pioneer factors: directing transcriptional regulators within the chromatin environment

10
Pioneer factors: directing transcriptional regulators within the chromatin environment Luca Magnani 1 , Je ´ro ˆ me Eeckhoute 2* and Mathieu Lupien 1* 1 Norris Cotton Cancer Center, Institute for Quantitative Biomedical Sciences, Department of Genetics, Dartmouth Medical School, Lebanon, NH 03756, USA 2 Universite ´ de Lille Nord de France, INSERM UMR1011, UDSL, F-59000 and Institut Pasteur de Lille, F-59019, Lille, France Chromatin is a well-known obstacle to transcription as it controls DNA accessibility, which directly impacts the recruitment of the transcriptional machinery. The recent burst of functional genomic studies provides new clues as to how transcriptional competency is regulated in this context. In this review, we discuss how these studies have shed light on a specialized subset of transcription factors, defined as pioneer factors, which direct recruit- ment of downstream transcription factors to establish lineage-specific transcriptional programs. In particular, we present evidence of an interplay between pioneer factors and the epigenome that could be central to this process. Finally, we discuss how pioneer factors, whose expression and function are altered in tumors, are also being considered for their prognostic value and should therefore be regarded as potential therapeutic targets. Thus, pioneer factors emerge as key players that connect the epigenome and transcription in health and disease. A role for pioneer factors in shaping the chromatin landscape to activate functional regulatory elements The human body consists of hundreds of distinct cell types that are specified by their unique transcriptional pro- grams. These programs are dependent on the interplay between regulatory elements, including enhancers, promo- ters and insulators (see Glossary) [14]. Specific DNA sequences in these regulatory elements are recognized by transcription factors that generate transcriptional reg- ulatory output signals [57]. These signals directly modu- late the transcription of mRNA from DNA by RNA polymerase II. Therefore, transcription factors are respon- sible for the spatial and temporal fine-tuning of transcrip- tional programs. Although they recognize and bind to specific DNA sequences, the recruitment of transcription factors is also dependent on the chromatin landscape (Box 1). Indeed, the distribution of nucleosomes and epigenetic modifications along the genome, as well as the three- dimensional chromatin organization, can regulate tran- scription factor recruitment and activities. For instance, transcription factor binding to chromatin is favored in nucleosome-free regions [3]. Consequently, gene regulation can be dynamically regulated by nucleosome positioning [8,9]. In this instance, enhancers differ significantly from proximal regulatory elements; namely promoters. For ex- ample, promoters generally lie in open and nucleosome- free chromatin regardless of the cell type, whereas acces- sibility to the enhancer is highly variable and cell-type specific [4]. Furthermore, different epigenetic modifica- tions characterize promoters and enhancers [1,5]. The chromatin landscape is therefore a critical barrier that guides the transcriptional potential in a cell type-specific manner by modulating enhancer activity. Understanding how the chromatin landscape is shaped at enhancers is therefore central to understanding cell type-specific com- mitment. Pioneer factors are an emerging class of chromatin- bound proteins grouped according to their biological func- tion, as opposed to homology of sequences and conserved domains. They contribute to the implementation of cell type-specific transcriptional programs upstream of tran- scription factors binding to enhancers [1017]. In general, pioneer factors can therefore be defined as factors that are first able to bind to enhancers and that subsequently Review Glossary Bookmarking: deposition of specific epigenetic modifications that label poised or functional enhancers. This can take place during development to allow delayed activation of enhancers. Chromatin landscape: defined as the combination of parameters that influence its function and that include the distribution of nucleosomes and epigenetic modifications across the genome as well as higher-order folding and related three-dimensional organization (Box 1). Enhancer: a transcriptional regulatory element that activates its target gene over a distance. Functional enhancers are epigenetically marked, sparse of nucleosome, recruit transcription factors and can contact target gene promoters through chromatin looping. Insulator: a transcriptional regulatory element that delineates chromatin domains acting as a barrier to the spread of epigenetic modifications or as an anchor for chromatin looping. Insulators are typically bound by the CTCF transcription factor. Nucleosome: consists of approximately 147 base pairs of DNA wrapped around histone octamers (made of H2A, H2B, H3 and H4). Pioneer factors: defined as a specific class of transcription factors that is required and sufficient to trigger competency of transcriptional regulatory sites (Box 2). Promoter: a transcriptional regulatory element found in the immediate proximity of the transcriptional start site of genes. Promoters recruit the basal transcriptional machinery and initiate transcription. Regulatory element: sequence of DNA bound by transcription factors that regulate gene transcriptional levels. Regulatory elements comprise promoters, enhancers and insulators. Corresponding author: Lupien, M. ([email protected]) * These authors contributed equally to this work. 0168-9525/$ see front matter ß 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.tig.2011.07.002 Trends in Genetics, November 2011, Vol. 27, No. 11 465

Upload: luca-magnani

Post on 13-Sep-2016

218 views

Category:

Documents


6 download

TRANSCRIPT

Pioneer factors: directingtranscriptional regulators within thechromatin environmentLuca Magnani1, Jerome Eeckhoute2* and Mathieu Lupien1*

1 Norris Cotton Cancer Center, Institute for Quantitative Biomedical Sciences, Department of Genetics, Dartmouth Medical School,

Lebanon, NH 03756, USA2 Universite de Lille Nord de France, INSERM UMR1011, UDSL, F-59000 and Institut Pasteur de Lille, F-59019, Lille, France

Review

Glossary

Bookmarking: deposition of specific epigenetic modifications that label poised

or functional enhancers. This can take place during development to allow

delayed activation of enhancers.

Chromatin landscape: defined as the combination of parameters that influence

its function and that include the distribution of nucleosomes and epigenetic

modifications across the genome as well as higher-order folding and related

three-dimensional organization (Box 1).

Enhancer: a transcriptional regulatory element that activates its target gene

over a distance. Functional enhancers are epigenetically marked, sparse of

nucleosome, recruit transcription factors and can contact target gene

promoters through chromatin looping.

Insulator: a transcriptional regulatory element that delineates chromatin

domains acting as a barrier to the spread of epigenetic modifications or as

an anchor for chromatin looping. Insulators are typically bound by the CTCF

transcription factor.

Nucleosome: consists of approximately 147 base pairs of DNA wrapped

around histone octamers (made of H2A, H2B, H3 and H4).

Pioneer factors: defined as a specific class of transcription factors that is

required and sufficient to trigger competency of transcriptional regulatory sites

(Box 2).

Promoter: a transcriptional regulatory element found in the immediate

proximity of the transcriptional start site of genes. Promoters recruit the basal

transcriptional machinery and initiate transcription.

Regulatory element: sequence of DNA bound by transcription factors that

Chromatin is a well-known obstacle to transcription as itcontrols DNA accessibility, which directly impacts therecruitment of the transcriptional machinery. The recentburst of functional genomic studies provides new cluesas to how transcriptional competency is regulated in thiscontext. In this review, we discuss how these studieshave shed light on a specialized subset of transcriptionfactors, defined as pioneer factors, which direct recruit-ment of downstream transcription factors to establishlineage-specific transcriptional programs. In particular,we present evidence of an interplay between pioneerfactors and the epigenome that could be central to thisprocess. Finally, we discuss how pioneer factors, whoseexpression and function are altered in tumors, are alsobeing considered for their prognostic value and shouldtherefore be regarded as potential therapeutic targets.Thus, pioneer factors emerge as key players that connectthe epigenome and transcription in health and disease.

A role for pioneer factors in shaping the chromatinlandscape to activate functional regulatory elementsThe human body consists of hundreds of distinct cell typesthat are specified by their unique transcriptional pro-grams. These programs are dependent on the interplaybetween regulatory elements, including enhancers, promo-ters and insulators (see Glossary) [1–4]. Specific DNAsequences in these regulatory elements are recognizedby transcription factors that generate transcriptional reg-ulatory output signals [5–7]. These signals directly modu-late the transcription of mRNA from DNA by RNApolymerase II. Therefore, transcription factors are respon-sible for the spatial and temporal fine-tuning of transcrip-tional programs. Although they recognize and bind tospecific DNA sequences, the recruitment of transcriptionfactors is also dependent on the chromatin landscape (Box1). Indeed, the distribution of nucleosomes and epigeneticmodifications along the genome, as well as the three-dimensional chromatin organization, can regulate tran-scription factor recruitment and activities. For instance,transcription factor binding to chromatin is favored innucleosome-free regions [3]. Consequently, gene regulationcan be dynamically regulated by nucleosome positioning

Corresponding author: Lupien, M. ([email protected])* These authors contributed equally to this work.

0168-9525/$ – see front matter � 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.tig.2011.0

[8,9]. In this instance, enhancers differ significantly fromproximal regulatory elements; namely promoters. For ex-ample, promoters generally lie in open and nucleosome-free chromatin regardless of the cell type, whereas acces-sibility to the enhancer is highly variable and cell-typespecific [4]. Furthermore, different epigenetic modifica-tions characterize promoters and enhancers [1,5]. Thechromatin landscape is therefore a critical barrier thatguides the transcriptional potential in a cell type-specificmanner by modulating enhancer activity. Understandinghow the chromatin landscape is shaped at enhancers istherefore central to understanding cell type-specific com-mitment.

Pioneer factors are an emerging class of chromatin-bound proteins grouped according to their biological func-tion, as opposed to homology of sequences and conserveddomains. They contribute to the implementation of celltype-specific transcriptional programs upstream of tran-scription factors binding to enhancers [10–17]. In general,pioneer factors can therefore be defined as factors that arefirst able to bind to enhancers and that subsequently

regulate gene transcriptional levels. Regulatory elements comprise promoters,

enhancers and insulators.

7.002 Trends in Genetics, November 2011, Vol. 27, No. 11 465

Box 1. Chromatin landscape

Chromatin is a macromolecule whose primary unit is the nucleosome.

Nucleosomes consist of approximately 147 base pairs of DNA wrapped

around histone octamers (made of H2A, H2B, H3 and H4) that form a

‘beads-on-a-string’ structure. However, nucleosomes are not regularly

spread along the DNA, as eukaryotic cells can dynamically reposition

nucleosomes to increase and/or decrease occupancy of specific

regions. For instance, transcriptional regulatory regions typically

consist of nucleosome-free DNA, making transcription factor recogni-

tion motifs more easily accessible. Moreover, chromatin adopts higher-

order compaction structures that result from modulation of internu-

cleosomal (linker) DNA bending levels through the linker histone H1.

Additionally, long-range interactions are also involved in shaping

chromatin three-dimensional conformation. Such contacts between

distant chromatin sites appear to be crucial to allow enhancers (long-

distance regulatory regions) to transmit signals to the promoters of

their target genes. Long-range interactions implicating insulatory

regions bound by the transcription factor CTCF are also important in

this context to allow and/or impede specific enhancer–promoter

contacts. Together, these different layers of chromatin complexity

and compaction levels therefore impact on the ability of transcription

factors to bind to chromatin and regulate gene expression.

An additional layer of information borne by chromatin consists of

epigenetic modifications of DNA and histones. This involves methyla-

tion and/or hydroxymethylation of cytosines within the DNA and

post-translational modifications (acetylation, methylation, etc.) of

histones. Various combinations of potential DNA and/or histone

modifications are found within the chromatin, depending on the

function of the regions under consideration. In addition, specific

combinations of epigenetic modifications are found at open versus

condensed chromatin regions and transcriptional regulatory events

involve changes in epigenetic modifications. Overall, combinations of

epigenetic modifications define a limited number of functionally

distinct chromatin domains across the genome. Importantly, epige-

netic modifications are involved in a dialog with transcription factors

where they modulate each other’s presence onto the chromatin.

Hence, work in recent years has pointed to a crucial role for epigenetic

modifications in control of transcription factor activities.

Thus, the chromatin landscape is defined as the combination of

parameters that influence its function and that include the distribution

of nucleosomes and epigenetic modifications across the genome, as

well as higher-order folding and related three-dimensional organiza-

tion [105].

Review Trends in Genetics November 2011, Vol. 27, No. 11

trigger their transcriptional competency (Box 2). In thisreview, we focus on recent advances in functional genomicsthat demonstrate how pioneer factors can impact thechromatin landscape to promote cell type-specific tran-scriptional programs. We also discuss evidence for theiremerging role in disease development and progression thatmakes them attractive prognostic markers and therapeu-tic targets in pathologies such as cancer.

Bookmarking enhancers: pioneer factors set the stageAs stated above, cell type-specific transcriptionalresponses are dependent on lineage-specific functionalenhancers [18–20]. Indeed, whereas the genetic informa-tion within an organism is conserved across cell types, agiven enhancer will only be functional in a subset of them.Furthermore, enhancers exhibit species-specific activities[21,22]. Deposition of specific epigenetic modificationscould play an important role in this process because theydistinguish functional from non-functional enhancers. For

Box 2. Pioneer factors

Pioneer factors are defined as a specific class of transcription factors

that are required and sufficient to trigger enhancer competency. In

line with this definition, pioneer factors are found bound to the

chromatin before activation of enhancers and gene expression

modulation. For example, FOXA factors bind the Alb1 enhancer in

primitive endoderm where it is not yet active and Alb1 is not yet

expressed [53]. Importantly, liver specification and Alb1 gene

expression are completely lost in mice when FOXA factors are

inactivated, providing genetic proof for their role as pioneer factors

during liver development [106]. More generally, pioneer factors have

crucial roles in development and differentiation. This implies that

pioneer factors have to recognize their DNA binding motifs in a

context that impedes binding of other transcription factors. Accord-

ingly, FOXA factors are able to bind to nucleosomal DNA [12].

Subsequently, pioneer factors have to trigger recruitment of

additional transcription factors directly responsible for the transcrip-

tional regulatory output signals. Direct protein interactions could help

tether additional transcription factors to regulatory sites. However,

even if this opportunity exists, in many instances pioneer factors

promote transcription factor binding to their own DNA recognition

motifs [19]. This implies that pioneer factors have to remodel the

466

example, mono- and dimethylation of lysine 4 on histone 3(H3K4me1/me2) are commonly associated with functionalenhancers, whereas H3K9me2 is found at non-functionalsites [18,19,23–28]. Therefore, how these enhancers arebookmarked by these epigenetic modifications is central tounderstanding the process guiding the commitment of cellsto a specific fate.

Importantly, pioneer factors can actively contribute tothis process. Indeed, the pioneer factor PU.1 was recentlyshown to promote H3K4me1 deposition at enhancers inthe course of macrophage and B-cell differentiation [20,29](Figure 1). PU.1 binding to the chromatin can be observedwithin an hour following its expression and is accompa-nied by nucleosome repositioning and chromatin opening[20,29]. Subsequently, H3K4me1, which is initially lack-ing at PU.1 binding sites, is gained [20]. H3K4me1 depo-sition coincides with the recruitment of transcriptionfactors guiding the cell type-specific transcriptional pro-gram promoting B cell or macrophage differentiation [20].

chromatin landscape locally (Box 1). First, pioneer factors could

reposition and/or evict nucleosome(s) to promote binding to nucleo-

some-free DNA [107]. However, a recent study suggested that FOXA

factors are not crucial for nucleosome distribution across the genome

of hepatocytes [61]. Second, pioneer factors could modulate higher-

order chromatin structures. For instance, FOXA factors could replace

the linker histone H1 and modulate linker DNA bending [12]. However,

the importance of higher-order chromatin structure in the regulation

of transcription factor accessibility to DNA still remains to be firmly

established [105]. Another attractive explanation for the role of

pioneer factors is that they could be required to trigger epigenetic

changes at enhancers. These changes can affect the chromatin-

binding affinity of proteins (with DNA or histone binding domains),

potentially resulting in direct or indirect regulation of transcription

factor binding [105].

Most potential pioneer factors known to date have been shown to

precede enhancer activation and/or to be required for recruitment of

prototypic transcription factors, which can not directly bind to

compacted chromatin. However, how pioneer factors fulfill this role in

vivo is still elusive and may involve different mechanisms, depending

on the chromatin landscape and the pioneer factor considered.

Pre B-cellMacrophageHepatocyteBreast cell

FOXD3

PU.1

FOXA1

AP-2γ

PBX1 GATA-3

and/orPF

PF

PF

PF

PFTF

etc.Prostate cel

FOXA1 FOXA1

FOXA2

CTCF

FOXD3

CTCF

TLE1

Differen

tiation

TRENDS in Genetics

Figure 1. Pioneer factors and activation of enhancers during cellular differentiation. During differentiation, cells progress through a diverse set of stages associated with

distinct transcriptional programs. These programs are implemented during development in a cell type-specific fashion by marking enhancers with histone modifications

(enhancer bookmarking). Pioneer factors (PF) play an active role in this process. In pluripotent cells (top DNA strand), the chromatin at cell type-specific enhancers is

selectively bookmarked. This is accomplished by arrays of epigenetic modifications, such as mono- or dimethylation of lysine 4 on histone H3 (H3K4me1/me2; green

circles), H3K9me2 (red squares), DNA methylation (red stars) or by transcription factors, such as the pioneer factor FOXD3 or the insulator CTCF. During the transition from

pluripotent to progenitor cells (center strand), pioneer factors (indicated by the colored ovals) are expressed in a cell type-specific manner. They can bind the chromatin at

bookmarked regions engaging in the translation of the histone modification into functional enhancers. This can involve sequential binding of pioneer factors, co-occupancy

or cross-regulation. In addition to simple binding at epigenetically marked enhancers, pioneer factors can also specify changes to the chromatin landscape directing the

deposition of a new set of epigenetic modifications. These include loss of DNA methylation and gain of H3K4me1/me2 (center strand). Furthermore, nucleosome

displacement also contributes to enhancers becoming active for transcription factor (TF) recruitment in committed cells (bottom strand). Hence, as cells commit to a specific

fate, the transcription factors they express will be able to bind the chromatin and potentiate target gene expression.

Review Trends in Genetics November 2011, Vol. 27, No. 11

Indeed, these events allow subsequent binding and activi-ty of the nuclear receptor liver X receptor b (LXR b) inmacrophages [20]. Therefore, PU.1 binding functions as amarker that defines the set of potential cis-regulatoryelements that will subsequently be bound by specifictranscription factors. The concept that PU.1 can repro-gram the chromatin landscape through epigenetic modifi-cation is consistent with the observation that its ectopic

expression can transform fibroblasts into macrophage-like cells [30].

The Forkhead family member, Forkhead box protein A1(FOXA1) also acts as a pioneer factor, bookmarkingenhancers by reprogramming the chromatin landscape(Figure 1) [31]. Indeed, FOXA1 induction and binding tochromatin upon neuronal differentiation of pluripotentcells is followed by accumulation of the H3K4me2

467

Review Trends in Genetics November 2011, Vol. 27, No. 11

epigenetic modification at bound enhancers [31]. Interest-ingly, this is also accompanied by the loss of DNA methyl-ation at those occupied enhancers [31,32].

Pioneer factors can also bookmark enhancers by pre-venting deposition of repressive epigenetic modifications.This has been observed for the Forkhead factor FOXD3(Figure 1). In embryonic stem cells, the pluripotencytranscription factor FOXD3 occupies an enhancer associ-ated with the albumin (Alb1) gene, maintaining it in apoised state (Figure 1) [14,33]. As embryonic stem cellsdifferentiate into endoderm, FOXD3 is replaced byFOXA1, which is required for subsequent activity ofthe enhancer and expression of the Alb1 gene [14,34].By contrast, in cell-lineages where FOXA1 expression isnot induced upon FOXD3 loss, the enhancer gains DNAmethylation. This blocks transcription factor recruitmentand prevents Alb1 expression [14]. Ectopic expression ofFOXD3 in these cells leads to loss of DNA methylation ofthe enhancer, whereas FOXA1 ectopic expression has noeffects [14]. This suggests that these factors are notinterchangeable and provides a rationale for FOXD3binding before FOXA1.

Taken together, these results demonstrate that book-marking by pioneer factors is dependent on facilitating thedeposition of active epigenetic signatures and preventingthat of repressive epigenetic modifications at regulatoryelements (Figure 1). Moreover, these data suggest thatenhancer bookmarking occurs at different stages of cellulardifferentiation.

Translating bookmarks: positive feedback betweenpioneer factors and epigenetic signaturesWhereas pioneer factor binding to the chromatin associ-ates with epigenomic reprogramming in differentiatedcells, epigenetic modifications also provide signals forpioneer factors. Indeed, pioneer factors can act as ‘readers’of epigenetic modifications. In agreement with FOXA1bookmarking functions, in vitro assays have previouslydemonstrated that FOXA1 can bind recombinant histonesdeprived of any post-translational modifications [35].However, nucleosome-harboring H3K4me1/me2 favorsbinding of FOXA1 in vivo (Figure 1) [19,36–38]. Indeed,removal of this epigenetic modification through the ectopicexpression of the histone demethylase KDM1 (lysine-spe-cific demethylase 1; LSD1) significantly abrogates FOXA1binding to chromatin [19,38]. FOXA2, another relatedForkhead family member, recognizes compacted chroma-tin at enhancers marked with H3K4me1 nucleosomes inislet and liver cells [39]. At most enhancers, FOXA2 isfound between two nucleosomes bearing methylation ofH3K4 [39], reminiscent of what was reported for thepioneer factor FOXA1 [40]. By contrast, recent work as-cribed the presence of H3K9me2 epigenetic modification toFOXA1 sites not involved in transcriptional regulation[37]. Comparing FOXA1 binding sites in breast, prostateor colon cancer cells reveals that cell type-specific tran-scriptional regulation is associated with FOXA1 sites thatlack H3K9me2 [37], in agreement with the repressivenature of H3K9 methylation [41]. Overall, this demon-strates that the binding of pioneer factors to the chromatinbenefits from their interplay with epigenetic modifications

468

to define functional enhancers driving transcriptionalregulation.

Other non-histone proteins also affect the activity ofpioneer factors. Indeed, the insulator protein CCCTC-bind-ing factor (CTCF) can influence the binding of pioneerfactors to the chromatin. Through genome-wide studies,it was recently demonstrated that a small fraction ofFOXA1 chromatin-bound sites are co-occupied by CTCF[42]. CTCF depletion reduces FOXA1 binding to thosesites, but favors its recruitment to sites bound by CTCFbut not by FOXA1 in control cells [42,43]. Therefore, CTCFbinding to chromatin appears to be able to both positivelyand negatively impact FOXA1 recruitment. CTCF wasrecently shown to modulate the position of nucleosomessurrounding its binding site [43]. How CTCF modulatespioneer factor recruitment to chromatin and what theimpact is on the chromatin structure remain to be investi-gated.

Enhancer activation through pioneer factor activitiesChromatin compaction varies greatly across the genomeand between cell types. Recent assessment of chromatinaccessibility through genome-wide DNAse I assays coupledwith massively parallel sequencing revealed distinctionsbetween functional and nonfunctional enhancers. Indeed,functional enhancers tend to lie in a open chromatinconformation, whereas nonfunctional distant regulatoryelements are found in compacted chromatin [44,45]. Iden-tical results were obtained using formaldehyde-assistedisolation of regulatory elements (FAIRE)-seq, a techniquemeasuring chromatin accessibility based on phenol-chlo-roform extraction of formaldehyde cross-linked chromatin[46]. Hence, in most instances, compacted chromatin is notable to direct binding of prototypic transcription factors[47].

One of the strategies adopted by transcription factors toovercome the chromatin barrier is to recruit proteins di-rectly that can remodel chromatin upon their recruitment[48–51]. Pioneer factors could also possess intrinsic chro-matin remodeling activities, as suggested by the ability ofFOXA1 to relax condensed chromatin structures in vitro inan ATP-independent manner [34]. Even if the mechanismsinvolved are not yet precisely defined, several studiesindicate that chromatin opening is central to pioneer factorfunctions. For instance, whereas PU.1–/– derived hemato-poietic stem cells fail to express the c-fms gene, re-expres-sion of PU.1 directly translates into chromatin remodelingboth at the promoter and enhancer associated with the c-fms gene, as assessed by DNase I in vivo foot printing [52].More importantly, PU.1 binding at the enhancer is re-quired to facilitate recruitment of early growth response2 (EGR-2), another transcription factor essential for mac-rophage development [52]. Similar conclusions were drawnbased on a genome-wide analysis in pre-B cells, wherePU.1 inducible expression resulted in nucleosome reposi-tioning around the linker region (the DNA sequence foundbetween two nucleosomes), as assessed by MNase diges-tion coupled with massively parallel sequencing [20].

FOXA1 was also reported to reposition nucleosomes invitro [53]. Interestingly, FOXA1 contains a winged helixmotif (FKH/WH) that is highly homologous to that found in

Review Trends in Genetics November 2011, Vol. 27, No. 11

histone H1 [12]. This homology allows FOXA1 to displacelinker histone H1 in vitro on chromatin templates and invivo in embryonic stem (ES) cells after retinoic acid stimu-lation [12,32,34,54]. Given that histone H1 normally favorschromatin compaction by bending the linker DNA andlinking distant nucleosomes to one another, this wouldresult in chromatin opening. Relaxation of chromatin byFOXA1 was evidenced using DNase I digestion [34].FOXA1 genome-wide remodeling actions have also beendemonstrated through FAIRE coupled with massivelyparallel sequencing [42,55]. In line with the existence ofinactive FOXA1-bound sites, it has been previously dem-onstrated that FOXA1 alone is not sufficient to promotecomplete relaxation of the chromatin [37]. However,FOXA1-bound regions deprived of nucleosomes were high-ly associated with cell type-specific transcription [41]. Suchchromatin remodeling ability associated with DNase Ihypersensitivity is also common to several other Forkheadfamily members, including FOXE1, FOXI1 and FOXO1[54–57]. Similarly, binding of the pioneer factor pre-B cellleukemia homeobox 1 (PBX1), previously reported to occu-py chromatin prior to transcription factor recruitment inthe course of muscle differentiation [58], and of the trans-ducin-like enhancer protein 1 (TLE1) in breast cancer cellsassociates with nucleosome depletion [59]. Indeed, FAIREassays in breast cancer cells depleted of TLE1 revealincreased nucleosome occupancy at their binding sites[59]. Chromatin remodeling abilities also distinguish an-other class of protein with pioneer factor characteristics;namely the GATA family. Ectopic expression of GATA-3 innaive primary CD4+ cells directly increases chromatinopenness at the IL-10 enhancer [60]. However, a recentstudy indicates that maintenance of nucleosome distribu-tion in mouse liver might not rely on pioneer factors [61].Indeed, FOXA1 and FOXA2 gene inactivation did not leadto nucleosome repositioning in liver cells [61]. Further-more, at some genomic regions, FOXA1 and FOXA2 bind tonucleosome-occupied sites [38,61]. This suggests that pio-neer factors have different roles at distinct classes ofenhancer [36]. The relative importance of the impact ofpioneer factors on nucleosome repositioning, displacementof the linker histone H1 or deposition of epigenetic marksat different subsets of enhancers, remain to be established.

Importantly, the transcriptional competency of regula-tory elements could derive from collaborations betweendifferent pioneer factors (Figure 1). This is observed duringliver development, where GATA and Forkhead familymembers cooperate to regulate the expression of theAlb1 gene. In addition to the transition between FOXD3and FOXA1 occupancy of the Alb1 enhancer during celldifferentiation to endoderm, GATA-4 can also be recruitedto the FOXA1-occupied enhancer. In fact, both in vitro andin vivo assays confirm that FOXA1 and GATA-4 co-occupythe Alb1 enhancer [12,13,16,62]. In agreement with therequired presence of both pioneer factors at the Alb1enhancer, depletion of either prevents the expression ofthe Alb1 gene [16]. FOXA1 cooperation with other pioneerfactors also applies in other cell types. For instance, inbreast cells, FOXA1 is found at sites occupied by theactivating protein 2 (AP-2)g pioneer factor [63]. The inter-dependency between these pioneer factors is highlighted

by the fact that each is required for the recruitment of theother [63]. By contrast, whereas FOXA1 can also cooperatewith TLE1 in breast cancer cells, the depletion of eitherpioneer factor does not affect the binding to chromatin ofthe other [59]. Cooperation between pioneer factors hasalso been reported during B cell development. Indeed, SRY(sex determining region Y)-box 2 (SOX2) is required for therecruitment of FOXD3 to the chromatin in embryonic stemcells. This than allows for the deposition of the H3K4me2epigenetic modification [64]. Similarly, in pro-B cells,SOX4 is required for the recruitment of the forkhead factorFOXP1 [64]. However, the co-occupancy of enhancers bypioneer factors does not always translate into cooperation.Indeed, in erythroid differentiation, PU.1 binding atGATA-1-bound regulatory elements leads to the repressionof GATA-1 target genes through the implementation ofrepressive epigenetic modifications [65]. Conversely,GATA-1 binding at PU.1-bound regulatory elements dur-ing myelopoiesis impairs the expression of PU.1 targetgenes [65].

In summary, epigenetically marked functional enhan-cers are remodeled and maintained in an open chromatinconformation through the action of pioneer factors. Theinterplay between pioneer factors through collaboration,substitution or cross-regulation further contributes to theestablishment of specific transcriptional programs. Thesemechanisms are important for development and cellulardifferentiation, and may be altered in diseases such ascancer, contributing to establishment of aberrant tran-scriptomes.

Pioneer factors in cancerCancer commonly associates with altered transcriptionalprograms. For example, it is possible to discriminate sev-eral subtypes of breast tumor based on their transcription-al profiles [66]. Changes to the chromatin landscapethrough chromatin remodeling and epigenetic modifica-tions play an active role in the establishment of theseanomalous programs [67–70]. Through their chromatinremodeling activity and their capacity to modulate thedeposition of epigenetic modifications, pioneer factorsare prime candidate drivers of changes in transcriptionalprograms seen in cancer. Accordingly, reports have dem-onstrated that genes encoding pioneer factors and theirgenomic activities are altered in multiple types of cancer(Figure 2).

Alterations to pioneer factor genes

Mutations within pioneer factor genes can disrupt theiractivity and contribute to cancer. For instance, the PU.1gene is mutated in 7% of patients with acute myeloidleukemia (AML) [71]. Although the effect of these muta-tions on the function of PU.1 as a pioneer factor has notbeen investigated clearly, they decrease its capacity tointeract with transcription factors, such as runt-relatedtranscription factor 1 (RUNX1) and JUN, and directlyimpact target gene expression [71]. In addition, transloca-tion events can target genes encoding pioneer factors. Forexample, a translocation event frequently involves PBX1in leukemia, where a fusion protein harboring the trans-activation domain of the transcription factor E2A and the

469

SNP-induceddifferential PF binding

PF

T/G

Altered chromatin landscape

Other cancer-related genetic and/or epigenetic alteration

Other cancer-relatedevents on partners

Altered PF function

PF-FUSION or PF

PF

PF-FUSION

or

PF

or

Overexpression

PF gene PF-fusion gene

TranslocationMutation

PF gene

Increased PF occupancy

PF PFPFPF

Gene level

Proteinlevel

Target level

PF

PF

PF

TRENDS in Genetics

Figure 2. Role of pioneer factors in cancer development. Cancer is characterized by aberrant expression profiles resulting in uncontrolled cell proliferation. Activities of

pioneer factors (PF) are altered in several different ways in cancer. At the gene level, pioneer factors may be overexpressed, mutated or rearranged through translocation

events (cancer-associated alteration is presented in red). As a consequence, pioneer factors see their protein level increase, their DNA binding specificity altered and their

interaction with partners modified. These events can be enhanced by additional cancer-related events targeting their partners. In addition, the genomic activity of pioneer

factors can be targeted in the course of cancer development. For instance, mutations or genetic variations within their DNA binding motif can modulate their binding

capacity. Overexpression of pioneer factors can result in increased occupancy at certain genomic locations and binding to de novo sites. Mutations and translocation events

in pioneer factor coding sequences can also alter their function by driving their recruitment to de novo sites or altering their activity at endogenous binding sites. Finally,

cancer-dependent de novo epigenetic reprogramming may guide pioneer factors to new sites. For example, de novo deposition of the epigenetic modification H3K4me2

(red/green circles) provides enhancers with a signature favoring FOXA1 binding in prostate cancer. Hence, these events highlight the multitude of way through which

pioneer factors can be affected in cancer. Abbreviation: SNP, single nucleotide polymorphism.

Review Trends in Genetics November 2011, Vol. 27, No. 11

DNA-binding domain of PBX1 is generated [72]. Thisresults in the expression of target genes through PBX1binding sites [72].

In addition to mutations and translocation events, pio-neer factors are generally overexpressed in cancer(Figure 2). This is seen in breast [73], ovarian [74] andprostate cancers [75] for PBX1. This also applies to AP-2g,which is commonly overexpressed in breast cancer [76–78],

470

where it facilitates estrogen receptor a (ERa) signaling[63]. FOXA1 overexpression has also been reported invarious types of cancer [79–81]. Moreover, DNA copy num-ber amplification of FOXA1 is found in metastatic prostatetumors [81] and in breast tumors [82]. Overexpression oramplification of FOXA1 protein levels in breast and pros-tate cancer cells is directly linked to its ability to specifychromatin binding and function of ERa and androgen

Review Trends in Genetics November 2011, Vol. 27, No. 11

receptor (AR) [10,15,19,37,42,83]. FOXA1 is also amplifiedand overexpressed in thyroid carcinoma, where it promotescell cycle progression by controlling the expression ofp27kip1 [84]. Hence, changes in the protein levels of pio-neer factors through their overexpression or amplificationare common in cancer. However, the mechanisms drivingthese events and how they impact on pioneer factor normalfunctions are, for the most part, yet to be identified.

Disrupting pioneer factor genomic activity

The chromatin-binding activity of pioneer factors is as-cribed to their capacity to recognize specific DNA motifs inenhancers [19,20,61]. Recently, genome-wide associationstudies have linked genetic variants, such as single nucle-otide polymorphisms (SNPs), to cancer development. In-terestingly, some of the SNPs associated with prostatecancer map to FOXA1 binding sites [85,86]. Importantly,these SNPs were shown to disrupt the genomic activity ofFOXA1 by modulating its binding to chromatin (Figure 2)[85]. Disruption of pioneer factor binding by SNPs istherefore potentially involved in cancer development.The ongoing whole-genome resequencing efforts led bythe 1000 Genomes Project is constantly expanding thelist of genetic variants found within the human genome[87]. Many of these are likely to map to pioneer factorbinding sites. Therefore, it will be critical to investigatethe functional implication of these genetic variants onpioneer factor activity and their potential role in diseasedevelopment. Similarly, cancer genome resequencingefforts led by the International Cancer Genome Consor-tium (ICGC) are likely to identify mutations in tumorsthat map to pioneer factor binding sites [88]. The func-tional implication for these mutations will also need to bedefined.

As an alternative to genetic variations, pioneer factorbinding can also associate with changes to the chromatinlandscape imposed by epigenetic modifications (Figure 2).For instance, prostate cancer progression from castration-sensitive to castration-resistant status is related to de novoFOXA1 binding favored by H3K4 methylation at sitesunique to castration-resistant prostate cancer cells [38].These new FOXA1 binding sites promote the expression ofthe ubiquitin-conjugating enzyme E2C gene (UBE2C),which is essential for the proliferation of castration-resis-tant prostate cancer cells [2,38]. Removal of H3K4 meth-ylation in these cells significantly reduced FOXA1 bindingand UBE2C expression, thereby decreasing cancer cellproliferation [38].

Taken together, these studies demonstrate that pioneerfactors are direct targets in cancer through modification oftheir coding sequence, modulation of their expression oralteration of their genomic activities (Figure 2). This canhave consequences for the initiation, proliferation or pro-gression of tumors. These characteristics make pioneerfactors very attractive from a translational point of viewfor providing prognostic and/or therapeutic values.

Pioneer factors as therapeutic targets and biomarkersProgress in the field of personalized medicine for cancer isintimately linked to the discovery of novel biomarkers forprognostic purposes and to an expansion in drugs available

to clinicians to treat patients. The list of relevant mecha-nisms that can be targeted in cancer is rapidly expandingthrough the discovery of compounds that modulate epige-netic events [89,90]. Although these compounds arecurrently restricted to modulation of histone post-transla-tional modifications (e.g. histone acetylation [90,91]) andDNA methylation [92], the central role of pioneer factors incancer makes them an attractive entry point for therapeu-tic intervention.

Pioneer factors as therapeutic targets

The activity of the pioneer factor PU.1 can be modulated toimpact the growth of different subtypes of AMLs. Forexample, PU.1 expression is required for the expressionof the colony stimulating factor 1 receptor (CSF1R) gene inthe AML subtype harboring the (MOZ)-TIF2 translocation[93]. CSF1R expression allows for maintenance of theleukemia stem cell population that gives rise to (MOZ)-TIF2 AML. Depletion of PU.1 blocks CSF1R expression,leading to a decrease in the leukemia stem cell populationand, therefore, in leukemogenesis [93]. By contrast, in theAML subtype lacking the (MOZ)-TIF2 fusion protein, PU.1acts as a pioneer factor to promote the genomic activity oftranscription factors promoting differentiation [20]. Ectop-ic PU.1 expression can also promote the differentiation ofhighly proliferative myeloid cells into nondividing maturemacrophage-like cells prone to apoptosis [94]. Interesting-ly, vitamin K2, which promotes PU.1 upregulation inmyeloid progenitor cells, induces differentiation and apo-ptosis in AML cell lines [95].

FOXA1 has also been proposed as a therapeutic targetowing to its role in ERa signaling in breast cancer [96].Unfortunately, therapeutic approaches to antagonizeFOXA1 activity are still missing. However, PBX1 mightbe an actionable target in cancer. Its genomic activity hasbeen linked to its capacity to interact with HOX factors[97]. Recently, small peptides designed to antagonize theinteractions between PBX1 and HOX factors were shownto promote apoptosis efficiently in melanoma, ovarian andlung cancer cell lines [98–100]. They were also revealed toblock proliferation of leukemia and pancreatic cancer cells[101]. Although more studies are required to define thespecificity of these small peptides against PBX1, recentwork using small interfering (si)RNAs against PBX1 hasdemonstrated that its depletion abrogates proliferation ofovarian and breast cancer cells both in vitro and in vivo,supporting its role as a prime candidate for intervention incancer [74].

The development of novel compounds that antagonizepioneer factor activity is likely to be impaired by theinherent difficulties of targeting proteins that localizepredominantly to the nucleus and that do not possessligand-binding abilities. Given that pioneer factors arecommonly overexpressed in cancer, leads for the develop-ment of alternative therapeutic applications may originatefrom an understanding of the mechanisms that regulatetheir expression.

Pioneer factors as prognostic biomarkers

Recent studies have revealed the prognostic potential ofpioneer factors. For example, FOXA1 acts as a biomarker

471

Box 3. Outstanding questions

� What are the remaining pioneer factors and how can they be

screened for?

� How do pioneer factors collaborate with one another?

� How do pioneer factors modulate chromatin condensation

(ex: impact on higher-order chromatin folding and long-range

chromatin loops)?

� How do pioneer factors lead to de novo deposition of epigenetic

signatures?

� How different are pioneer factor activities in normal versus

cancerous cells?

� What are the mechanisms regulating pioneer factor expression?

Review Trends in Genetics November 2011, Vol. 27, No. 11

in prostate cancer, where its overexpression is associatedwith metastasis [81]. FOXA1 and GATA-3 expressioncharacterizes luminal breast tumors and is a predictor ofhigher survival [79,80,102]. This is in agreement with theircoexpression with ERa, a clinically validated marker ofgood outcome in patients with breast cancer [103]. Simi-larly, PBX1 is a prognostic marker for ovarian cancers [74].Finally, the prognostic value of the pioneer factor PU.1 hasrecently been proposed. Indeed, PU.1 expression levelspositively correlate with remission in chronic myeloidleukemia (CML) [104]. Hence, although the study of pio-neer factors is still in its infancy, it conceals the potential toderive novel translational applications.

Concluding remarksDefining the functional relationship between chromatinand transcription factors is at the heart of current under-standing of transcriptional regulation. Pioneer factors playan essential role in this process because they initiatefunctional competency of enhancers through chromatinremodeling. However, much work remains to be done toidentify all pioneer factors and their mode(s) of action (Box3). In particular, it is still not precisely understood howpioneer factors cooperate with one another to remodelchromatin and induce transcriptional competency, howthey modulate chromatin condensation or lead to de novodeposition of epigenetic signatures to define functionalregulatory elements. Finally, it will be critical to delineatethe function of pioneer factors in disease development andprogression, and to take into account how genetic variantsand mutations currently being identified through genomeresequencing efforts will impact their activity.

References1 Bushey, A.M. et al. (2008) Chromatin insulators: regulatory

mechanisms and epigenetic inheritance. Mol. Cell 32, 1–92 Chen, Z. et al. (2011) Phospho-MED1-enhanced UBE2C locus looping

drives castration-resistant prostate cancer growth. EMBO J. 30,2405–2419

3 Levine, M. (2010) Transcriptional enhancers in animal developmentand evolution. Curr. Biol. 20, R754–R763

4 Bulger, M. and Groudine, M. (2011) Functional and mechanisticdiversity of distal transcription enhancers. Cell 144, 327–339

5 Farnham, P.J. (2009) Insights from genomic profiling of transcriptionfactors. Nat. Rev. Genet. 10, 605–616

6 Graf, T. and Enver, T. (2009) Forcing cells to change lineages. Nature462, 587–594

7 Pan, Y.P. et al. (2010) Mechanisms of transcription factor selectivity.Trends Genet. 26, 75–83

8 Fussner, E. et al. (2011) Living without 30 nm chromatin fibers.Trends Biochem. Sci. 36, 1–6

472

9 Bai, L. and Morozov, A.V. (2010) Gene regulation by nucleosomepositioning. Trends Genet. 26, 476–483

10 Laganiere, J. et al. (2005) Location analysis of estrogen receptor alphatarget promoters reveals that FOXA1 defines a domain of the estrogenresponse. Proc. Natl. Acad. Sci. U.S.A. 102, 11651–11656

11 Bossard, P. and Zaret, K.S. (2000) Repressive and restrictivemesodermal interactions with gut endoderm: possible relation toMeckel’s Diverticulum. Development 127, 4915–4923

12 Cirillo, L.A. et al. (1998) Binding of the winged-helix transcriptionfactor HNF3 to a linker histone site on the nucleosome. EMBO J. 17,244–254

13 Gualdi, R. et al. (1996) Hepatic specification of the gut endoderm invitro: cell signaling and transcriptional control. Genes Dev. 10, 1670–

168214 Xu, J. et al. (2009) Transcriptional competence and the active marking

of tissue-specific enhancers by defined transcription factors inembryonic and induced pluripotent stem cells. Genes Dev. 23,2824–2838

15 Carroll, J.S. et al. (2005) Chromosome-wide mapping of estrogenreceptor binding reveals long-range regulation requiring theforkhead protein FoxA1. Cell 122, 33–43

16 Bossard, P. and Zaret, K.S. (1998) GATA transcription factors aspotentiators of gut endoderm differentiation. Development 125, 4909–

491717 Lupien, M. and Brown, M. (2009) Cistromics of hormone-dependent

cancer. Endocr. Relat. Cancer 16, 381–38918 Heintzman, N.D. et al. (2009) Histone modifications at human

enhancers reflect global cell-type-specific gene expression. Nature459, 108–112

19 Lupien, M. et al. (2008) FoxA1 translates epigenetic signaturesinto enhancer-driven lineage-specific transcription. Cell 132, 958–

97020 Heinz, S. et al. (2010) Simple combinations of lineage-determining

transcription factors prime cis-regulatory elements required formacrophage and B cell identities. Mol. Cell 38, 576–589

21 Odom, D.T. et al. (2007) Tissue-specific transcriptional regulation hasdiverged significantly between human and mouse. Nat. Genet. 39,730–732

22 Schmidt, D. et al. (2010) Five-vertebrate ChIP-seq reveals theevolutionary dynamics of transcription factor binding. Science 328,1036–1040

23 Birney, E. et al. (2007) Identification and analysis of functionalelements in 1% of the human genome by the ENCODE pilotproject. Nature 447, 799–816

24 Koch, C.M. et al. (2007) The landscape of histone modifications across1% of the human genome in five human cell lines. Genome Res. 17,691–707

25 Heintzman, N.D. et al. (2007) Distinct and predictive chromatinsignatures of transcriptional promoters and enhancers in thehuman genome. Nat. Genet. 39, 311–318

26 Aday, A.W. et al. (2011) Identification of cis regulatory features in theembryonic zebrafish genome through large-scale profiling ofH3K4me1 and H3K4me3 binding sites. Dev. Biol. DOI: 10.1016/J.YDBIO.2011.03.007

27 Rada-Iglesias, A. et al. (2011) A unique chromatin signature uncoversearly developmental enhancers in humans. Nature 470, 279–283

28 Creyghton, M.P. et al. (2010) Histone H3K27ac separates active frompoised enhancers and predicts developmental state. Proc. Natl. Acad.Sci. U.S.A. 107, 21931–21936

29 Ghisletti, S. et al. (2010) Identification and characterization ofenhancers controlling the inflammatory gene expression programin macrophages. Immunity 32, 317–328

30 Feng, R. et al. (2008) PUA and C/EBP alpha/beta convert fibroblastsinto macrophage-like cells. Proc. Natl. Acad. Sci. U.S.A. 105, 6057–

606231 Serandour, A.A. et al. (2011) Epigenetic switch involved in activation

of pioneer factor FOXA1-dependent enhancers. Genome Res. 21, 555–

56532 Taube, J.H. et al. (2010) Foxa1 functions as a pioneer transcription

factor at transposable elements to activate AFP during differentiationof embryonic stem cells. J. Biol. Chem. 285, 16135–16144

33 Xu, J. et al. (2007) Pioneer factor interactions and unmethylated CpGdinucleotides mark silent tissue-specific enhancers in embryonic stemcells. Proc. Natl. Acad. Sci. U.S.A. 104, 12377–12382

Review Trends in Genetics November 2011, Vol. 27, No. 11

34 Cirillo, L.A. et al. (2002) Opening of compacted chromatin by earlydevelopmental transcription factors HNF3 (FoxA) and GATA-4. Mol.Cell 9, 279–289

35 Sekiya, T. et al. (2009) Nucleosome-binding affinity as a primarydeterminant of the nuclear mobility of the pioneer transcriptionfactor FoxA. Genes Dev. 23, 804–809

36 Wang, D. et al. (2011) Reprogramming transcription by distinctclasses of enhancers functionally defined by eRNA. Nature 474,390–394

37 Eeckhoute, J. et al. (2009) Cell-type selective chromatin remodelingdefines the active subset of FOXA1-bound enhancers. Genome Res. 19,372–380

38 Wang, Q. et al. (2009) Androgen receptor regulates a distincttranscription program in androgen-independent prostate cancer.Cell 138, 245–256

39 Hoffman, B.G. et al. (2010) Locus co-occupancy, nucleosomepositioning, and H3K4me1 regulate the functionality of FOXA2-,HNF4A-, and PDX1-bound loci in islets and liver. Genome Res. 20,1037–1051

40 He, H.S.H.S. et al. (2010) Nucleosome dynamics define transcriptionalenhancers. Nat. Genet. 42, 343–347

41 Barski, A. et al. (2007) High-resolution profiling of histonemethylations in the human genome. Cell 129, 823–837

42 Hurtado, A. et al. (2011) FOXA1 is a key determinant of estrogenreceptor function and endocrine response. Nat. Genet. 43, 27–42

43 Fu, Y.T. et al. (2008) The insulator binding protein CTCF positions 20nucleosomes around its binding sites across the human genome. PLoSGenet. 4, e1000138

44 Sabo, P.J. et al. (2006) Genome-scale mapping of DNase Isensitivity in vivo using tiling DNA microarrays. Nat. Methods3, 511–518

45 Boyle, A.P. et al. (2008) High-resolution mapping andcharacterization of open chromatin across the genome. Cell 132,311–322

46 Giresi, P.G. et al. (2007) FAIRE (formaldehyde-assisted isolation ofregulatory elements) isolates active regulatory elements from humanchromatin. Genome Res. 17, 877–885

47 Razin, S.V. et al. (2007) Chromatin domains and regulation oftranscription. J. Mol. Biol. 369, 597–607

48 Winkler, D.D. and Luger, K. (2011) The histone chaperone Fact:structural insights and mechanisms for nucleosome reorganization.J. Biol. Chem. 286, 18369–18374

49 Hargreaves, D.C. and Crabtree, G.R. (2011) ATP-dependentchromatin remodeling: genetics, genomics and mechanisms. CellRes. 21, 396–420

50 John, S. et al. (2011) Chromatin accessibility pre-determinesglucocorticoid receptor binding patterns. Nat. Genet. 43, 264–268

51 Wu, J.I. et al. (2009) Understanding the words of chromatinregulation. Cell 136, 200–206

52 Krysinska, H. et al. (2007) A two-step, PU.1-dependent mechanism fordevelopmentally regulated chromatin remodeling and transcriptionof the c-fms gene. Mol. Cell. Biol. 27, 878–887

53 Zaret, K.S. (2002) Regulatory phases of early liver development:paradigms of organogenesis. Nat. Rev. Genet. 3, 499–512

54 Yan, J.Z. et al. (2006) The forkhead transcription factor FoxI1 remainsbound to condensed mitotic chromosomes and stably remodelschromatin structure. Mol. Cell. Biol. 26, 155–168

55 Eeckhoute, J. et al. (2009) Defining specificity of transcription factorregulatory activities. J. Cell Sci. 122, 4027–4034

56 Cuesta, I. et al. (2007) The forkhead factor FoxE1 binds to thethyroperoxidase promoter during thyroid cell differentiation andmodifies compacted chromatin structure. Mol. Cell. Biol. 27, 7302–

731457 Hatta, M. and Cirillo, L.A. (2007) Chromatin opening and stable

perturbation of core histone:DNA contacts by FoxO1. J. Biol.Chem. 282, 35583–35593

58 Berkes, C.A. et al. (2004) Pbx marks genes for activation by MyoDindicating a role for a homeodomain protein in establishing myogenicpotential. Mol. Cell 14, 465–477

59 Holmes, K.A. et al. (2011) Transducin-like enhancer protein 1mediates estrogen receptor binding and transcriptional activity inbreast cancer cells. Proc. Natl. Acad. Sci. U.S.A. DOI: 10.1073/pnas.1018863108

60 Shoemaker, J. et al. (2006) GATA-3 directly remodels the IL-10 locusindependently of IL-4 in CD4+ T cells. J. Immunol. 176, 3470–3479

61 Li, Z.Y. et al. (2011) The nucleosome map of the mammalian liver. Nat.Struct. Mol. Biol. 18, U742–U746

62 Smale, S.T. (2010) Pioneer factors in embryonic stem cells anddifferentiation. Curr. Opin. Genet. Dev. 20, 519–526

63 Tan, S.K. et al. (2011) AP-2g regulates estrogen receptor-mediatedlong-range chromatin interaction and gene transcription. EMBO J.30, 2569–2581

64 Liber, D. et al. (2010) Epigenetic priming of a pre-B cell-specificenhancer through binding of Sox2 and Foxd3 at the ESC stage.Cell Stem Cell 7, 114–126

65 Burda, P. et al. (2010) The role of PU.1 and GATA-1 transcriptionfactors during normal and leukemogenic hematopoiesis. Leukemia 24,1249–1257

66 Sorlie, T. et al. (2001) Gene expression patterns of breast carcinomasdistinguish tumor subclasses with clinical implications. Proc. Natl.Acad. Sci. U.S.A. 98, 10869–10874

67 Meissner, A. (2010) Epigenetic modifications in pluripotent anddifferentiated cells. Nat. Biotechnol. 28, 1079–1088

68 Bernstein, B.E. et al. (2006) A bivalent chromatin structure marks keydevelopmental genes in embryonic stem cells. Cell 125, 315–326

69 Meissner, A. et al. (2008) Genome-scale DNA methylation maps ofpluripotent and differentiated cells. Nature 454, 766–770

70 Mikkelsen, T.S. et al. (2008) Dissecting direct reprogramming throughintegrative genomic analysis. Nature 454, 49–55

71 Mueller, B.U. et al. (2002) Heterozygous PU.1 mutations areassociated with acute myeloid leukemia. Blood 100, 998–1007

72 Aspland, S.E. et al. (2001) The role of E2A-PBX1 in leukemogenesis.Oncogene 20, 5708–5717

73 Xiao, Y. et al. (2011) The lymphovascular embolus of inflammatorybreast cancer exhibits a Notch 3 addiction. Oncogene 30, 287–300

74 Park, J.T. et al. (2008) Identification of Pbx1, a potential oncogene, asa Notch3 target gene in ovarian cancer. Cancer Res. 68, 8852–8860

75 Kikugawa, T. et al. (2006) PLZF regulates Pbx1 transcription andPbx1-HoxC8 complex leads to androgen-independent prostate cancerproliferation. Prostate 66, 1092–1099

76 Gee, J.M.W. et al. (2009) Overexpression of TFAP2C in invasivebreast cancer correlates with a poorer response to anti-hormonetherapy and reduced patient survival. J. Pathol. 217, 32–41

77 Friedrichs, N. et al. (2005) Distinct spatial expression patterns of AP-2alpha and AP-2gamma in non-neoplastic human breast and breastcancer. Mod. Pathol. 18, 431–438

78 Williams, C.M. et al. (2009) AP-2gamma promotes proliferation inbreast tumour cells by direct repression of the CDKN1A gene. EMBOJ. 28, 3591–3601

79 Badve, S. et al. (2007) FOXA1 expression in breast cancer: correlationwith luminal subtype A and survival. Clin. Cancer Res. 13, 4415–

442180 Thorat, M.A. et al. (2008) Forkhead box A1 expression in breast cancer

is associated with luminal subtype and good prognosis. J. Clin.Pathol. 61, 327–332

81 Jain, R.K. et al. (2011) High-level expression of forkhead-box proteinA1 in metastatic prostate cancer. Histopathology 58, 766–772

82 Hu, X. et al. (2009) Genetic alterations and oncogenic pathwaysassociated with breast cancer subtypes. Mol. Cancer Res. 7, 511–

52283 Carroll, J.S. et al. (2006) Genome-wide analysis of estrogen receptor

binding sites. Nat. Genet. 38, 1289–129784 Nucera, C. et al. (2009) FOXA1 is a potential oncogene in anaplastic

thyroid carcinoma. Clin. Cancer Res. 15, 3680–368985 Jia, L. et al. (2009) Functional enhancers at the gene-poor 8q24

cancer-linked locus. PLoS Genet. 5, e100059786 Lu, Y. et al. (2011) Functional annotation of risk loci identified

through genome-wide association studies for prostate cancer.Prostate 71, 955–963

87 Siva, N. (2008) 1000 Genomes project. Nat. Biotechnol. 26, 25688 Hudson, T.J. et al. (2010) International network of cancer genome

projects. Nature 464, 993–99889 Sahin, M. et al. (2010) DNA methylation or histone modification

status in metastasis and angiogenesis-related genes: a newhypothesis on usage of DNMT inhibitors and S-adenosylmethioninefor genome stability. Cancer Metastasis Rev. 29, 655–676

473

Review Trends in Genetics November 2011, Vol. 27, No. 11

90 Carafa, V. et al. (2011) Histone deacetylase inhibitors: recent insightsfrom basic to clinical knowledge & patenting of anti-cancer actions.Recent Patents Anti-Cancer Drug Discov. 6, 131–145

91 Tan, J.H. et al. (2010) Novel histone deacetylase inhibitors in clinicaltrials as anti-cancer agents. J. Hematol. Oncol. 3, 5

92 Gronbaek, K. et al. (2008) Epigenetic changes in cancer as potentialtargets for prophylaxis and maintenance therapy. Basic Clin.Pharmacol. Toxicol. 103, 389–396

93 Aikawa, Y. et al. (2010) PU.1-mediated upregulation of CSF1R iscrucial for leukemia stem cell potential induced by MOZ-TIF2. Nat.Med. 16, 580–585

94 Durual, S. et al. (2007) Lentiviral PU.1 overexpression restoresdifferentiation in myeloid leukemic blasts. Leukemia 21, 1050–1059

95 Sada, E. et al. (2010) Vitamin K2 modulates differentiation andapoptosis of both myeloid and erythroid lineages. Blood 116, 1627

96 Nakshatri, H. and Badve, S. (2007) FOXA1 as a therapeutic target forbreast cancer. Expert Opin. Ther. Targets 11, 507–514

97 Moens, C.B. and Selleri, L. (2006) Hox cofactors in vertebratedevelopment. Dev. Biol. 291, 193–206

98 Morgan, R. et al. (2010) Targeting HOX and PBX transcription factorsin ovarian cancer. BMC Cancer 10, 89

474

99 Plowright, L. et al. (2009) HOX transcription factors are potentialtherapeutic targets in non-small-cell lung cancer (targeting HOXgenes in lung cancer). Br. J. Cancer 100, 470–475

100 Morgan, R. et al. (2007) Antagonism of HOX/PBX dimer formationblocks the in vivo proliferation of melanoma. Cancer Res. 67, 5806–5813

101 Aulisa, L. et al. (2009) Inhibition of cancer cell proliferation bydesigned peptide amphiphiles. Acta Biomaterialia 5, 842–853

102 Ademuyiwa, F.O. et al. (2010) Expression of Forkhead-box protein A1,a marker of luminal A type breast cancer, parallels low oncotype DX21-gene recurrence scores. Mod. Pathol. 23, 270–275

103 Yoon, N.K. et al. (2010) Higher levels of GATA3 predict better survivalin women with breast cancer. Hum. Pathol. 41, 1794–1801

104 Albajar, M. et al. (2008) PU.1 expression is restored upon treatment ofchronic myeloid leukemia patients. Cancer Lett. 270, 328–336

105 van Steensel, B. (2011) Chromatin: constructing the big picture.EMBO J. 30, 1885–1895

106 Lee, C.S. et al. (2005) The initiation of liver development is dependenton Foxa transcription factors. Nature 435, 944–947

107 Mcpherson, C.E. et al. (1993) An active tissue-specific enhancer andbound transcription factors existing in a precisely positionednucleosomal array. Cell 75, 387–398