mchanistic qstionconcrns how thy ar rcritd to and dispacd fromthir targt gns dring inag spcification. Thanswr to this qstion not ony has impications for orfndamnta ndrstanding of inag choic dringdopmnt and diffrntiation, bt may aso consid-raby contribt to or ndrstanding of th initiatingnts in cancr.

    Transcription factors recruit PcG proteins. In Drosophilamelanogaster, sra transcription factors ar rqird

    to rcrit PcG protins to polycomb repressive elements(PRes) dring dopmnt51. On sch transcriptionfactor, ncodd byYy1 (aso known as Pho), has rcntybn shown to co-occpy most PRes with PRC1 andPRC2 componnts in D.melanogastermbryos andara19,52. Th PRe in D. melanogasteris not an asiyrcognizab DNA sqnc motif as it is not a singtranscription factor binding sit. Instad, it is a coc-tion of binding sits, dfind as an mnt of srahndrd bas pairs. To dat, PRes ha not bn dfindin mammaian cs, dspit th mapping of sra tho-sand binding sits for th PcG protins1214. This sggststhat many diffrnt mammaian transcription factorscontribt to th rcritmnt of th PcG protins. Infact, if on ooks at th targt gns rgatd by th PcGprotins in mammaian cs and considrs how thy arxprssd in diffrnt c typs, it bcoms diffict toimagin that ony a fw transcription factors ar inodin PcG rcritmnt and dispacmnt. It is iky that thrqirmnt of mtip transcription factors confrs amch gratr fxibiity of targt gn rgation. On thisbasis, it wi b ssntia to dfin ths transcription fac-tors, bcas thir drgation cod b ky to indc-ing cancr that is, thy cod work as oncogns ortmor spprssors.

    So which transcription factors contro th associa-tion of PcG protins with thir targt gns? It has bn

    stimatd that th hman gnom ncods approxi-maty 2,600 transcription factors53,54. W propos thatc fat transcription factors (CFTFs) ar strong can-didats for th rgation of PcG protin rcritmntto and dissociation from thir targt gns. W dfinCFTFs as a transcription factors that fnction to rg-at c fat dcisions dring ithr mbryognsis oradt c diffrntiation. Intrstingy, most if nota CFTFs ar thmss PcG targt gns1214. Somxamps incd th Hox, Sox, Rnx, Fox, Pax andGata transcription factor famiis. Fnctionay, thyar known to rgat many ky c fat dcisions, bothin stm cs and dring diffrntiation, by actiating

    a gl

    The Polycomb group (PcG) proteins regulate cell fate decisions during development

    and differentiation. They form multiprotein repressive complexes called Polycomb

    repressive complexes (PRCs), which modify chromatin.

    The PcG proteins bind and repress the promoters of hundreds of genes encoding

    proteins with roles in cell fate determination.

    It is unclear how PcG proteins are displaced and recruited to different subsets of

    target genes during cell fate decisions. However, cell fate transcription factors

    (CFTFs) and long non-coding RNAs (ncRNAs) are emerging as potential regulators.

    There is growing evidence that many PcG target genes are silenced in advanced

    cancer and that this may be the result of an epigenetic switch to DNA methylation

    during neoplastic progression.

    Several PcG proteins are known to be deregulated in cancer. We propose that the

    deregulation of CFTFs and long ncRNAs also leads to the misexpression of PcG

    target genes.

    REVIEWS

    774 | NOveMBeR 2009 | vOluMe 9 www.tu.cm/w/cc

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.uniprot.org/uniprot/Q15910http://www.ncbi.nlm.nih.gov/gene/648?ordinalpos=3&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/23512?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/43819?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/43819?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/23512?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/648?ordinalpos=3&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.uniprot.org/uniprot/Q15910
  • 7/30/2019 Bracken 2009

    3/12

    |

    K27

    Me3

    Off

    EED

    EZH2

    EED

    SUZ12

    SET

    PSC

    PC

    SCML

    RIN

    G

    CD

    PH

    PSC

    PC

    SCML

    RIN

    G

    CD

    PH

    H2AK119

    Ub

    PRC1

    PRC1PRC2

    PRC2

    PRC2

    K27

    Me3

    K27

    Me3

    K27

    Me3

    K27

    Me3

    K27

    Me3

    EED

    EZH2

    EED

    SUZ12

    SET

    EED

    EZH2

    EED

    SUZ12

    SET

    Oncogene

    A gene that contributes to

    cancer progression as a

    consequence of overexpression

    or of dominantly acting

    mutations that alter the activity

    and/or specificity of the gene

    product.

    MicroRNAThese single-stranded RNAs

    are ~21 to 23 nucleotides in

    length and regulate gene

    expression by partial

    complementary base pairing to

    mRNAs and recruitment to the

    RNA-induced silencing

    complex to inhibit translation

    (and possibly increase

    degradation) of mRNA.

    Polycomb repressive

    element

    A DNA sequence of varying

    length, but often of several

    hundred base pairs, to which

    the PcG proteins can be

    recruited. PREs have been

    defined in D.melanogaster,

    but not in other organisms so

    far. They are composed of a

    collection of transcription

    factor binding sites, defined as

    an element.

    CFTF

    Any transcription factor that

    functions to regulate cell fate

    decisions during differentiation

    and development.

    Embryonic stem cell

    A cell derived from the innercell mass of an early-stage

    embryo known as a blastocyst

    in mice and an epiblast in

    humans. They are immortal,

    can be propagated in vitro and

    are pluripotent.

    SET domain

    A conserved domain that

    catalyses histone lysine

    methyltransferase activity and

    is found in a wide variety of

    chromatin-modifying proteins,

    for example EZH2, SU(VAR)H1,

    MLL1 and G9A.

    th targt gns of spcif ic fats and aso by rprssingatrnati-fat gns5558. emrging data sggst thatsra w-charactrizd CFTFs xrcis thir rosin c fat dcisions throgh rcritmnt of PcG pro-tins to thir targt gns (FIG. 3a). Anaogos to YY1 inD. melanogaster, th thr embryonic stem cell (eS c)cor transcription factors OCT4, SOX2 and NANOGco-occpy a sbst of PcG targt gns in hman eScs14. Sra mammaian transcription factors physi-cay associat with PcG protins, sch as YY1 (REF. 59),RING1 and YY1-binding protin(RYBP)60, PlZF61,62,GATA3 (REF. 63) and e2F6 (REF. 64). Frthrmor, YY1,IKAROS and PlZF ha bn shown to b rqirdfor PcG association with targt gns dring mamma-ian dopmnt and diffrntiation59,61,65. It is ikythat othr CFTFs dispac PcG protins from thirtargt gns dring mammaian car diffrntia-tion (FIG. 3b). Spporting this possibiity is th findingthat tiss-spcific TATA box-binding protin (TBP)-associatd factors (TAFs) can dispac PcG protinsfrom th promotrs of ky diffrntiation gns dr-

    ing trmina diffrntiation ofD. melanogastergrmstm cs66. Takn togthr, ths data sggst thata arg nmbr of diffrnt CFTFs cod fnction inmany diffrnt tiss typs to rgat PcG fnctionand thrfor inag choics.

    Long non-coding RNAs (ncRNAs) recruit PcG proteins.long ncRNAs ar a sbst of ncRNAs that ar ongrthan 200 ncotids and ha dirs car fnctions(BOX 1). Intrstingy, thy ar aso mrging as rcrit-rs of PcG protins to targt gns67. Rcnty, th ongncRNAs HOTAIR, KCNq1OT1 and REPA ha bnshown to rcrit PcG protins to chromatin throghintraction with th PRC2 compx6871. For xamp,Rinn et al. idntifid HOTAIR, which is xprssd fromwithin th HOXCocs, and dmonstratd that it isrqird to rprss transcription in trans across 40kb of th HOXD ocs69. This rprssion is mdiatdthrogh a dirct intraction btwn HOTAIR and thPRC2 compx. Th oss of th PRC2-mdiatd tri-mthyation of ysin 27 of histon H3 (H3K27m3)on th HOXD ocs in HOTAIR-dptd cs d thathors to spcat that HOTAIR rcrits th PRC2compx to this gnomic rgion. Rcnty, sra tho-sand additiona ong ncRNAs ha bn idntifid inboth mos and hman cs, bt itt is known abotthir fnction69,72,73. Th fact that many ha diffrn-

    tia xprssion pattrns dring inag spcificationsggsts that thy may ha important ros dringdopmnt and diffrntiation69,72,73. Intrstingy,on-fifth of a hman ong ncRNAs idntifid to datha bn rportd to physicay associat with thPRC2 compx, sggsting that thy may ha a gnraro in rcriting PcG protins to thir targt gns 73.Intrigingy, Khai et al. carrid ot RNA forscncin situ hybridization on HOTAIR and for additionaPcG-associatd ong ncRNAs and rportd mtipspcks in th c nci, sggsting that thy mightfnction to rcrit PcG protins to many diffrnt tar-gt gns. Th finding that RNA intrfrnc-mdiatd

    knockdown of ong ncRNAs did not affct th xprs-sion s of gns ocatd in cis frthr spports thida that ong ncRNAs fnction in trans.

    So, how do ong ncRNAs rcrit PcG protins tothir targt gns? Crrnt mods (FIG. 3c) ar basdon th proposd abiitis of ong ncRNAs to bindgnomic DNA in a sqnc-spcific mannr at thpromotrs of targt gns and to rcrit PcG pro-tins throgh intraction with th SET domains and/or

    Figure 1 | Cdtd ct Pycmb p

    cmpx. Two major Polycomb repressive complexes

    (PRCs) have been described. The PRC2 complex contains

    the histone methyltransferase enhancer of zeste

    homologue 2 (EZH2), which together with embryonicectoderm development (EED) and suppressor of zeste 12

    homolog (SUZ12) catalyses the trimethylation of histone H3

    at lysine K27 (H3K27me3). The EZH2 SET domain confers

    this activity. Multiple forms of the PRC1 complex exist and

    these contain combinations of at least four PC proteins

    (CBX2, CBX4, CBX7 and CBX8), six PSC proteins (BMI1,

    MEL18, MBLR, NSPC1, RNF159 and RNF3), two RING

    proteins (RNF1 and RNF2), three PH proteins (HPH1, HPH2

    and HPH3) and two SCML proteins (SCML1 1 and SCML2).

    Some results have suggested that PRC1 complexes are

    recruited by the affinity of chromodomains in chromobox

    (Cbx) proteins to the H3K27me3 mark. PRC1 recruitment

    results in the RNF1 and RNF2-mediated ubiquitylation of

    histone H2A on lysine 119, which is thought to be important

    for transcriptional repression. PC, Polycomb; PSC, Posteriorsex combs ; SCML, Sex combs on midleg .

    REVIEWS

    NATuRe RevIeWS |CanCer vOluMe 9 | NOveMBeR 2009 |775

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.uniprot.org/uniprot/Q01860http://www.uniprot.org/uniprot/P48431http://www.uniprot.org/uniprot/Q9H9S0http://www.uniprot.org/uniprot/Q05516http://www.uniprot.org/uniprot/Q13422http://www.ncbi.nlm.nih.gov/sites/entrezhttp://www.ncbi.nlm.nih.gov/sites/entrezhttp://www.uniprot.org/uniprot/Q13422http://www.uniprot.org/uniprot/Q05516http://www.uniprot.org/uniprot/Q9H9S0http://www.uniprot.org/uniprot/P48431http://www.uniprot.org/uniprot/Q01860
  • 7/30/2019 Bracken 2009

    4/12

  • 7/30/2019 Bracken 2009

    5/12

    |

    Off

    Off

    On

    On

    On

    On

    On

    PcG

    PcG

    PcG

    PcG

    Recruitment of PcGs by CFTFs

    Displacement of PcGs by CFTFs

    Recruitment of PcGs by long ncRNAs

    Coordinated regulation of PcGsby long ncRNAs and CFTFs

    Polycomb targetgene promoter

    Long ncRNApromoter

    Differentiation

    Differentiation

    Differentiation

    Differentiation

    PcG targetgene promoter

    Long ncRNApromoter

    a

    b

    c

    d

    CFTF2

    CFTF2

    OffPcG

    Off

    PcG

    PcG

    Off

    CFTF1

    CFTF3

    CFTF4 CFTF5

    th pol rol of cFtF Rna r

    What is known abot th ro of CFTFs and ongncRNAs in cancr? Th gntic idnc spportinga ro for transcription factors in cancr is probabystrongr than for any othr fnctiona grop of pro-tins. For xamp,MYCis on of th bst charactr-izd hman oncogns, and TP53 (which ncodsp53 in hmans) and RB1 ar th two most stdidhman tmor-spprssor gns. Thr is aso strongidnc that at ast 30 CFTFs ar gnticay atrdand contribt to cancr in a tiss-spcific mannr(TABLE 1). Th prcis mchanisms of action ar stipoory ndrstood in many cass. Howr, th i-dnc sggsts that thir norma ros in th rgationof inag-spcific c fat dcisions bcom prtrbdon thir mtation, ampification, transocation anddtion in cancr. Som spcific xamps incd thoncognsMYB85, SOX2(REF. 180),MITF86 and GATA2(REF. 87), and th tmor spprssors GATA3 (REF. 88),CEBP89, IKAROS and PAX5(REF. 90). Importanty, inaddition to bing gnticay atrd in hman can-

    crs, mos mods ha stabishd th significancof CFTFs as cancr-rant gns (TABLE 1).

    Th mod mrging is that CFTFs can b sbdiiddinto two casss on th basis of thir norma fnction,and that th drgation of both casss can potn-tiay contribt to th formation of non-diffrntiatdor tmor-initiating cs (FIG. 4). Oncogns bongto th first cass, as thy ar normay xprssd in

    stm or prognitor cs, and th tmor spprssorsar in th scond cass, as thy ar normay xprssddring diffrntiation and ar rqird for in-ag spcification. W propos that th drgationof ithr cass of CFTFs ads to th accmation of

    Box 1 | Log o-og Rna

    Long non-coding RNAs (ncRNAs) are > 200 nucleotides

    in length.

    Around 3,500 human and 2,000 mouse long ncRNAs

    have been identified to date.

    Long ncRNAs are often transcribed from gene loci that

    are overlapping and interspersed among coding genes.

    Long ncRNAs were thought to be transcriptional noise.

    However, the fact that many long ncRNAs are expressed

    in tightly regulated temporal and regional patterns

    suggests that they have important biological functions.

    Some long ncRNAs have been shown to have diverse

    cellular functions, including imprinting, X chromosome

    inactivation, chromatin remodelling and transcriptional

    regulation.

    Several long ncRNAs are emerging as regulators of

    chromatin-modifying complex (such as polycomb

    repressive complex 2 (PRC2), MLL, G9A, CoREST and

    SMCX) recruitment to target genes.

    Some long ncRNAs are emerging as candidate

    oncogenes and tumour-suppressor genes. (For further

    information see REF. 67).

    Figure 3 | Ptt mcm by wc c t tcpt ct d g -cdg rna uct t

    gut Pycmb gup pt ct wt tgt g dug g cc d pcct.

    | Cell fate transcription factors (CFTFs) recruit Polycomb group (PcG) proteins to target genes during lineage decisions.

    b | CFTFs induce the dissociation of PcG proteins from target genes during lineage decisions.c | Long non-coding RNAs

    (ncRNAs) recruit PcG proteins to target genes during lineage decisions. d | Coordinated action of CFTFs and long ncRNAs

    is necessary to recruit PcG proteins to or dissociate them from target genes during lineage determination. The long

    ncRNAs can function either in cis orin trans.

    REVIEWS

    NATuRe RevIeWS |CanCer vOluMe 9 | NOveMBeR 2009 |777

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.ncbi.nlm.nih.gov/gene/4602?ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/4602?ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/2624?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/5079?ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/5079?ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/2624?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/4602?ordinalpos=2&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSum
  • 7/30/2019 Bracken 2009

    6/12

    Pluripotent cell

    A type of stem cell that is

    capable of differentiating into

    all of the derivatives of the

    three germ layers: ectoderm,

    endoderm and mesoderm.

    cs incapab of ndrgoing diffrntiation (FIG. 4).Ths pr-tmorignic cs thn ha th potn-tia to frthr progrss to bcom tmors aftr thaccmation of additiona gntic and/or pignticatrations. To istrat this hypothsis w dscribsom xamps of ths two casss of CFTFs andpay particar attntion to thos CFTFs for whichthr is idnc of a fnctiona intraction with PcGprotins.

    OCT4 is normay xprssd in pluripotent cells ofth ary mbryo and in eS cs, and it is rqird formaintaining ths cs in an ndiffrntiatd stat91.

    A potntia oncognic actiity of OCT4 was radwhn it was shown to b highy xprssd in hmangrm c tmors and was rqird for thir con-tind growth92. In addition, th ctopic xprssionof OCT4 bocks prognitor c diffrntiation andcass dyspasia in pithia tisss93. Importanty,OCT4 occpis sra hndrd PcG targt gns inhman eS cs and is thoght to contribt to thsstaind rcritmnt of PcG protins to th promot-rs of diffrntiation gns14. Thrfor, th prga-tion of OCT4 in cancr might ad to th prsistnt orsstaind PcG-mdiatd rprssion of diffrntiation

    Table 1 | C t tcpt ct tt dgutd um cc

    G m r Gtc tt um cc edc cc Tgt g

    ETV5 Oncogene Translocated in prostate cancer126 In vitromodel126

    ETV7 Oncogene Overexpressed in lymphoma127 In vivomodel127

    GATA6 Oncogene Amplified in pancreatic cancer128 In vitromodel128

    HOXA9 Oncogene Translocated in myeloid leukaemia102 In vivomodel129

    LMO1 Oncogene Translocated in T cell leukaemia130 In vivomodel131

    LMO2 Oncogene Translocated in T cell leukaemia130 In vivomodel132

    MITF Oncogene Amplified in melanoma133 In vitromodel133 CDKN2AINK4A (REF. 134)

    MYB Oncogene Mutated in colon cancer and transclocated in T-ALL85 In vivomodel135

    MYCN Oncogene Amplified in neuroblastoma136 In vivo model137

    OTX2 Oncogene Amplified in medulloblastoma138,139 In vitromodel139

    PAX3 Oncogene Translocated in alveolar rhabdomyosarcoma140 In vivo model141

    PLZF Oncogene Translocated in acute promyelocytic leukaemia142 In vivo model143

    RUNX1 Oncogene Translocated in AML144 In vivomodel145 NF1(REF. 146)

    TAL1 Oncogene Translocated or mutated in T-ALL131,147 In vivomodel131,147 CD4(REF. 147)

    TBX2 Oncogene Amplified in breast cancer148

    In vitromodel148

    CDKN2AARF

    (REF. 148)

    TITF1 Oncogene Amplified in lung cancer149,150 In vitromodel149,150

    CDX2 TS Mutated in colon cancer151,152 In vivomodel153 CDKN1A(REF. 154)

    CEBPA TS Mutated in AML155 In vivomodel156

    FOXP3 TS Mutated and deleted in breast cancer157,158 In vitroand in vivomodels157,158 ERBB2(REF. 158)andSKP2(REF. 157)

    GATA3 TS Silenced and mutated in breast cancer159,160 In vivomodel159 FOXA1(REF. 159) andCDKN2C161

    HOXA5 TS Silenced in breast cancer162 In vitromodel162 TP53(REF. 162)

    IKAROS TS Deleted in AML90, 107 In vitromodel65 HES1(REF. 65)

    ING1 TS Mutated in squamous cell carcinoma163,164 In vivomodel165

    KLF6 TS Mutated in prostate cancer166 and deregulated in GBM167 In vivomodel167 ATF3(REF. 168)

    PAX5 TS Mutated, deleted or fused in ALL107 In vitromodel107 CD19andCD72(REF. 107)

    PU.1 TS Inactivated or mutated in AML169,170 In vivomodel171, 172 JUNB171

    RUNX3 TS Mutated or silenced in gastric cancer173 In vivomodel174

    SMAD4 TS Mutated in pancreatic cancer175 In vivomodel176

    WT1 Both Mutated in hepatic cancer177,178 In vitromodel179 CDKN1A (REF. 179)

    ALL, acute lymphoblastic leukaemia; AML, acute myeloid leukaemia; ATF3, activating transcription factor 3; CEBPA, CCAAT/enhancer binding protein-; CDKN1A,cyclin-dependent kinase inhibitor 2A; CDKN2A, cyclin-dependent kinase 2A; CDKN2C, cyclin-dependent kinase inhibitor 2C; CDX2, caudal type homeobox 2;ETV, ets variant; Fox, forkhead box; GBM, glioblastoma multiforme; HES1, hairy and enhancer of split 1; Hox, homeobox; LMO, LIM domain only; ING1, inhibitor ofgrowth family, member 1; KLF6, Kruppel-like factor 6; MITF, microphthalmia-associated transcription factor; NF1, neurofibromin 1; OTX2, orthodenticle homeobox2; Pax, paired box; Runx, runt-related transcription factor; SKP2, S-phase kinase-associated protein 2; TAL1, T-cell acute lymphocytic leukaemia 1; T-ALL, T cellacute lymphoblastic leukaemia; TBX2, T-box 2; TS, tumour suppressor; WT1, Wilms tumor 1.

    REVIEWS

    778 | NOveMBeR 2009 | vOluMe 9 www.tu.cm/w/cc

    2009 Macmillan Publishers Limited. All rights reserved

  • 7/30/2019 Bracken 2009

    7/12

    gns and a consqnt bock of th abiity of cs torspond to diffrntiation cs (FIG. 4). Sra othrCFTFs, sch as MYB85, PlZF94, HOXA9 (REFS 9597),PAX3(REF. 98) and PAX7(REF. 99), ar known to fnc-tion in tiss-spcific stm and prognitor cs andha bn fond to ha gain of fnction in cancr.PlZF rcrits BMI1 and th associatd PRC1 com-px to rprss th Hoxdocs dring mos dop-mnt61. PlZF is xprssd in hamatopoitic stm andprognitor cs and is an ssntia rgator of spr-matogonia stm c maintnanc94,100. Importanty,th PlZFrtinoic acid rcptor- (RAR) fsion pro-

    tin, ik th promyocytic kamia (PMl)RARfsion protin, can abrranty rcrit PcG protinsto targt gns dring cancr dopmnt62,101. Thisraiss th possibiity that othr CFTFs form fsionprotins with this abiity. For xamp, HOXA9 isxprssd in hamatopoitic stm cs and pro-gnitors and is transocatd in myoid kamia102.Simiary, th PAX3 and PAX7 CFTFs fnction dr-ing mbryonic myognsis (msc dopmnt) andar transocatd in aoar rhabdomyosarcoma achidhood cancr of skta msc cs 58. Sraothr CFTFs that ar normay xprssd in ndiffr-ntiatd cs ar drgatd in cancr withot bing

    xprssd as fsion protins (TABLE 1). For xamp,MYB is xprssd in coon stm cs and prognitorcs; it is gnticay disrptd in coon cancr by amtation in an intron, ading to highr xprssions85. SOX2, ik OCT4, occpis a sbst of PcGtargt gns in eS cs and is aso xprssd in tiss-spcific stm and prognitor cs, incding nra,ng and osophaga cs103105. Intrstingy, SOX2is ampifid in both ng and osophaga sqamosc carcinomas, sggsting that its ro in cancr isto maintain cs in a pr-trminay diffrntiatdstat180. It wi b intrsting to discor whthrMYB, PAX3, PAX7 and othr CFTFs ar inodin rgating PcG targt gns and/or whthr thyrgat PcG fnction.

    W propos that th scond grop of CFTFs pro-mots diffrntiation by rcriting PcG protins tostm c gns and/or by dispacing PcG protins fromdiffrntiation targt gns (FIG.4). Whn inactiatd incancr (for xamp by dtion or mtation) this wodad to cs bing nab to rprss stm c gns and/

    or actiat a programm of diffrntiation gns. Twopotntia xamps, PAX5 and IKAROS, ar rqirdfor B ymphocyt diffrntiation58,106 and ar fr-qnty dtd in act ymphobastic kamias90,107.Intrstingy, mic with oss of fnction ofIkaros or d-tion ofPax5 ha rdcd H3K27m3 at crtain oci,sggsting that ths CFTFs fnctionay intract withPcG protins65,108. Anothr xamp is GATA3, whichis known to b rqird for mina c diffrntiationin brast pithia88 and is mtatd in brast cancrs109.C/eBP, howr, is rqird for granocytic diffrn-tiation of bipotnt granocyt-macrophag prognitorcs and is mtatd in act myoid kamia89. Mostof ths inag-spcific CFTFs rmain to b charactr-izd in trms of thir intractions with PcG protins andpigntic modifirsper se (TABLE 1).

    Th ida that th drgation of CFTFs can changc fat and ad to tmor dopmnt has bnhighightd by th rcnt intrst in car rpro-gramming110112 (BOX 3). Adt somatic cs can bindcd to trans-diffrntiat into c typs of othrinags or d-diffrntiat into mbryonic stm-ikcs cad indcd pripotnt stm cs. It is nowcar that th controd gain or oss of xprssion ofspcific sts of CFTFs in diffrnt contxts has thpowr to rprogramm c idntity. This has bnshown to ad to a rstting of th pigntic andscap

    in ths cs113,114, frthr spporting th hypothsisthat th drgation of n on CFTF cod potn-tiay indc pigntic rprogramming and contribtto tmor initiation. Importanty, it is aso iky thatoncognic CFTFs (sch as OCT4 and SOX2) whnactiatd or tmor spprssi CFTFs (sch as PAX5and IKAROS) whn inactiatd ad to a bock of thdiffrntiation of immatr cs or a d-diffrntiationof mor matr cs (FIG. 4).

    Do CFTFs aways ha to b gnticay atrd asotind in TABLE 1? Th most iky answr is no. It isw stabishd that car signaing pathways arcommony drgatd in hman cancr 115. Most if

    Box 2 | Ovrvw of rrpom mhoolog

    Gb y mrna xp

    Expression microarrays. These are used to quantify mRNA expression levels in a cell.

    They consist of an arrayed series of thousands of microscopic spots of DNA

    oligonucleotides, each representing a gene, which are used as probes to hybridize a

    cDNA or cRNA sample. After hybridization, the microarray is scanned and software

    used to determine the expression levels of the mRNAs represented. Expression

    microarrays are restricted to the detection of genes that are represented on the array,and this can be limited in certain cases. For example, to cover the entire human

    genome would require as many as 20 chips. This becomes technically challenging,

    labour intensive and expensive.

    RNA sequencing. This is a relatively new technology that applies high-throughput

    next-generation sequencing technologies to sequence cDNA to obtain information

    about the RNA content of a sample120. This method is both quantitatively and

    qualitatively superior to expression arrays. It allows the accurate quantification of

    genes expressed at low levels and qualitatively allows the monitoring of all

    non-annotated genes, including long non-coding RNAs.

    Gm-wd ct y

    ChIP-on-chip or ChIP-chip. Chromatin immunoprecipitation (ChIP) is a method to

    map the DNA location of transcription factors, chromatin remodellers or histone

    modifications. The principle underpinning this assay is that DNA binding proteins in

    living cells are bound to DNA. By using an antibody that is specific to the DNA

    binding protein, one can immunoprecipitate the proteinDNA complex. The

    immunoprecipitated DNA is then isolated and detected by hybridizing the amplified

    DNA on a microarray (chip) (ChIP-on-chip or ChIP-chip). The lysate used for the

    immunoprecipitation can be prepared from non-treated cells, or cells treated with

    formaldehyde, which cross-links proteins bound to DNA.

    ChIPsequencing. This is a recent advancement of the global analysis of

    transcription factor binding sites by ChIP. It applies high-throughput

    next-generation sequencing technologies to sequence the ChIP DNA. This method

    is better than ChIP-chip, because it allows the researcher to ascertain the location

    of binding sites anywhere in the genome. Although this is also possible with

    ChIP-chip, for the human genome this would require as many as 20 chips.

    Moreover, the ChIPsequencing technology generates results with greater

    resolution and higher signal-to-noise values.

    REVIEWS

    NATuRe RevIeWS |CanCer vOluMe 9 | NOveMBeR 2009 |779

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.uniprot.org/uniprot/P23760http://www.uniprot.org/uniprot/P23759http://www.uniprot.org/uniprot/P23759http://www.uniprot.org/uniprot/P23760
  • 7/30/2019 Bracken 2009

    8/12

    |

    On

    On

    OnPcGCFTF1

    CFTF1

    Stem cell gene

    Off

    PcGPcGPcG

    CFTF1CFTF1CFTF1

    Off

    PcG

    PcG

    Off

    Stem cell gene

    Stem cell geneDifferentiation gene

    Differentiation gene

    Differentiation gene

    Stem cell

    Tumour-initiating cell

    Differentiated cell

    Tumour-initiating cell

    Differentiation

    Gain of CFTF1 activitye.g. in OCT4 or SOX2

    Loss of CFTF2 activitye.g. in PAX5 or C/EBP

    Tumourprogression

    Tumour

    Additional genetic andepigenetic alterations

    Tumourprogression

    Additional genetic andepigenetic alterations

    Tumour

    Stem cell gene Differentiation gene

    CFTF2

    Off Off

    CFTF3

    CFTF3PcG

    a

    b c

    not a of ths pathways rgat c fat dci-sions by controing th abndanc and/or actiity ofdownstram ffctor CFTFs. So, for xamp, athoghOCT4 has not bn shown to b gnticay atrdin grm c cancrs, it rsponds to signaing fromfibrobast growth factors, th kamia inhibitoryfactorsigna transdcr and actiator of transcription3 pathway, th transforming growth factor-bonmorphognic protin pathway and th noda andWnt pathways, any of which cod b drgatd116.It is thrfor ogica to assm that many additionaCFTFs ar drgatd in cancr as a consqnc ofatrd signaing pathways. Consistnt with this, s-ra CFTFs ar pignticay, rathr than gnticay,drgatd in cancr. For xamp, PAX2 is prg-atd in ndomtria cancr 117, and HOXA5 is down-rgatd in brast cancr109.

    Th potntia of ong ncRNAs as drirs of tmorformation is aso apparnt. Thr is mrging, if so farimitd, idnc that ong ncRNA gns ar inddcancr rant. Th xprssion ofHOTAIR is highr inmtastatic brast cancr than in primary brast pithiacs (R. Gptha and H.Y. Chang, prsona commnica-tion). Moror, ths obsrations proid idncthat HOTAIR contribts to th mtastatic phnotyp,and that this corratd with th abrrant rcritmnt ofPcG protins to mtip targt gns. In anothr rcntstdy, Y et al. sarchd for antisns transcripts asso-ciatd with 21 w-known tmor spprssor gns118.Thy idntifid a 34.8 kb transcript (which thy cad

    p15AS) that was associatd with th cycin-dpndntkinas inhibitor and PcG targt gn CDKN2B, which isfrqnty sincd in kamia. Th athors xamindth xprssion of both CDKN2B andp15AS in kamic

    Figure 4 | G d c t tcpt ct my d t t mt tumu-ttgc. This model illustrates how the loss or gain of function of two putative cell fate transcription factors (CFTFs)

    may lead to the formation of a tumour-initiating cell. | Normal differentiation of a stem or progenitor cell. The

    levels of CFTF1 decrease during differentiation and the levels of CFTF2 and CFTF3 increase. The decrease of CFTF1

    and the increase of CFTF2 lead to displacement of Polycomb group (PcG) proteins from the promoter of the

    differentiation gene. The increase in CFTF3 levels leads to the recruitment of PcG proteins to the promoter of the

    stem cell gene. b | Conversion of normal stem cells to tumour-initiating cells. In this case, the levels of CFTF1

    become aberrantly high in the stem cells and as a consequence the differentiation gene remains repressed,

    rendering the cell insensitive to differentiation signals. c | The conversion of differentiated cells to

    tumour-initiating cells. In this scenario, CFTF2 function is lost in a differentiated cell (for example, by mutation or

    deletion of the gene) and therefore the cell reverts or de-differentiates to a more stem cell-like state or to a

    tumour-initiating cell in which the differentiation gene is aberrantly silenced. Notably, it is also possible that CFTF1

    could be activated in a differentiated cell or that CFTF2 could be deleted or mutated in a stem cell or progenitor.

    In addition, the loss of CFTF3 activity could lead to an inability to repress the stem cell gene.

    REVIEWS

    780 | NOveMBeR 2009 | vOluMe 9 www.tu.cm/w/cc

    2009 Macmillan Publishers Limited. All rights reserved

    http://www.ncbi.nlm.nih.gov/gene/100048912?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/gene/100048912?ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSum
  • 7/30/2019 Bracken 2009

    9/12

    |

    ES and iPScells

    B cells

    T cells

    Pluripotent cells Progenitors Mature cells

    Neural progenitors

    Blood progenitors

    Pancreatic progenitors

    Mesenchymal progenitors

    Neutrophils

    Macrophages

    Eosinophils

    Mast cells

    Megakaryocytes

    Red blood cells

    Skin fibroblasts

    Neurons

    Astrocytes

    Oligodendrocytes

    Acinar cells

    Beta cells7

    7

    2

    2

    3

    3

    4

    4

    OCT4, KLF4, SOX2 and MYC

    6

    6

    5

    5

    OCT4 and KLF4

    OCT4, KLF4, SOX2, MYC and C/EBP

    OCT4, KLF4, SOX2 and MYC

    Loss of PAX5

    C/EBP

    1

    1

    PDX1, NGN3 and MAFA

    kocyts and fond that in most cassp15AS xprs-sion was incrasd with a concomitant dcras in INK4Bxprssion. ectopic xprssion of p15AS was shown toincras DNA mthyation s at th CDKN2B pro-motr. An intrsting and so far nxpord possibiity isthat th abrranty high s of this ong ncRNA that arobsrd in cancr cod ad to th prmannt rprs-sion of th CDKN2B ocs throgh PcG rcritmnt andsbsqnt accmation of DNA mthyation. Anothrstdy idntifid a 7 kb ong ncRNA, namd hcn, as amarkr for mos hpatocar carcinoma (HCC)119.exprssion of this ong ncRNA was fond to b ightfodhighr in a mos mod of HCC compard with matchd

    norma ir tiss. This high xprssion was obsrd ina stags of HCC, impicating it as a potntia initiatingsion in th dopmnt in cancr. Frthrmor, thathors idntifid a hman orthoog ofhcn, mtastasisassociatd ng adnocarcinoma transcript 1(MALAT1),which is highy xprssd in hman cancrs. It wi bintrsting to dtrmin th bioogica fnction of thsong ncRNAs and in particar whthr thy fnctionto rcrit PcG protins to targt gns dring inagchoics and whthr thir drgation contribts tocancr. It is car that ong ncRNAs rprsnt a prom-ising candidat st of potntia oncogns and tmorspprssor gns.

    Prpv

    Crrnt rsarch fforts ar dirctd at ndrstandingth mchanisms by which PcG protins ar rcritdto and dispacd from thir targt gns dring in-ag spcification. Both CFTFs and ong ncRNAs armrging as ky rgators of ths nts. In th nxtfw yars, w wi s a nmbr of paprs in which

    th targt gns of cancr-rant CFTFs wi bdinatd. Anaogos to th nraing of th tran-scriptiona ntworks controing eS cs, w xpctthat simiar fforts wi stabish th transcriptionantworks of adt stm cs, prognitors and diffr-ntiatd cs. Stdis wi aso addrss th hypothsisthat PcG rcritmnt is rgatd by CFTFs and ongncRNAs dring inag choic. It wi b importantto dtrmin whthr spcific sbsts of PcG targtgns ar actiatd or rprssd by spcific CFTFsor ong ncRNAs dring inag choics. Moror, itwi b intrsting to dtrmin to what xtnt CFTFsrgat th xprssion of ong ncRNAs, and whthrong ncRNAs dictat th rcritmnt of th PcG pro-tins. Ths stdis wi proid important informa-tion rgarding th mocar mchanisms that contronorma c fat dcisions and aso how th drga-tion of ky payrs (that is, CFTFs and ong ncRNAs)might ad to cancr. For instanc, do th many atra-tions in CFTFs that ha bn docmntd in ariostmors (TABLE 1) contribt to th atrd pig-ntic profis obsrd in ths tmors? Crrnty,rmarkaby itt is known abot ths spcifictranscriptiona atrations and how thy ar inf-ncd by th drgation of th CFTFs and/orong ncRNAs.

    Or th nxt 5 yars many fnctionay important

    ong ncRNAs wi probaby b ncord. It wi b fas-cinating to arn how many of th ong ncRNAs wimrg as bona fid oncogns and tmor spprssorgns. Anticancr thrapis targting inag-spcificCFTFs or ong ncRNAs may ha adantags or drgsdirctd at PcG protins or DNA mthyation nzyms,as thy ar mor iky to b c typ spcific. In conc-sion, it is car that a bttr ndrstanding of th ro ofCFTFs and ong ncRNAs in modating th pignticactiity of th PcG protins wi proid mor targtsfor anticancr thrapy, and thrfor is promising forth taior-mad indiidaizd tratmnt of cancrpatints.

    Box 3 | th powr of rrpo for: llulr rprogrmmg

    Terminally differentiated mammalian cells are considered to be stable and rarely,

    apart from in aberrant situations such as cancer development, do they de- or

    trans-differentiate121. Owing to this property it was originally believed that the fates

    imposed on somatic cells during development were final (or terminal), and that cells

    could not be reprogrammed. However, the successful cloning of animals by somatic

    cell nuclear transfer demonstrated that unidentified factor(s) in the oocyte had theability to revert or alter developmental decisions. Since these initial experiments,

    several studies have demonstrated that cell fate transcription factors(CFTFs), when

    deleted or induced exogenously, can cause a switch in the fate of various somatic

    cell types111 (see the figure). For example, the removal ofPax5 from mouse B

    lymphocytes results in their de-differentiation to blood progenitor cells that are

    capable of forming several haematopoietic lineages122. On the exogenous induction

    of CCAAT/enhancer-binding protein- (C/EBP), B cells and T cells can beconverted into macrophages123,124. Pancreatic exocrine cells can be converted to

    insulin producing -cells in vivo, by a combination of neurogenin 3 (NGN3),pancreatic and duodenal homeobox 1 (PDX1) and MAFA CFTFs125. In their seminal

    paper, Takahashi and Yamanaka demonstrated that four CFTFs (OCT4, SOX2, KLF4

    and MYC) were capable of reprogramming adult mouse skin fibroblasts into a

    population of cells that, in many ways, are both molecularly and functionally

    indistinguishable from embryonic stem (ES) cells112. These cells, termed induced

    pluripotent stem (iPS) cells, have also been derived from human skin fibroblasts andseveral other cells of alternative lineages111.

    REVIEWS

    NATuRe RevIeWS |CanCer vOluMe 9 | NOveMBeR 2009 |781

    2009 Macmillan Publishers Limited. All rights reserved

  • 7/30/2019 Bracken 2009

    10/12

    1. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman,

    I. L. Stem cells, cancer, and cancer stem cells. Nature

    414, 105111 (2001).

    2. Stingl, J. & Caldas, C. Molecular heterogeneity of

    breast carcinomas and the cancer stem cell

    hypothesis. Nature Rev. Cancer7, 791799 (2007).

    3. Vescovi, A. L., Galli, R. & Reynolds, B. A. Brain tumour

    stem cells. Nature Rev. Cancer6, 425436 (2006).

    4. Feinberg, A. P., Ohlsson, R. & Henikoff, S. The

    epigenetic progenitor origin of human cancer. Nature

    Rev. Genet.7, 2133 (2006).

    The authors discuss the epigenetic progenitormode of cancer, in which tumour deveopment is

    proposed to invove the ateration of gene

    epression patterns owing to epigenetic changes.

    This mode is under active investigation.5. Ohm, J. E. & Baylin, S. B. Stem cell chromatin

    patterns: an instructive mechanism for DNA

    hypermethylation? Cell Cycle6, 10401043 (2007).

    6. Ben-Porath, I. et al. An embryonic stem cell-like gene

    expression signature in poorly differentiated

    aggressive human tumors. Nature Genet.40,

    499507 (2008).7. Ohm, J. E. et al. A stem cell-like chromatin pattern

    may predispose tumor suppressor genes to DNA

    hypermethylation and heritable silencing. Nature

    Genet.39, 237242 (2007).

    8. Schlesinger, Y. et al. Polycomb-mediated methylation

    on Lys27 of histone H3 pre-marks genes for de novo

    methylation in cancer. Nature Genet.39, 232236

    (2007).

    9. Widschwendter, M. et al. Epigenetic stem cell

    signature in cancer. Nature Genet.39, 157158

    (2007).

    References 79 estabished that PcG target genes

    are frequent aberrant hpermethated b DNA

    methation in cancer.10. Sparmann, A. & van Lohuizen, M. Polycomb silencers

    control cell fate, development and cancer. Nature Rev.

    Cancer6, 846856 (2006).

    11. Pietersen, A. M. & van Lohuizen, M. Stem cell

    regulation by polycomb repressors: postponing

    commitment. Curr. Opin. Cell Biol.20, 201207

    (2008).

    12. Boyer, L. A. et al. Polycomb complexes repress

    developmental regulators in murine embryonic stem

    cells. Nature441, 349353 (2006).

    13. Bracken, A. P., Dietrich, N., Pasini, D., Hansen, K. H. &

    Helin, K. Genome-wide mapping of Polycomb target

    genes unravels their roles in cell fate transitions.

    Genes Dev.20, 11231136 (2006).

    14. Lee, T. I. et al. Control of developmental regulators by

    Polycomb in human embryonic stem cells. Cell125,301313 (2006).

    References 1214 were the first studies to

    perform genome-wide mapping of target genes for

    the PcG proteins.

    15. Pasini, D., Bracken, A. P., Hansen, J. B., Capillo, M. &

    Helin, K. The Polycomb Group protein Suz12 is

    required for embryonic stem cell differentiation. Mol.

    Cell. Biol.27, 37693779 (2007).16. Ezhkova, E. et al. Ezh2 orchestrates gene expression

    for the stepwise differentiation of tissue-specific stem

    cells. Cell136, 11221135 (2009).

    17. Mohn, F. & Schubeler, D. Genetics and epigenetics:

    stability and plasticity during cellular differentiation.

    Trends Genet.25, 129136 (2009).

    18. Mohn, F. et al. Lineage-specific polycomb targets and

    de novo DNA methylation define restriction and

    potential of neuronal progenitors. Mol. Cell30,

    755766 (2008).

    19. Oktaba, K. et al. Dynamic regulation by polycomb

    group protein complexes controls pattern formationand the cell cycle in Drosophila. Dev. Cell15,

    877889 (2008).20. Lessard, J. & Sauvageau, G. Bmi-1 determines the

    proliferative capacity of and leukaemic stem cells.

    Nature423, 255260 (2003).

    21. Molofsky, A. V. et al. Bmi-1 dependence distinguishes

    neural stem cell self-renewal from progenitor

    proliferation. Nature425, 962967 (2003).

    22. Park, I. K. et al. Bmi-1 is required for maintenance of

    adult self-renewing haematopoietic stem cells. Nature

    423, 302305 (2003).

    23. Dietrich, N. et al. Bypass of senescence by the

    polycomb group protein CBX8 through direct binding

    to the INK4A-ARF locus. EMBO J.26, 163748

    (2007).

    24. Gil, J., Bernard, D., Martinez, D. & Beach, D.

    Polycomb CBX7 has a unifying role in cellular lifespan.

    Nature Cell Biol.6, 6772 (2004).

    25. Gil, J. & Peters, G. Regulation of the

    INK4b-ARF-INK4a tumour suppressor locus: all for

    one or one for all. Nature Rev. Mol. Cell Biol.7,

    667677 (2006).

    26. Bracken, A. P. et al. The Polycomb group proteins bind

    throughout the INK4A-ARF locus and are

    disassociated in senescent cells. Genes Dev.21,

    52530 (2007).

    27. Bruggeman, S. W. et al.Ink4a andArfdifferentially

    affect cell proliferation and neural stem cell self-

    renewal in Bmi1-deficient mice. Genes Dev.19,

    14381443 (2005).28. Chen, H. et al. Polycomb protein Ezh2 regulates

    pancreatic -cell Ink4a/Arf expression andregeneration in diabetes mellitus. Genes Dev.23,

    975985 (2009).29. Dhawan, S., Tschen, S. I. & Bhushan, A. Bmi-1 regulates

    the Ink4a/Arf locus to control pancreatic beta-cell

    proliferation.Genes Dev.23, 906911 (2009).

    30. Leung, C. et al. Bmi1 is essential for cerebellar

    development and is overexpressed in human

    medulloblastomas. Nature428, 337341 (2004).

    31. Molofsky, A. V., He, S., Bydon, M., Morrison, S. J. &

    Pardal, R. Bmi-1 promotes neural stem cell self-

    renewal and neural development but not mouse

    growth and survival by repressing the p16Ink4a and

    p19Arf senescence pathways. Genes Dev.19,

    14321437 (2005).

    32. Lowe, S. W. & Sherr, C. J. Tumor suppression by Ink4a-

    Arf: progress and puzzles.Curr. Opin. Genet. Dev.13,

    7783 (2003).

    33. Vire, E. et al. The Polycomb group protein EZH2

    directly controls DNA methylation. Nature439,

    871874 (2006).

    34. Mohammad, H. P. et al. Polycomb CBX7 promotes

    initiation of heritable repression of genes frequently

    silenced with cancer-specific DNA hypermethylation.

    Cancer Res.69, 63226330 (2009).

    35. Bea, S. et al.BMI1 gene amplification and

    overexpression in hematological malignancies occur

    mainly in mantle cell lymphomas. Cancer Res.61,

    24092412 (2001).

    36. Rubio-Moscardo, F. et al. Mantle-cell lymphoma

    genotypes identified with CGH to BAC microarrays

    define a leukemic subgroup of disease and predict

    patient outcome. Blood105, 44454454 (2005).

    37. van Lohuizen, M. et al. Identification of cooperating

    oncogenes in E mu-myc transgenic mice by provirus

    tagging. Cell65, 737752 (1991).

    38. Jacobs, J. J. et al. Bmi-1 collaborates with c-Myc in

    tumorigenesis by inhibiting c-Myc-induced apoptosis

    via INK4a/ARF. Genes Dev.13, 26782690 (1999).

    39. Koontz, J. I. et al. Frequent fusion of theJAZF1andJJAZ1genes in endometrial stromal tumors. Proc.

    Natl Acad. Sci. USA98, 63486353 (2001).

    40. Bachmann, I. M. et al. EZH2 expression is associated

    with high proliferation rate and aggressive tumor

    subgroups in cutaneous melanoma and cancers of the

    endometrium, prostate, and breast.J. Clin. Oncol.24,

    268273 (2006).

    41. Bracken, A. P. et al. EZH2 is downstream of the

    pRB-E2F pathway, essential for proliferation and

    amplified in cancer. EMBO J.22, 532335 (2003).

    42. Kleer, C. G. et al. EZH2 is a marker of aggressive

    breast cancer and promotes neoplastic transformation

    of breast epithelial cells. Proc. Natl Acad. Sci. USA

    100, 1160611611 (2003).

    43. Saramaki, O. R., Tammela, T. L., Martikainen, P. M.,

    Vessella, R. L. & Visakorpi, T. The gene for polycomb

    group protein enhancer of zeste homolog 2 (EZH2) is

    amplified in late-stage prostate cancer. Genes

    Chromosom. Cancer45, 639645 (2006).

    44. van Kemenade, F. J. et al. Coexpression of BMI-1 andEZH2 polycomb-group proteins is associated with

    cycling cells and degree of malignancy in B-cell non-

    Hodgkin lymphoma. Blood97, 38963901 (2001).

    45. Varambally, S. et al. The polycomb group protein

    EZH2 is involved in progression of prostate cancer.

    Nature419, 624629 (2002).

    46. Vekony, H. et al. High expression of Polycomb group

    protein EZH2 predicts poor survival in salivary gland

    adenoid cystic carcinoma.J. Clin. Pathol.61,

    744749 (2008).

    47. Weikert, S. et al. Expression levels of the EZH2

    polycomb transcriptional repressor correlate with

    aggressiveness and invasive potential of bladder

    carcinomas. Int. J. Mol. Med.16, 349353 (2005).

    48. Friedman, J. M. et al. The putative tumor suppressor

    microRNA-101 modulates the cancer epigenome by

    repressing the polycomb group protein EZH2. Cancer

    Res.69, 26232629 (2009).

    49. Varambally, S. et al. Genomic loss of microRNA-101

    leads to overexpression of histone methyltransferase

    EZH2 in cancer. Science322, 16951699 (2008).

    50. Raaphorst, F. M., Otte, A. P. & Meijer, C. J. Polycomb-

    group genes as regulators of mammalian

    lymphopoiesis. Trends Immunol.22, 682690 (2001).

    51. Ringrose, L. & Paro, R. Polycomb/Trithorax response

    elements and epigenetic memory of cell identity.

    Development134, 223232 (2007).52. Kwong, C. et al. Stability and dynamics of polycomb

    target sites in Drosophila development. PLoS Genet.

    4, e1000178 (2008).53. Ashburner, M. et al. Gene ontology: tool for the

    unification of biology. The Gene Ontology Consortium.

    Nature Genet.25, 2529 (2000).

    54. Babu, M. M., Luscombe, N. M., Aravind, L., Gerstein,

    M. & Teichmann, S. A. Structure and evolution of

    transcriptional regulatory networks. Curr. Opin. Struct.

    Biol.14, 283291 (2004).

    55. Blyth, K., Cameron, E. R. & Neil, J. C. The RUNX

    genes: gain or loss of function in cancer. Nature Rev.

    Cancer5, 376387 (2005).

    56. Myatt, S. S. & Lam, E. W. The emerging roles of

    forkhead box (Fox) proteins in cancer. Nature Rev.

    Cancer7, 847859 (2007).

    57. Pearson, J. C., Lemons, D. & McGinnis, W. Modulating

    Hoxgene functions during animal body patterning.

    Nature Rev. Genet.6, 893904 (2005).

    58. Robson, E. J., He, S. J. & Eccles, M. R. A PANorama of

    PAXgenes in cancer and development. Nature Rev.

    Cancer6, 5262 (2006).

    59. Caretti, G., Di Padova, M., Micales, B., Lyons, G. E. &

    Sartorelli, V. The Polycomb Ezh2 methyltransferase

    regulates muscle gene expression and skeletal muscle

    differentiation. Genes Dev.18, 26272638 (2004).

    60. Garcia, E., Marcos-Gutierrez, C., del Mar Lorente, M.,

    Moreno, J. C. & Vidal, M. RYBP, a new repressor protein

    that interacts with components of the mammalian

    Polycomb complex, and with the transcription factor

    YY1.EMBO J.18, 34043418 (1999).61. Barna, M. et al. Plzf mediates transcriptional

    repression ofHoxD gene expression through

    chromatin remodeling. Dev. Cell3, 499510 (2002).

    62. Boukarabila, H. et al. The PRC1 Polycomb group

    complex interacts with PLZF/RARA to mediate leukemic

    transformation. Genes Dev.23, 11951206 (2009).

    63. Hosokawa, H. et al. Regulation of Th2 cell

    development by Polycomb group gene bmi1 through

    the stabilization of GATA3.J. Immunol.177,

    76567664 (2006).

    64. Trimarchi, J. M., Fairchild, B., Wen, J. & Lees, J. A.

    The E2F6 transcription factor is a component of the

    mammalian Bmi1-containing polycomb complex. Proc.Natl Acad. Sci. USA98, 15191524 (2001).

    65. Kleinmann, E., Geimer Le Lay, A. S., Sellars, M.,

    Kastner, P. & Chan, S. Ikaros represses the

    transcriptional response to Notch signaling in T-cell

    development. Mol. Cell. Biol.28, 74657475 (2008).

    66. Chen, X., Hi ller, M., Sancak, Y. & Fuller, M. T. Tissue-

    specific TAFs counteract Polycomb to turn on terminal

    differentiation. Science310, 869872 (2005).

    67. Mercer, T. R., Dinger, M. E. & Mattick, J. S. Long non-

    coding RNAs: insights into functions. Nature Rev.

    Genet.10, 155159 (2009).68. Pandey, R. R. et al. Kcnq1ot1 antisense noncoding

    RNA mediates lineage-specific transcriptional silencing

    through chromatin-level regulation. Mol. Cell32,

    232246 (2008).

    69. Rinn, J. L. et al. Functional demarcation of active and

    silent chromatin domains in human HOX loci by

    noncoding RNAs. Cell129, 13111323 (2007).

    This paper identified HOTAIR as a ong ncRNA that

    is transcribed from the HOXC ocus, which theauthors showed associates with the PRC2 compe

    and is required for recruitment of PcGs to theHOXD ocus.

    70. Terranova, R. et al. Polycomb group proteins Ezh2 and

    Rnf2 direct genomic contraction and imprinted

    repression in early mouse embryos. Dev. Cell15,

    668679 (2008).

    71. Zhao, J., Sun, B. K., Erwin, J. A., Song, J. J. & Lee, J. T.

    Polycomb proteins targeted by a short repeat RNA to

    the mouse X chromosome. Science322, 750756

    (2008).

    72. Dinger, M. E. et al. Long noncoding RNAs in mouse

    embryonic stem cell pluripotency and differentiation.

    Genome Res.18, 14331445 (2008).

    73. Khalil, A. M. et al. Many human large intergenic

    noncoding RNAs associate with chromatin-modifying

    complexes and affect gene expression. Proc. Natl

    Acad. Sci. USA106, 1166711672 (2009).

    REVIEWS

    782 | NOveMBeR 2009 | vOluMe 9 www.tu.cm/w/cc

    2009 Macmillan Publishers Limited. All rights reserved

  • 7/30/2019 Bracken 2009

    11/12

    74. Hogg, J. R. & Collins, K. Structured non-coding RNAs

    and the RNP Renaissance. Curr. Opin. Chem. Biol.12,

    684689 (2008).

    75. Krajewski, W. A., Nakamura, T., Mazo, A. & Canaani,

    E. A motif within SET-domain proteins binds single-

    stranded nucleic acids and transcribed and

    supercoiled DNAs and can interfere with assembly of

    nucleosomes. Mol. Cell. Biol.25, 18911899 (2005).

    76. Akhtar, A., Zink, D. & Becker, P. B. Chromodomains

    are protein-RNA interaction modules. Nature407,

    405409 (2000).

    77. Bernstein, E. et al. Mouse polycomb proteins binddifferentially to methylated histone H3 and RNA and

    are enriched in facultative heterochromatin. Mol. Cell.

    Biol.26, 25602569 (2006).

    78. Muchardt, C. et al. Coordinated methyl and RNA

    binding is required for heterochromatin localization

    of mammalian HP1. EMBO Rep.3, 975981(2002).

    79. Cawley, S. et al. Unbiased mapping of transcription

    factor binding sites along human chromosomes

    21 and 22 points to widespread regulation of

    noncoding RNAs. Cell116, 499509 (2004).

    A andmark paper, which estabished that three

    transcription factors do not primari bind at the

    promoters of known coding genes, as previous

    assumed.80. Guttman, M. et al. Chromatin signature reveals over a

    thousand highly conserved large non-coding RNAs in

    mammals. Nature458, 223227 (2009).

    This paper identified 1,600 conserved ong

    ncRNAs in humans, and showed the differentia

    epression of some of these during embronic

    deveopment and in response to DNA damage.81. Lefterova, M. I. et al. PPAR and C/EBP factors

    orchestrate adipocyte biology via adjacent binding on

    a genome-wide scale. Genes Dev.22, 29412952

    (2008).

    82. Nielsen, R. et al. Genome-wide profiling of PPAR:RXRand RNA polymerase II occupancy reveals temporal

    activation of distinct metabolic pathways and changes

    in RXR dimer composition during adipogenesis. Genes

    Dev.22, 29532967 (2008).

    83. Robertson, A. G. et al. Genome-wide relationship

    between histone H3 lysine 4 mono- and tri-

    methylation and transcription factor binding. Genome

    Res.18, 19061917 (2008).

    84. Zheng, Y. et al. Genome-wide analysis of Foxp3 target

    genes in developing and mature regulatory T cells.

    Nature445, 936940 (2007).

    85. Ramsay, R. G. & Gonda, T. J. MYB function in normal

    and cancer cells. Nature Rev. Cancer8, 523534

    (2008).86. Garraway, L. A. & Sel lers, W. R. Lineage dependency

    and lineage-survival oncogenes in human cancer.

    Nature Rev. Cancer6, 593602 (2006).

    87. Zhang, S. J. et al. Gain-of-function mutation of

    GATA-2 in acute myeloid transformation of chronic

    myeloid leukemia. Proc. Natl Acad. Sci. USA105,

    20762081 (2008).

    88. Kouros-Mehr, H., Kim, J. W., Bechis, S. K. & Werb, Z.

    GATA-3 and the regulation of the mammary luminal

    cell fate. Curr. Opin. Cell Biol.20, 164170 (2008).

    89. Nerlov, C. The C/EBP family of transcription factors: a

    paradigm for interaction between gene expression and

    proliferation control. Trends Cell Biol.17, 318324

    (2007).

    90. Mullighan, C. G. et al. BCR-ABL1 lymphoblastic

    leukaemia is characterized by the deletion of Ikaros.

    Nature453, 110114 (2008).

    91. Nichols, J. et al. Formation of pluripotent stem cells

    in the mammalian embryo depends on the POU

    transcription factor Oct4. Cell95, 379391(1998).

    92. Gidekel, S., Pizov, G., Bergman, Y. & Pikarsky, E.

    Oct-3/4 is a dose-dependent oncogenic fate

    determinant. Cancer Cell4, 361370 (2003).

    93. Hochedlinger, K., Yamada, Y., Beard, C. & Jaenisch, R.

    Ectopic expression of Oct-4 blocks progenitor-cell

    differentiation and causes dysplasia in epithelial

    tissues. Cell121, 465477 (2005).

    94. Reid, A. et al. Leukemia translocation gene, PLZF, is

    expressed with a speckled nuclear pattern in early

    hematopoietic progenitors. Blood86, 45444552

    (1995).95. Ivanova, N. B. et al. A stem cell molecular signature.

    Science298, 601604 (2002).

    96. Sauvageau, G. et al. Differential expression of

    homeobox genes in functionally distinct CD34+

    subpopulations of human bone marrow cells. Proc.

    Natl Acad. Sci. USA91, 1222312227 (1994).

    97. Thorsteinsdottir, U. et al. Overexpression of the

    myeloid leukemia-associated Hoxa9 gene in bone

    marrow cells induces stem cell expansion. Blood99,

    121129 (2002).

    98. Lang, D. et al. Pax3 functions at a nodal point in

    melanocyte stem cell differentiation. Nature433,

    884887 (2005).

    99. Olguin, H. C. & Olwin, B. B. Pax-7 up-regulation

    inhibits myogenesis and cell cycle progression in

    satellite cells: a potential mechanism for self-renewal.

    Dev. Biol.275, 375388 (2004).

    100. Kotaja, N. & Sassone-Corsi, P. Plzf pushes stem cells.Nature Genet.36, 551553 (2004).

    101. Villa, R. et al. Role of the polycomb repressive complex

    2 in acute promyelocytic leukemia. Cancer Cell11,

    513525 (2007).102. Nakamura, T. et al. Fusion of the nucleoporin gene

    NUP98 to HOXA9 by the chromosome translocation

    t(7;11)(p15;p15) in human myeloid leukaemia. Nature

    Genet.12, 154158 (1996).

    103. Que, J. et al. Multiple dose-dependent roles for Sox2

    in the patterning and differentiation of anterior foregut

    endoderm. Development134, 25212531 (2007).

    104. Que, J., Luo, X., Schwartz, R. J. & Hogan, B. L.

    Multiple roles for Sox2 in the developing and adult

    mouse trachea. Development136, 18991907

    (2009).

    105. Chen, X. et al. Multilayered epithelium in a rat model

    and human Barretts esophagus: similar expression

    patterns of transcription factors and differentiation

    markers. BMC Gastroenterol.8, 1 (2008).

    106. Georgopoulos, K. Haematopoietic cell-fate decisions,

    chromatin regulation and ikaros. Nature Rev.

    Immunol.2, 162174 (2002).

    107. Mullighan, C. G. et al. Genome-wide analysis of

    genetic alterations in acute lymphoblastic leukaemia.

    Nature446, 758764 (2007).

    108. Xu, C. R., Schaf fer, L., Head, S. R. & Feeney, A. J.

    Reciprocal patterns of methylation of H3K36 and

    H3K27 on proximal vs. distal IgVH genes are

    modulated by IL-7 and Pax5. Proc. Natl Acad. Sci.

    USA105, 86858690 (2008).

    109. Tlsty, T. D. Luminal cells GATA have it. Nature Cell Biol.

    9, 135136 (2007).

    110. Gurdon, J. B. & Melton, D. A. Nuclear reprogramming

    in cells. Science322, 18111815 (2008).

    111. Hochedlinger, K. & Plath, K. Epigenetic

    reprogramming and induced pluripotency.

    Development136, 509523 (2009).

    112. Takahashi, K. & Yamanaka, S. Induction of pluripotent

    stem cells from mouse embryonic and adult fibroblast

    cultures by defined factors. Cell126, 663676

    (2006).This is a semina paper that describes how the

    epression of just four CFTFs, OCT4, SOx2, KlF4

    and MyC, coud de-differentiate skin fibrobasts

    into embronic stem ces. It inspired efforts to

    generate patient-specific tissues for regenerative

    medicine. From a cancer perspective, together with

    subsequent papers, it highights the

    reprogramming power of CFTFs.113. Deng, J. et al. Targeted bisulfite sequencing reveals

    changes in DNA methylation associated with nuclear

    reprogramming. Nature Biotechnol.27, 353360

    (2009).

    114. Maherali, N. et al. Directly reprogrammed fibroblasts

    show global epigenetic remodeling and widespread

    tissue contribution. Cell Stem Cell1, 5570 (2007).115. Hanahan, D. & Weinberg, R. A. The hallmarks of

    cancer. Cell100, 5770 (2000).

    116. Boiani, M. & Scholer, H. R. Regulatory networks in

    embryo-derived pluripotent stem cells. Nature Rev.

    Mol. Cell Biol.6, 872884 (2005).117. Wu, H. et al. Hypomethylation-linked activation of

    PAX2 mediates tamoxifen-stimulated endometrial

    carcinogenesis. Nature438, 981987 (2005).

    118. Yu, W. et al. Epigenetic silencing of tumour suppressor

    genep15 by its antisense RNA. Nature451,

    202206 (2008).

    This paper identified a ong ncRNA that represses

    CDKN2B.

    119. Lin, R., Maeda, S., Liu, C., Karin, M. & Edgington, T. S.

    A large noncoding RNA is a marker for murine

    hepatocellular carcinomas and a spectrum of human

    carcinomas. Oncogene26, 851858 (2007).120. Wang, Z., Gerstein, M. & Snyder, M. RNA-Seq: a

    revolutionary tool for transcriptomics. Nature Rev.

    Genet.10, 5763 (2009).

    121. Zhou, Q. & Melton, D. A. Extreme makeover:

    converting one cell into another. Cell Stem Cell3,

    382388 (2008).

    122. Nutt, S. L., Heavey, B., Rolink, A. G. & Busslinger, M.

    Commitment to the B-lymphoid lineage depends on

    the transcription factor Pax5. Nature401, 556562

    (1999).

    123. Laiosa, C. V., Stadtfeld, M., Xie, H., de Andres-Aguayo,

    L. & Graf, T. Reprogramming of committed T cell

    progenitors to macrophages and dendritic cells by C/

    EBP and PU.1 transcription factors. Immunity25,731744 (2006).

    124. Xie, H., Ye, M., Feng, R. & Graf, T. Stepwise

    reprogramming of B cells into macrophages. Cell117,

    663676 (2004).125. Zhou, Q., Brown, J., Kanarek, A., Rajagopal, J. &

    Melton, D. A. In vivo reprogramming of adult

    pancreatic exocrine cells to -cells. Nature455,627632 (2008).

    126. Helgeson, B. E. et al. Characterization of

    TMPRSS2:ETV5 and SLC45A3:ETV5 gene fusions in

    prostate cancer. Cancer Res.68, 7380 (2008).

    127. Cardone, M. et al. The novel ETS factor TEL2

    cooperates with Myc in B lymphomagenesis. Mol. Cell.

    Biol.25, 23952405 (2005).

    128. Kwei, K. A. et al. Genomic profiling identifies GATA6 as

    a candidate oncogene amplified in pancreatobiliary

    cancer. PLoS Genet.4, e1000081 (2008).

    129. Kroon, E., Thorsteinsdott ir, U., Mayotte, N.,

    Nakamura, T. & Sauvageau, G. NUP98-HOXA9

    expression in hemopoietic stem cells induces chronic

    and acute myeloid leukemias in mice. EMBO J.20,

    350361 (2001).

    130. Grutz, G. G. et al. The oncogenic T cell LIM-protein

    Lmo2 forms part of a DNA-binding complex

    specifically in immature T cells. EMBO J.17,

    45944605 (1998).

    131. Chervinsky, D. S. et al. Disordered T-cell development

    and T-cell malignancies in SCL LMO1 double-

    transgenic mice: parallels with E2A-deficient mice.

    Mol. Cell. Biol.19, 50255035 (1999).

    132. Fisch, P. et al. T-cell acute lymphoblastic lymphoma

    induced in transgenic mice by the RBTN1 and RBTN2

    LIM-domain genes. Oncogene7, 23892397 (1992).

    133. Garraway, L. A. et al. Integrative genomic analyses

    identify MITF as a lineage survival oncogene amplified

    in malignant melanoma. Nature436, 117122

    (2005).

    134. Loercher, A. E., Tank, E. M., Delston, R. B. & Harbour,

    J. W. MITF links differentiation with cell cycle arrest in

    melanocytes by transcriptional activation of INK4A.

    J. Cell Biol.168, 3540 (2005).

    135. Lidonnici, M. R., Corradini, F., Waldron, T., Bender,

    T. P. & Calabretta, B. Requirement of c-Myb for

    p210(BCR/ABL)-dependent transformation of

    hematopoietic progenitors and leukemogenesis. Blood111, 47714779 (2008).

    136. Brodeur, G. M., Seeger, R. C., Schwab, M., Varmus,

    H. E. & Bishop, J. M. Amplification of N-myc in

    untreated human neuroblastomas correlates with

    advanced disease stage. Science224, 11211124

    (1984).

    137. Weiss, W. A., Aldape, K., Mohapatra, G., Feuerstein,

    B. G. & Bishop, J. M. Targeted expression of MYCN

    causes neuroblastoma in transgenic mice. EMBO J.

    16, 29852995 (1997).

    138. Boon, K., Eberhart, C. G. & Riggins, G. J. Genomic

    amplification of orthodenticle homologue 2 in

    medulloblastomas. Cancer Res.65, 703707 (2005).

    139. Di, C. et al. Identification of OTX2 as a

    medulloblastoma oncogene whose product can be

    targeted by all-trans retinoic acid. Cancer Res.65,

    919924 (2005).

    140. Barr, F. G. et al. Rearrangement of the PAX3 paired

    box gene in the paediatric solid tumour alveolar

    rhabdomyosarcoma. Nature Genet.3, 113117(1993).

    141. Keller, C. et al. Alveolar rhabdomyosarcomas in

    conditional Pax3:Fkhrmice: cooperativity of Ink4a/

    ARF and Trp53 loss of function. Genes Dev.18,

    26142626 (2004).

    142. Chen, Z. et al. Fusion between a novel Kruppel-like

    zinc finger gene and the retinoic acid receptor- locusdue to a variant t(11;17) translocation associated with

    acute promyelocytic leukaemia. EMBO J.12,

    11611167 (1993).

    143. He, L. Z. et al. Distinct interactions of PML-RAR andPLZF-RAR with co-repressors determine differentialresponses to RA in APL. Nature Genet.18, 126135

    (1998).

    144. Zhang, J., Kalkum, M., Yamamura, S., Chait, B. T. &

    Roeder, R. G. E protein silencing by the leukemogenic

    AML1-ETO fusion protein. Science305, 12861289

    (2004).

    REVIEWS

    NATuRe RevIeWS |CanCer vOluMe 9 | NOveMBeR 2009 |783

    2009 Macmillan Publishers Limited. All rights reserved

  • 7/30/2019 Bracken 2009

    12/12