Genomic and Biochemical Insights into the Specificity of ...

39
Genomic and Biochemical Insights into the Specificity of ETS Transcription Factors Peter C. Hollenhorst, 1 Lawrence P. McIntosh, 2 and Barbara J. Graves 3,4 1 Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana 47405; email: [email protected] 2 Department of Biochemistry and Molecular Biology, Department of Chemistry, and The Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3; email: [email protected] 3 Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah Health Sciences, Salt Lake City, Utah 84112; email: [email protected] 4 Howard Hughes Medical Institute, Chevy Chase, Maryland 20815 Annu. Rev. Biochem. 2011. 80:437–71 First published online as a Review in Advance on May 3, 2011 The Annual Review of Biochemistry is online at biochem.annualreviews.org This article’s doi: 10.1146/annurev.biochem.79.081507.103945 Copyright c 2011 by Annual Reviews. All rights reserved 0066-4154/11/0707-0437$20.00 Keywords autoinhibition, ChIP-Seq, DNA binding, ETS domain, PNT domain, protein dynamics Abstract ETS proteins are a group of evolutionarily related, DNA-binding tran- scriptional factors. These proteins direct gene expression in diverse nor- mal and disease states by binding to specific promoters and enhancers and facilitating assembly of other components of the transcriptional ma- chinery. The highly conserved DNA-binding ETS domain defines the family and is responsible for specific recognition of a common sequence motif, 5 -GGA(A/T)-3 . Attaining specificity for biological regulation in such a family is thus a conundrum. We present the current knowledge of routes to functional diversity and DNA binding specificity, includ- ing divergent properties of the conserved ETS and PNT domains, the involvement of flanking structured and unstructured regions appended to these dynamic domains, posttranslational modifications, and protein partnerships with other DNA-binding proteins and coregulators. The review emphasizes recent advances from biochemical and biophysical approaches, as well as insights from genomic studies that detect ETS- factor occupancy in living cells. 437 Annu. Rev. Biochem. 2011.80:437-471. Downloaded from www.annualreviews.org by University of British Columbia on 09/16/11. For personal use only.

Transcript of Genomic and Biochemical Insights into the Specificity of ...

Page 1: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

Genomic and BiochemicalInsights into the Specificityof ETS Transcription FactorsPeter C. Hollenhorst,1 Lawrence P. McIntosh,2

and Barbara J. Graves3,4

1Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana 47405;email: [email protected] of Biochemistry and Molecular Biology, Department of Chemistry, and TheMichael Smith Laboratories, University of British Columbia, Vancouver, British Columbia,Canada V6T 1Z3; email: [email protected] of Oncological Sciences, Huntsman Cancer Institute, University of UtahHealth Sciences, Salt Lake City, Utah 84112; email: [email protected] Hughes Medical Institute, Chevy Chase, Maryland 20815

Annu. Rev. Biochem. 2011. 80:437–71

First published online as a Review in Advance onMay 3, 2011

The Annual Review of Biochemistry is online atbiochem.annualreviews.org

This article’s doi:10.1146/annurev.biochem.79.081507.103945

Copyright c© 2011 by Annual Reviews.All rights reserved

0066-4154/11/0707-0437$20.00

Keywords

autoinhibition, ChIP-Seq, DNA binding, ETS domain, PNT domain,protein dynamics

Abstract

ETS proteins are a group of evolutionarily related, DNA-binding tran-scriptional factors. These proteins direct gene expression in diverse nor-mal and disease states by binding to specific promoters and enhancersand facilitating assembly of other components of the transcriptional ma-chinery. The highly conserved DNA-binding ETS domain defines thefamily and is responsible for specific recognition of a common sequencemotif, 5′-GGA(A/T)-3′. Attaining specificity for biological regulationin such a family is thus a conundrum. We present the current knowledgeof routes to functional diversity and DNA binding specificity, includ-ing divergent properties of the conserved ETS and PNT domains, theinvolvement of flanking structured and unstructured regions appendedto these dynamic domains, posttranslational modifications, and proteinpartnerships with other DNA-binding proteins and coregulators. Thereview emphasizes recent advances from biochemical and biophysicalapproaches, as well as insights from genomic studies that detect ETS-factor occupancy in living cells.

437

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 2: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

Contents

INTRODUCTION. . . . . . . . . . . . . . . . . 438Evolution of the ETS Family . . . . . 439Biochemical and Biological

Diversity . . . . . . . . . . . . . . . . . . . . . . 439Extensive Coexpression . . . . . . . . . . . 440

ETS DOMAIN: BIOCHEMICALOVERVIEW OF DNA BINDNGAND AUTOINHIBITION . . . . . . 441Structure and DNA-Binding

Interface . . . . . . . . . . . . . . . . . . . . . . 441DNA-Sequence Preferences . . . . . . 442Structural Elements Appended

to the ETS Domain . . . . . . . . . . . 443Autoinhibition of ETS1 DNA

Binding: A DynamicMechanism . . . . . . . . . . . . . . . . . . . 444

Autoinhibition in Other ETSProteins . . . . . . . . . . . . . . . . . . . . . . 446

PNT DOMAIN: DIVERSEFUNCTION ANDREGULATION . . . . . . . . . . . . . . . . . 448PNT-Domain Structure and

Appended Elements . . . . . . . . . . . 448Ras Signal Transduction and

MAP-Kinase Docking . . . . . . . . . 448Role of the PNT Domain in

Phosphorylation-EnhancedInteractions of ETS1and CBP . . . . . . . . . . . . . . . . . . . . . . 449

PNT-Domain Self-Association. . . . 450GENOMIC APPROACHES TO

THE SPECIFICITYQUESTION: REDUNDANTAND SPECIFIC BINDING . . . . . 450Redundant Binding of ETS

Proteins in Living Cells . . . . . . . . 451Specific Binding of ETS Proteins

in Living Cells . . . . . . . . . . . . . . . . 453COOPERATIVE PARTNERSHIPS

PROVIDE ETS FAMILYSPECIFICITY: GENOMICAND BIOCHEMICALPERSPECTIVES . . . . . . . . . . . . . . . . 454Ternary Complex Factors/Serum

Response Factor . . . . . . . . . . . . . . . 454SPI1/IRF4 . . . . . . . . . . . . . . . . . . . . . . . 455ETS1/RUNX1 and

Autoinhibition . . . . . . . . . . . . . . . . 455ETS1/PAX5 . . . . . . . . . . . . . . . . . . . . . 456ETS/FOX . . . . . . . . . . . . . . . . . . . . . . . 456ETV6 Self-Association and

Autoinhibition . . . . . . . . . . . . . . . . 456Other Dimeric Configurations . . . . 457

GENOMIC INSIGHTS INTOONCOGENIC ETSFACTORS . . . . . . . . . . . . . . . . . . . . . . 458ERG and Androgen Receptor in

Prostate Cancer . . . . . . . . . . . . . . . 458EWS-FLI1 in Ewing’s Sarcoma . . . 458

ETS domain:conserved structuraldomain in all ETSproteins that isnecessary andsufficient for DNAbinding

PNT domain:structural domainconserved inapproximatelyone-third of ETSproteins, whichfunctions in proteininteractions andresembles the SAMdomain

INTRODUCTIONAn overarching conundrum for eukaryotictranscription factors is how a set of relatedproteins displaying highly conserved DNA-binding domains execute diverse biologicalroles. This review addresses the current state ofbiochemical and genomic investigation of thisspecificity question in the ETS family. Initially,we focus on the common and distinct DNA-binding properties of the ETS domain. Next,we discuss the PNT domain to illustrate thepotential of semiconserved domains to providea route to diverse function by different family

members. Then, we consider new studies thatprovide the first views of what is happening incells at the level of genome-wide ETS-factorDNA occupancy. We include genomic analysisof oncogenic ETS genes as these studieschallenge our thinking about specificity andredundancy. Both biochemical and genomicapproaches demonstrate the role of proteinpartnerships as a specificity mechanism. Weemphasize systems for which biochemical andbiophysical details are available because theseprovide a robust framework both for definingthe specificity problem and informing the many

438 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 3: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

SRF: serum responsefactor

Intrinsicallydisordered proteinsequences: regions ofproteins that arebiologically active, yetlack a well-definedthree-dimensionalstructure underphysiologicalconditions

experimental investigations aimed at under-standing the role of ETS factors in biologicalregulation. To complement our structural andgenomics focus, we refer the reader to previousreviews that cover biological properties of theETS factors in more detail (1–6).

Evolution of the ETS Family

The ETS proteins are encoded by a genefamily that is defined by the conserved ETSdomain (Figure 1a,b) and present throughoutthe metazoan phyla (7). Family phylogenysuggests the duplication of an ancestral geneearly in metazoan evolution. The continuedgrowth of the family from invertebrates,represented by the 8 and 10 paralogs inDrosophila melanogaster and Caenorhabditiselegans, respectively (Supplemental Table 1:For all Supplemental Material, follow thelink from the Annual Reviews home page athttp://www.annualreviews.org), to verte-brates, represented by 28 human genes, waslikely facilitated by large-scale duplicationsof vertebrate genomic regions (8). Hallmarksof these evolutionary steps are seen in thedendogram of the family (Figure 2) as well asin the relative positions of structural domains(Figure 1a).

An understanding of the overall relatednessof ETS family members is critical for fram-ing the question of functional specificity. Forexample, there are subfamilies with higher de-grees of sequence similarity in both their ETSdomains and PNT domains (9) (Figures 1a

and 2). Closely related members of a subfam-ily may display redundant functions, whereasthose in different subfamilies may have uniquebiochemical properties that could be utilized indistinct biological pathways.

Biochemical and Biological Diversity

Structural or functional domains and posttrans-lational modifications that are found outside ofthe highly conserved ETS domain illustrate thepotential for unique properties of subfamiliesor individual ETS proteins. The PNT domain

(Figure 1a,c) has variable functions, discussedbelow, depending on the subfamily in whichit resides. The OST domain appears unique toGABPA (Figure 1a,d ) and serves in cofactorrecruitment (10). The B-box, found in thethree members of the ternary complex factor(TCF) subfamily, forms an induced structuralelement that interacts with serum responsefactor (SRF) on DNA (11). Although examinedin only a few cases by biophysical methods(12–15), the remaining regions of the ETSfactors, including those functioning as transac-tivation domains, likely display minimal stablesecondary or tertiary structure. These intrin-sically disordered regions can have numerouspossible roles, including serving as flexiblelinkers, linear peptide motifs for binding topartner proteins, and sites of posttranslationalmodifications (16). Figure 1 is annotated toshow only those examples of phosphorylationthat are covered in this review owing to theirinterplay with structured domains. Othermodifications found on ETS factors, includingubiquitinylation, sumoylation, glycosylation,and acetylation, have been reviewed elsewhere(17, 18). In conclusion, the overall diversifi-cation of the known biochemical propertiesof the family members suggests that differentfamily members will play distinct biologicalroles.

Diverse biological roles of individualETS family members are also suggested bythe broad range of phenotypes displayedin mouse genetic disruptions (Figure 2).Genetic studies in C. elegans (19) and Drosophila(20) demonstrate this same trend, with avariety of phenotypes suggesting specializedfunctions ranging from organ cell fate andneuronal development to oogenesis and aging(Supplemental Table 1) (21–26). Althoughclassic genetics approaches can be limitedas a result of potential redundancy withinthe family, this diversity strongly indicatesthat different ETS family members carry outdistinct biological roles. Indeed, recent reportsthat include analyses of transcriptional targetsimplicate specific ETS genes in development ofmammalian hematopoietic stem cells (27–29),

www.annualreviews.org • ETS Family of Transcription Factors 439

Supplemental Material

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 4: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

endothelial cells (30–32), and T cells (33).Although many putative transcriptional targetshave been identified over the past 20 years forETS family members through experimentalanalyses of transcriptional control elements byin vitro and cell-based assays (3), we focus onlyon genome-wide occupancy approaches andevaluate their contribution to the evaluation ofthe authentic targets of ETS factors.

Extensive CoexpressionIn spite of the biological and biochemical evi-dence that different ETS proteins can executediverse roles, an extensive overlap in the ex-pression of various family members is found inmany human tissues. Coexpression of the ETSgenes has been systematically surveyed at themRNA level in more than 20 tissues and trans-formed cell lines (34–35). Half of the family

ETV1 (ER81)

ETV4 (PEA3, E1AF)

ETV5 (ERM)

FEV

ERF

ETV3 (PE1, PEP1, METS)

ETV3L

ETV2 (ER71)

ELF1

ELF2 (NERF)

ELF4 (MEF)

SPIB

SPIC

ETV6 (TEL)

ETV7 (TEL2)

FLI1

ERG

ELF3 (ESE1, ESX)

ELF5 (ESE2)

EHF (ESE3)

SPDEF (PDEF)

ETV6/RUNX1 (TEL/AML1)

EWS/FLI1

GABPA (GABPa, E4TF, ELG)

Human ETS family

ETS fusion oncoproteins

ELK1 P

ELK3 (NET, SAP2, ERP) P

ELK4 (SAP1) P

a

G

G

A A

NC

b

N

C

c

N

C

d

ETS1 P P

ETS2 P P

SPI1 (PU.1, Sfpi1) P

ETS domain PNT domain OST domain B-box C-terminal RUNX1 N-terminal EWS

ETS1 ETS domain

ERGPNT domain

GABPAOST domain

Carbon Oxygen Nitrogen

440 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 5: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

members are ubiquitously expressed in all celltypes tested. In any particular cell type, 14to 25 family members are coexpressed. Thischallenges the simplest notion that each familymember is restricted to a tissue or cell type fora site of action. In some cases, however, high-level expression is confined to a single cell type(e.g., SPDEF in the prostate, SPI1 in myeloidcells). Additional levels of regulation beyondmRNA expression, such as translational con-trol, alternative splicing, posttranslational mod-ifications, cellular localization, and protein sta-bility, will add to the complexity of this picture.Therefore, systematic proteomic studies thatdetermine active protein levels will be neces-sary to describe the relative prevalence of ETSfactors in diverse tissues.

Expression profiles in transformed cell linesshow strong similarity to those in matching nor-mal tissues, although exceptions are worth not-ing. There is high expression of SPDEF andESE subfamily members in mammary tumors(35), and a striking prevalence of ETV4 expres-sion in cell lines versus normal matching adulttissues (34). The high expression levels of ERGand PEA3 subfamily members in a subset ofprostate tumors led to the discovery of com-mon chromosomal rearrangements associatedwith this human cancer (36). Thus, relative dif-ferences in expression of ETS genes may havea significant role in both normal biology andcancer.

ETS DOMAIN: BIOCHEMICALOVERVIEW OF DNA BINDNGAND AUTOINHIBITION

Structure and DNA-Binding Interface

The overall structure and DNA-bindingmechanism of the ETS domain is based onhigh-resolution molecular models of ten familymembers derived from both X-ray crystal-lography and nuclear magnetic resonance(NMR) spectroscopy (Figure 1b, Supple-mental Table 2) (37–51). The conserved ETSdomain consists of three α-helices on a smallfour-stranded, antiparallel β-sheet scaffold.This variant of the prototypic helix-turn-helixDNA-binding motif, termed the wingedhelix architecture, is also found in eukaryoticFOX proteins, heat shock factors, and linkerhistones as well as the prokaryotic CAP protein(52). The ETS domain binds DNA over aregion spanning 12 to 15 base pairs (bp), butit displays sequence preference for only ∼9 bp(numbered 1–9 in this review) with a central,invariant 5′-GGA(A/T)-3′ core. The interfaceis characterized both by direct readout, whichinvolves base-pair contact by ETS-domainsidechains, and indirect readout, which utilizesthe sequence-dependent positioning of thephosphodiester backbone. The most importantdirect interactions in the major groove involvehydrogen bonding between the two invariantarginines in the recognition helix H3 and the

←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−Figure 1Structural and functional domains of the ETS family of transcription factors. (a) Nomenclature and domain organization of the 28paralogous human ETS proteins (grouped according to the dendogram of Figure 2). We use HUGO nomenclature (191) for all ETSgenes and proteins, with alternative names provided. Oncogenic ETS proteins, which are created by human chromosomerearrangements, are illustrated by two examples. Many ETS genes produce multiple protein products via alternative splicing/start sites,and in these cases, a single polypeptide was chosen arbitrarily. Boxes identify the DNA-binding ETS domain (red ), PNT domain( green), OST domain (blue), and B-box (magenta) of the ETS factors as well as the C-terminal portion of RUNX1 (light brown) and theN-terminal portion of EWS (dark brown). The circled P symbolizes a phosphorylated region discussed in the text. Not identified areadditional regions involved in transcriptional activation/repression, posttranslational modifications (including kinase docking andsumoylation), nuclear import/export, turnover, and so forth. (b) Ribbon diagram of the ETS1 ETS domain bound to a DNA duplex(1k79.pdb with the inhibitory helices deleted for clarity). The side chains of R391, R394, and Y395, which provide base-specificcontacts to core GGAA, are also shown in stick format. Ribbon diagrams of (c) the ERG PNT domain (1sxe.pdb) and (d ) the GABPAOST domain (1juo.pdb). See Supplemental Tables 2 and 3 for a referenced summary of all published ETS and PNT domainstructures. Molecular diagrams were rendered with PyMOL (http://pymol.org) with coordinate files from the Protein Data Bank(http://pdb.org).

www.annualreviews.org • ETS Family of Transcription Factors 441

Supplemental Material

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 6: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

PEA3

TCF

ELF

ESE

SPI

TEL

ETS

ERG

ERF

Defective in mouse deletionETS geneETS subfamily

SPI1 Lethal, myeloid, B cells (197-203)

SPIB B cells (204)

SPIC Red pulp macrophages (205)

ETV6 Yolk sac, hematopoietic stem cells (206,207)

ETV7 N/A, no mouse ortholog ELF3 Intestine (208)

EHF N/AELF5 Lethal, embryo patterning, mammary gland (209,210)

ELF4 NK cells, NK-T cells (211)

ELF2 N/AELF1 No defect (212)

SPDEF Intestine (213,214)

ETV1 Neuronal connections (215)

ETV5 Male fertility (216)

ETV4 Male fertility, motor neurons, mammary gland (217,218)

ELK3 Vasculature (219)

ELK4 Thymocytes, peripheral T cells (220)

ELK1 No defect (221)

ETS1 T cells, NK cells, NK-T cells (159,222,223)

ETS2 Placenta, hair follicles (224)

ETV2 Lethal, blood, vasculature (225,226)

ETV3 N/AN/AETV3L

ERF Placenta (227) ERG Definitive hematopoiesis* (194)

FLI1 Vasculature, megakaryocytes, B cells (228-231)

FEV 5-HT neurons (232,233)

GABPA Lethal, T cells (234)

Class I Class II Class III Class IV

Figure 2Diversity in function of ETS family. (left) A dendogram shows the degree of relatedness of 28 human ETS domain sequences, built byClustalW (192) using the neighbor-joining method (193). The length of each horizontal line indicates estimated evolutionary distance.Branches that separate an individual subfamily are labeled in red. Classes defined by differences in the in vitro derived binding site (62)are indicated by background colors. (right) A list of selected tissues or functions that are defective in each mouse gene deletion, ascataloged at http://www.knockoutmouse.org. See also citations 159 and 197–234, which are linked to defects in mouse deletion infigure. For the sake of brevity, some phenotypes are not listed. N/A indicates the gene deletion has not been reported. The asteriskindicates that this phenotype results from a point mutation of ERG that affects transactivation ability rather than a gene deletion (194).Mice with deletions or mutations of two members of a subfamily have phenotypes that indicate overlapping roles in endothelial cells(ETS1 and ETS2) (30), hematopoietic stem cells (ERG and FLI1) (28), thymocyte development (ELK1 and ELK4) (33), B-cellreceptor signal transduction (SPI1 and SPIB) (195), and limb-bud development (ETV1 and ETV5) (196).

two guanines of the GGA(A/T) core. Addi-tional direct and water-mediated hydrogenbonding, hydrophobic, and electrostatic inter-actions involved in the interface are providedprimarily by residues in the “turn” between he-lices H2 and H3, the “wing” between strands S3and S4, and the N terminus of helix H1. Strik-ingly, there are no direct contacts to any basepair outside of the central GGA(A/T), thus im-plicating sequence-dependent backbone con-figurations for specificity in the flanking regions(53). Indeed, thermodynamic studies revealedan ∼400-fold variation of the affinity of SPI1depending on sequences flanking the GGA

core (54). The structural basis for this indirectreadout is currently not well defined, althoughDNA duplexes bound by ETS domains are typ-ically distorted from an ideal B-DNA geometry.

DNA-Sequence Preferences

The selectivity of multiple ETS proteins hasbeen determined by in vitro methods thatdemonstrate a binding preference for a partic-ular DNA sequence. Ten proteins were assayedin independent experiments in a variety oflaboratories that selected preferred sequencesfrom pools of DNA duplexes, using either the

442 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 7: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

SELEX (systematic evolution of ligands byexponential enrichment) or length-encodedmultiplex approach (53, 55–61). More recently,26 mouse and 27 human ETS proteins weresubjected to either microwell binding assaysor protein-binding microarrays to determinesequence preferences (62). The results of suchanalyses performed by different laboratoriesand across the entire ETS family are strik-ing in their similarity. The most commonhigh-affinity, consensus sequence identified,ACCGGAAGT, consists of an invariant GGAcore surrounded by positions with lowerpreference (see also Genomic Approaches tothe Specificity Question, section below).

Sequence preferences observed in system-atic analysis of the whole family suggestfour classes of ETS proteins that track withsubfamilies (Figure 2) (62). Class I, con-taining more than half of the ETS family(PEA3, TCF, ETS, ERF, and ERG subfam-ilies), displays a consensus sequence identicalto the most common in vitro–derived sequence(ACCGGAAGT). Class II contains eight mem-bers (TEL, ESE, and ELF subfamilies) and hasa consensus that differs primarily in the firstnucleotide (CCCGGAAGT). Members of thissecond group tend also to be more sensitiveto the adenine-to-thymine substitution at po-sition 7, as has been previously reported forELF1 (63–65). However, variation in the affin-ity for the alternative GGAT core can also oc-cur between Class I members, such as ELK1and ELK4 (58). Class III is composed of onlythe SPI subfamily, whose members prefer bind-ing sites with an adenine-rich sequence 5′ tothe GGA (57). Class IV includes only SPDEF,which has a unique preference for a GGATcore sequence instead of GGAA (66). Quan-titative DNA-binding studies indicate that theaffinity of different ETS proteins for the sameconsensus sequence many vary by only ∼10-fold, whereas the affinity of the same proteinfor variant consensus sites differs by ∼5- to 80-fold (53, 56, 65).

A rationale for some sequence preferencescomes from detailed biochemical investi-gations. For example, two TCF subfamily

members, ELK1 and ELK4, have identicalDNA-recognition helices, but detailed crys-tallographic and mutagenic studies revealedthat residues at the DNA-binding interface,including a key tyrosine in helix H3, differconformationally owing to distinct tertiarycontacts with adjacent, nonconserved residues(41, 42). Collectively, these effects restrict theability of ELK1, but not ELK4, to bind vari-ants of the ETS consensus sequence includingGGAT.

In conclusion, the differences in DNA-sequence preference of most ETS proteinsidentified from in vitro studies are subtle. Fur-thermore, these sequences, which were selectedowing to their relative affinity in vitro, could oc-cur rarely in transcriptional-control elementsas a result of this lack of selectivity. Speci-ficity in cells could derive predominantly frominteractions with cofactors, including partnertranscription factors, as discussed below. Loweraffinity sites that vary from the high-affinityselected sequences could provide more speci-ficity by poising ETS factors to be dependenton these cofactors.

Structural Elements Appendedto the ETS Domain

Structural elements appended to the ETS do-main regulate its function and demonstrate di-versification in the family (Figures 3a and 4).In several ETS proteins, a short α- or 310-helix, or in the case of ELF3 an ordered coilsequence, lies immediately C terminal to the fi-nal β-strand of the ETS domain. This may bean even more common feature than currentlyrecognized because the analogous C-terminalregions have not been characterized structurallyfor all family members. These flanking residuescontribute to the tertiary structure and stabil-ity of several ETS domains (51, 67). GABPA,ETS1, ETS2, and ETV6 have two C-terminalhelices, termed H4 and H5. In the case ofGABPA, these serve as a platform for bind-ing GABPB1 (GABPβ), which indirectly en-hances DNA affinity, slowing the half-time fordissociation by at least 100-fold (Figure 4a)

www.annualreviews.org • ETS Family of Transcription Factors 443

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 8: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

H1 S1 S2 H2 H3 S3 S4 H4 H5HI-2HI-1

H0 H1 H2 H3 H4 H5H2'

ETS domain

**

ETS1, ETS2

GABPA, ETV6

ELK4

FLI1, SPI1, SPDEF, ELF3, ELF5, ELK1

ERG, FLI1, ETV6, Yan

GABPA, SPDEF

ETS1, ETS2, Pnt-P2

PNT domain

SAM domain

H1

S1

S2

H2

H3S3

S4

H4

H5

HI-2

HI-1

H0

H1

H2H3

H4

H5

H2'

**

a

b

SRR

N

PNT

C

N

ETS

Figure 3Appended structures to the conserved ETS and PNT domains expand regulatory potential. (a) The core ETS domain (red ) containsthree α-helices (rectangles) on a four-stranded, antiparallel β-sheet (arrows). Appended N- and C-terminal helices (cyan), as well as anordered C-terminal coil region in ELF3 (not shown) and dynamic sequences such as the serine-rich region (SRR) of ETS1, provideadditional functional diversity. Shown (right) is a ribbon diagram of residues 301–441 of ETS1 (1r36.pdb), with the appended helices(cyan) packed against helix H1 of the core ETS domain. Helices HI-1, HI-2, H4, and H5 inhibit DNA binding of ETS1. Helix HI-1,which is distal from the recognition helix H3, unfolds upon DNA binding as part of the autoinhibition mechanism (Figures 4c,d and5a). (b) The core PNT domain ( green) consists of four α-helices and a small α- or 310-helix (H2′) with a fold similar to that of the SAMdomain. Although not shown for clarity, residues preceding H2 form a conserved helical-like turn. Again, appended N-terminal (H0,H1) helices (orange) provide additional routes to functional diversity. Shown (right) is the ribbon diagram of residues 29–138 of ETS1(2jv3.pdb), including the PNT domain, which constitute a docking site for both MAP kinase and the TAZ1 domain of an ETS1cofactor, CBP (Figure 5b). Two conserved phosphoacceptors (asterisk) preceding the dynamic helix H0 of ETS1 are indicated. SeeSupplemental Tables 2 and 3 for a referenced summary of all published ETS and PNT domain structures. The codes shown are fromthe Protein Data Bank (http://pdb.org).

(40). The two appended C-terminal helices inETV6 participate in DNA-binding autoinhi-bition, likely via a steric mechanism (50). InETS1 and, by sequence similarity, ETS2 andtheir Drosophila ortholog Pnt-P2, two more he-lices, HI-1 and HI-2, reside N terminal to theETS domain and pack with helices H4 and H5(Figure 3a, right) (45, 48, 68). The resultinghelical bundle is central to the allosteric au-toinhibition of DNA binding by these familymembers (discussed below).

Autoinhibition of ETS1 DNA Binding:A Dynamic MechanismSeveral ETS proteins exhibit autoinhibition ofDNA binding. Diagnostic of this phenomenonis a reduction in the overall affinity for DNA ofthe full-length protein relative to smaller frag-ments that retain the ETS domain. Autoinhi-bition offers many routes for the regulation ofproteins, including alternative splicing or pro-teolysis that would remove inhibitory elements,and posttranslational modifications or protein

444 Hollenhorst · McIntosh · Graves

Supplemental Material

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 9: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

ETS domain

Appended helices

B-box

Partner

Monomer units of ETV6 polymer

GABPA / GABPB1

ETS1 / PAX5

ETV6 / ETV6 / ETV6

ELK4 / SRF

ETS1 / ETS1

a

c

b

e

d

f

SPI1 / IRF4

Figure 4Structurally characterized ETS protein partnerships. Shown are molecular models of the ETS partnerships involving the ETS domainsof (a) GABPA with GABPB1 (1awc.pdb), (b) ELK4 with serum response factor (SRF) (Ihbx.pdb), (c) ETS1 with PAX5 (1 mdm.pdb),(d ) the ETS1 homodimer (2nny.pdb), and (e) SPI1 with IRF4 (46). In each case, the ribbon diagrams are colored as ETS domain (red ),appended helices (cyan), B-box (magenta), and partner ( yellow). ( f ) Three monomer units of an ETV6 PNT domain polymer associatedhead-to-tail ( greens; 1ji7.pdb). See Supplemental Tables 2 and 3 for a referenced summary of all published ETS and PNT domainstructures. The codes listed are from the Protein Data Bank (http://pdb.org).

www.annualreviews.org • ETS Family of Transcription Factors 445

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 10: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

partnerships that can either reinforce or antag-onize inhibition (69).

The best-characterized example of au-toinhibition among the ETS proteins is thatof ETS1, in which all the above regulatorymechanisms are at play. Autoinhibition ofETS1 DNA binding was discovered initiallyby deletion analyses (56, 70, 71). However,two naturally occurring variants confirm thebiological relevance of inhibitory sequences.The oncogenic version of ETS1 found in thep135 oncoprotein of the E26 retrovirus has theC-terminal inhibitory sequences deleted. Itshigher DNA-binding activity may contributeto oncogenesis (72). Likewise, an alternativelyspliced ETS1 isoform, which lacks the N-terminal inhibitory sequences encoded by exonVII, binds DNA with higher affinity and slightlybroader specificity (73). As discussed below,ETS1 autoinhibition is reinforced by phospho-rylation (74, 75) and counteracted by proteinpartnerships, such as those with RUNX1 (alsotermed AML1/CBFα2) and PAX5.

Detailed analyses implicate the three struc-tural components of ETS1 in the mech-anism of autoinhibition, namely the ETSdomain, the appended inhibitory module(Figure 3a), and an adjacent intrinsically dis-ordered serine-rich region (SRR) (Figure 5a).The inhibitory module comprises a helical bun-dle (HI-1, HI-2, H4, and H5) and interfaceswith H1 of the ETS domain. Collectively,these two components, the inhibitory moduleand ETS domain, are described as the regu-latable unit of ETS1. The basic framework ofETS1 autoinhibition was established by earlystudies demonstrating that HI-1 unfolds uponDNA binding, thus suggesting a thermody-namic penalty to reduce net DNA affinity (76).Subsequent structural analyses showed that theinhibitory module is distal to the DNA-bindinginterface of ETS1, indicative of an allostericmechanism linking binding and helix unfolding(45, 48).

More recent NMR-based studies have re-vealed that dynamic properties across a widetimescale range are central to ETS1 autoinhibi-tion (Figure 5a). Helix HI-1 is only marginally

stable and thus poised to unfold. The recogni-tion helix H3 also shows significant local fluc-tuations (48) supporting the hypothesis thatDNA-binding domains, in general, requireflexibility for high-affinity DNA recognition(77, 78). Furthermore, a network of hydropho-bic residues spanning from helix HI-1 to H3 un-dergoes millisecond to microsecond timescalemotions (79). Most importantly, the ETS do-main appended with all four inhibitory helicesbinds DNA with half the affinity of the isolatedETS domain, whereas the presence of an adja-cent SRR reduces binding 10- to 20-fold. Moststrikingly, the SRR is intrinsically disorderedand interacts only transiently with the ETS do-main and inhibitory module (80).

Collectively, these data lead to the currentmodel of ETS1 autoinhibition in which theregulatable unit exists in a conformationalequilibrium between a flexible state competentfor DNA binding and a more rigid inactivestate (Figure 5a). Transient interactions withthe SRR allosterically favor the latter state.Increasing levels of multisite phosphorylationof the SRR by CaM kinase II leads to aprogressive reduction in DNA affinity bypromoting these transient interactions (79).Consistent with in vivo studies that show a rolefor phosphorylation (81), this rheostatic mech-anism provides a graded control of ETS1 DNAaffinity (82). Although the mechanism linkingHI-1 unfolding with DNA binding is notreadily apparent in static molecular structures(45), molecular dynamics simulations point tocorrelated motions involving HI-1, H4, andH1 that change upon hydrogen bonding of H1to the phosphodiester backbone of DNA (83).This theoretical evidence for a conduit fromthe DNA interface through the regulatableunit is consistent with the dynamics of thisregion observed in the absence of DNA byNMR.

Autoinhibition in Other ETS Proteins

Other ETS factors also exhibit DNA-bindingautoinhibition. A C-terminal inhibitory se-quence, bearing helix H5, strongly attenuates

446 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 11: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

P

P

PP

PP

PPPP

P

HI-1

HI-2

HI-1

HI-2

H5

HI-2

ETS

SRR

SRR and unfolded HI-1

H1–H3

Rigid, inactive Flexible, active

– P

+ P

a

-----

H1

H0

H2–H5

-----+

++

+

+

TAZ1

H1

H0

++

-P-

----

H0

+ P

– P

PNT

Closed Open

H2–H5

b

5H1

PPP

PP

PP

PP

PP

SRR

ETSdomain

PNTdomain

helicalbundle

Appendedinhibitory module

Appendedhelices

Figure 5Dynamic properties of the ETS and PNT domains regulate ETS1 function. (a) ETS1 autoinhibitioninvolves a conformational equilibrium between a rigid inactive state and a flexible active state. Upon DNAbinding, helix HI-1 unfolds. Transient phosphorylation-dependent interactions of the unstructuredserine-rich region (SRR) with the regulatable unit, formed by the core ETS domain (red ) and the appendedinhibitory module (cyan), stabilize the inactive state (80). This stabilization increases progressively withincreasing levels of CaM kinase II multisite phosphorylation of the SRR, leading to rheostatic control ofETS1 DNA binding (79). (b) The PNT domain of ETS1 interacts with the TAZ1 domain of CBP.Phosphorylation of ETS1 causes a conformational change and increases the affinity of the interaction (97).The ETS1 PNT domain consists of a core helical bundle ( green) with appended helices H0 and H1 (orange).MAP kinase phosphorylation of Thr38 and Ser41 shifts a conformational equilibrium of the dynamic helixH0 toward a more open state. This conformational switch, along with increased complementary electrostaticinteractions, favors binding of the TAZ1 domain ( yellow). Note that the structure of the PNT-TAZ1complex has not been determined, and though not shown in panel a or b, both the unmodified andphosphorylated forms of ETS1 can bind CBP and DNA, respectively.

ETV6 DNA binding by steric blockage ofits ETS domain DNA-binding interface(Figure 3a) (50). Members of the PEA3subfamily, including ETV4 (12, 84) andETV5 (85), contain autoinhibitory sequencesappended to both sides of their ETS domains.However, based on proline-scanning muta-genesis, these sequences do not appear to behelical. A cooperative partnership with USF-1enhances the affinity of ETV4 for DNA, at

least in part by interacting with these inhibitorysequences (84).

In cases of the ESE and TCF subfamilies,DNA binding is repressed by sequences dis-tant from the ETS domain. The C-terminalETS domain of ELF3 is inhibited by its cen-tral transactivation domain (86), whereas re-lated ELF5 is inhibited by sequences at its Nterminus (87). The TCF subfamily members,ELK1, ELK3, and ELK4, exhibit even more

www.annualreviews.org • ETS Family of Transcription Factors 447

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 12: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

MAP kinase:mitogen-activatedprotein kinase

complex inhibitory behaviors involving an in-terplay of the B-box, the C-terminal transac-tivation domain, and a NID (NET inhibitorydomain), as well as interactions with the helix-loop-helix Id proteins (88–90). Furthermore,phosphorylation of the transactivation domainresults in enhanced DNA binding (91).

An interesting question is whether theautoinhibition mechanisms in these otherETS proteins have features in common withETS1. A critical feature of autoinhibition inETS1 is the dynamic character of the ETSdomain. The dynamic character of DNA-binding domains, in general, is proposed to benecessary for nonspecific scanning of DNA insearch of specific, high-affinity sites (77, 78),with motions quenched upon specific complexformation. Thus, all ETS domains could havethese dynamic properties. Different appendedinhibitory elements of diverse ETS factorscould impinge on these dynamic propertiessimilarly to how the inhibitory helices of ETS1regulate its motions. Importantly, autoinhi-bition sets up the potential for regulation ofDNA binding through protein partnerships, asdescribed below.

PNT DOMAIN: DIVERSEFUNCTION AND REGULATION

PNT-Domain Structure andAppended Elements

Approximately one-third of ETS familymembers encode a second structured do-main, termed the PNT domain (Figure 1a).This ∼80-residue conserved domain playsunique roles in different ETS proteins.High-resolution molecular models of the PNTdomain are derived from both X-ray crystallog-raphy and NMR spectroscopy performed onone Drosophila and multiple vertebrate proteins(Supplemental Table 3) (92–97). The coreof this domain is formed by four α-helices(H2–H5) along with a short 310- or α-helix(H2′) (Figures 1c and 3b), which is highlysimilar to that of the more widespread SAMdomain. SAM domains exhibit remarkablydiverse association states and function in a

variety of protein-protein and -RNA inter-actions (98). Similar to the ETS domain,additional helical elements (H0 and H1)appended to the conserved-core of the PNTdomain provide unique regulatory paths fordistinct family members (Figure 3b).

Ras Signal Transduction andMAP-Kinase Docking

The transcriptional activation by vertebrateETS1 and ETS2 and the Drosophila orthologPnt-P2 of Ras-responsive genes is increased bymitogen-activated protein (MAP) kinase phos-phorylation of threonine and serine residuesimmediately preceding their highly similarPNT domains (68, 99). Detailed in vitro ki-netic studies revealed that the PNT domainsfacilitate signaling by enhancing substrate bind-ing (ETS1 or ETS2 to the MAP kinaseERK2 and Drosophila Pnt-P2 to the ortholo-gous Rolled kinase) rather than altering thekinetic properties of the enzymes (100–102).Thus, the PNT domains are docking mod-ules, interacting with complementary dock-ing sites on the kinases (103, 104). This pre-sumably increases the effective local concen-tration of the adjacent phosphoacceptors atthe catalytic sites of the enzymes, thereby en-hancing both the specificity and rate of theirmodification (105).

Although a detailed structural model ofa PNT-domain/MAP-kinase complex hasnot been solved, insights into the interfaceshave been obtained from mutagenesis studies,chemical footprinting, and competition exper-iments with peptides that bind known dockingsites on kinases. Key to PNT-domain bindingare residues from helices H2, H4, and H5 sur-rounding an exposed phenylalanine side chain(100, 102). Thus, unlike the well-characterizedlinear peptide motifs that bind docking sites onkinases (106, 107), the PNT domains interactvia a three-dimensional surface. Conversely,this region of the ETS1 PNT domain has beenmodeled to associate with the ERK2 substrate-binding groove centered near loop 12 and theαG helix (108). Additional contacts involving

448 Hollenhorst · McIntosh · Graves

Supplemental Material

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 13: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

the unstructured N-terminal tail of ETS1 withthe common D-recruitment site on the kinasealso have been proposed (109, 110). To bothaccommodate this docking mode and positionthe phosphoacceptors in the active site of thekinase, a structural change in the PNT domain,such as the unfolding of helix H0, would berequired. Indeed, as discussed below, this helixis dynamic and forms part of a phospho-switchmechanism for CBP recruitment.

In contrast, members of the TCF subfam-ily (ELK1, ELK3, and ELK4) lack a PNT do-main and dock on the D- and F-recruitmentsites of MAP kinases via linear peptide se-quences (88, 89). These sequences, termedthe D- and FxFP-motifs, respectively, flankthe phosphoacceptor-containing transactiva-tion domains of the TCF factors. In additionto increasing phosphorylation efficiency and di-recting acceptor specificity, these motifs differ-entially target TCF subfamily members to theMAP-kinase isoforms ERK2, JNK, or p38. Theresulting phosphorylation regulates the TCFproteins at the levels of DNA binding, coac-tivator or -repressor recruitment, and cellularlocalization (2, 88, 91, 111).

Role of the PNT Domain inPhosphorylation-EnhancedInteractions of ETS1 and CBP

The mechanism by which Ras/MAP-kinasesignaling enhances ETS1-regulated geneexpression via phosphorylation-enhancedrecruitment of the general transcriptionalcoactivator CBP has been characterized re-cently using cell-based assays and biophysicalmeasurements (65, 97, 112). ERK2 phosphory-lates Thr38, as well as a nonconsensus acceptorSer41, within the unstructured N-terminalregion of ETS1, immediately adjacent to itsPNT domain. NMR spectroscopic studiesrevealed that, in addition to the four α-helicesthat match the SAM domain (H2-H5), thePNT domain of ETS1 is appended with twoadditional helices (H0-H1) (Figure 3b). Im-portantly, helix H0 is only marginally stable and

undergoes significant local structural fluctua-tions (97).

Dual phosphorylation perturbs a “closed-open” conformational equilibrium of the PNTdomain, displacing the dynamic helix H0 fromthe core bundle (Figure 5b) (97). These mod-ifications increase the affinity of ETS1 for theTAZ1 (or CH1) domain of CBP by ∼30-fold(97). NMR-monitored titration experimentsmapped TAZ1 binding to both the phospho-acceptors and helices H0, H2, and H5 of theETS1 PNT domain. Reciprocal experimentsmapped the interaction surface of the TAZ1domain bound by ETS1. This same CBP re-gion is also implicated in several peptide modelsof transactivation domains bound to the TAZ1domain (113). The charge complementarity ofthese surfaces indicates that electrostatic forcesact in concert with the conformational equi-librium to mediate phosphorylation effects. Asdiscussed above, the same interface of the PNTdomain also serves as a docking site for theERK2 kinase. Thus, the PNT domain of ETS1is key to relaying signal transduction, both“accepting” and “transmitting” the effects ofphosphorylation.

As is the case for many transcription factors,other ETS proteins function in associationwith CBP or its closely related coactivator,p300. GABPA contains an OST domain,which displays a ubiquitin-like fold andbinds the TAZ1 and TAZ2 domains of CBP(Figure 1a,d ) (10). ELK1 binds p300, andRas/MAP-kinase-dependent phosphorylationwithin a transactivation domain enhances theinteraction (89, 114–116). In addition, directinteractions between CBP and p300 havebeen reported for SPI1 (117–118) and ETV1(119, 120). Note, however, ELK1, ETV1, andSPI1 have no PNT domain (Figure 1a), andthe structural bases for CBP or p300 bindingremain to be established. The Mediator hasalso been identified as a coactivator for theTCF subfamily (121–124). The interplay ofmultiple coactivators for transcription factors,including the ETS proteins, remains an areaof current investigation.

www.annualreviews.org • ETS Family of Transcription Factors 449

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 14: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

Promoter: a genomicregion in relativelyclose proximity to thetranscription start sitethat regulatestranscription of thenearby gene

Enhancer: a genomicregion that regulatestranscription atvariable distances fromthe transcription startsite, often greater than1 kb

ChIP:chromatinimmunoprecipitation

ChIP-Seq: agenomics techniquethat combineschromatinimmunoprecipitationwith massively parallelsequencing to identifyDNA sequencesbound by proteins inliving cells

PNT-Domain Self-Association

The PNT domain of ETV6 and its Drosophilaortholog Yan form helical homopolymers ina head-to-tail configuration as deduced fromcrystallographic studies (Figure 4f ) (93, 94,102). The polymer results from the associa-tion of two complementary surfaces of neigh-boring PNT domains, termed mid-loop (ML)and end-helix (EH). Mutant proteins bearing asubstituted hydrophobic to charged residue oneither the ML or EH surface are monomeric.However, a mixture of two mutants, onewith an ML-mutated surface and one with anEH-mutated surface, yields dimers by stableassociation of their remaining wild-type inter-faces. This dimer has been used as a model poly-mer and subjected to functional analysis of therole of self-association by ETV6 and Yan (26,50, 96, 102). As discussed below, the dimericform of ETV6 facilitates DNA binding, butother roles for a longer polymer are possible,such as recruitment of cofactors. With the ex-ception of Yan, ETV6, and likely ETV7 (125),all other PNT domains characterized to datehave one or both of these surfaces blocked byamino acid substitutions or appended helicesand, thus, are monomeric (95, 126).

The EGF-receptor signaling pathwayin Drosophila eye development provides awell-studied example of the interplay of PNTdomains from ETS family members that relieson both kinase docking and self-association(reviewed in Reference 127). Yan functionsas a repressor and self-associates via its PNTdomain (26). Pnt-P2 acts as an activator ofcommon developmental genes. These twoETS factors are targets of the DrosophilaMAP kinase Rolled. Upon signaling, Rolledphosphorylates Yan, leading to its export fromthe nucleus and subsequent degradation in thecytoplasm. Mae, which bears a PNT domainbut no ETS domain (128, 129), facilitates Yanphosphorylation by serving as a docking site forRolled. In addition, the affinity of Mae for Yanis 1000-fold higher than Yan for itself; thus, thepresence of Mae favors Yan depolymerization(96). In parallel, phosphorylation of Pnt-P2

adjacent to its PNT domain stimulates acti-vation of its target genes presumably owing toenhanced recruitment of CBP. The Mae PNTdomain binds to the PNT domain of Pnt-P2,blocking its Rolled docking site and, thereby,preventing its phosphorylation-dependentactivation (102). Thus, PNT-domain homo-and hetero-typic interactions provide a robustresponse to signaling.

The PNT domain is present in certain sub-families (ETS, TEL, ERG) plus GABPA andSPDEF, but absent in others (SPI, ERF, TCF,ELF) (Figures 1a and 2). Multiple evolution-ary events likely led to the loss of this domain asthe family duplicated and diversified. The PNTdomains in the ERG and ELF subfamilies, aswell as in SPDEF, have no reported activities.We anticipate that potential novel functions areyet to be discovered.

GENOMIC APPROACHES TOTHE SPECIFICITY QUESTION:REDUNDANT ANDSPECIFIC BINDING

Recent genome-wide analyses of ETS-factoroccupancy provide a new approach to thespecificity question by identifying genomicregions, both promoters and enhancers, boundby individual ETS proteins in living cells.Genomics technology combines chromatinimmunoprecipitation (ChIP) with eithermicroarrays (ChIP-chip) or massively parallelsequencing (ChIP-Seq) for identification ofthe factor-bound DNA regions (130, 131).Nine ETS proteins have been assayed forgenome-wide occupancy by either promotermicroarrays (ETS1, GABPA, ELF1, ELK1,EWS-FLI1, and SPI1) (132–135) or high-throughput sequencing (GABPA, ETS1, ERG,FLI1, EWS-ERG, EWS-FLI1, SPI1, SPDEF,ETV1, and ELF1) (29, 62, 65, 136–140). (SeeSupplemental Table 4 for public databaseaccess.) Although there are caveats with thecurrent state of this technology (see Caveatsof Genome-Wide Occupancy Techniques,sidebar), recent progress with these approacheshas generated interesting occupancy patterns

450 Hollenhorst · McIntosh · Graves

Supplemental Material

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 15: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

Housekeepinggenes: a looselydefined set of genesthat are expressed in allcell types and functionin normal cell growthand maintenance

CpG islands:genomic regions withmany occurrences ofthe DNA sequence5′-CG-3′, which ismodified by DNAmethyltransferase tobear a methyl-cytosine

for ETS proteins. Furthermore, the availabilityof multiple data sets from several familymembers is providing insights into specificityand genomic views of cooperative partnerships.

Redundant Binding of ETS Proteinsin Living Cells

Genome-wide occupancy data for several ETSproteins have been compared and found to havea high degree of similarity (Figure 6). The re-ported overlap for ETS1 and GABPA showsmost GABPA-bound regions are also occupiedby ETS1 in Jurkat cells (65, 136). Many of thesesame regions are occupied by ELK1 in HeLacells (134, 141). In Figure 6b, we comparedata for ELF1 (62) and GABPA from Jurkat Tcells (136) and SPI1 from mouse macrophages(138). From even these limited data sets, whichuse different techniques, cell lines, and species,overlapping occupancy is observable with sta-tistically significant frequency. We speculatethat the redundancy may encompass the en-tire family because the diverse ETS proteins,which have already been implicated, includingGABPA, ETS1, ELK1, ELF1, and SPI1, rep-resent five subfamilies.

Overlapping binding within the resolutionof the ChIP technology could be the result ofclusters of binding sites specific for each familymember. Alternatively, interchangeable occu-pancy of multiple ETS factors at one site overtime could be sampled in the pool of multiplecells (Figure 7). Favoring the latter hypoth-esis, high-resolution comparisons of bindingdata for ETS1 and GABPA reveal sharp colo-calization of binding events (65). Furthermore,bioinformatics analyses of regions with broadredundancy identified a striking enrichment ofthe sequence CCGGAAGT (132, 134). Thissequence motif matches exactly the in vitro-derived DNA-binding consensus of all but fourETS proteins (Classes I and II) (Figure 6a).Thus, the sequence motifs in redundantly oc-cupied regions are likely able to bind most, ifnot all, ETS proteins.

DNA regions occupied redundantly bymany ETS proteins are most frequently found

CAVEATS OF GENOME-WIDE OCCUPANCYTECHNIQUES

Microarrays and massively parallel sequencing can identify the se-quences of DNA derived from ChIP experiments. Mapping thesesequences to the genome results in peaks of reads above back-ground that indicate the genomic region bound by the immuno-precipitated transcription factor. Lists of bound regions identi-fied by peak-calling algorithms are not comprehensive, but theygenerally reflect cut-off values that compromise between false-negative and false-positive results. Resolution ranges from tensto hundreds of base pairs depending on the quality of the peak(136, 190). Bioinformatics approaches can predict binding se-quences by searching for overrepresented patterns in large num-bers of bound regions or by directly searching for a particularmotif. Neither method proves the use of a particular motif. It isalso important to consider that bound regions may differ by celltype or growth condition. Another caveat of the current technol-ogy is the limited information regarding the function of a boundtranscription factor, including information about which gene(s)may be targets of regulation. Currently, bound regions are usu-ally assigned to the closest genes to make functional correlations.Compilation of correlative genomics and bioinformatics data isessential at this stage with anticipation that technology can drivehigh-throughput tests of functionality in the future.

a short distance (∼20 to 40 bp) upstream oftranscription start sites and are correlatedwith characteristics of active promoters, asevidenced by DNase I sensitivity and presenceof histone H3K4 tri-methylation (65, 142,143). There is also an indication from geneontology studies that “housekeeping-type”genes are more likely to be characterized by thisredundant ETS-factor binding. Searching foroverrepresented motifs in the human genomeidentified an ETS-binding sequence located∼25 bp upstream of the transcription start siteof CpG island-containing housekeeping genepromoters (144–146). It has been proposedthat housekeeping genes, which are activelyexpressed in most cell types, could be regulatedby different ETS factors in different cell types.The frequency of this sequence (in ∼25% ofhuman promoters) (65), the association withcommonly expressed housekeeping genes, and

www.annualreviews.org • ETS Family of Transcription Factors 451

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 16: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

GABPA

SPI1

ELF1

1527

201

2114

1108

653

1286

3430

1 2 3 4 5 6 7 8 9 10

In vivo

GABPA

ETS1

RUNX

ELK1

GABPASRF

4376992

376

445148

84

42

a b

c d

In vitro

1278

382144

5466299

7835

Figure 6Genomic analyses via ChIP-chip and ChIP-Seq detect specific and redundant ETS binding. (a) Redundantlyoccupied regions in cells have a DNA sequence similar to preferred ETS-binding sequences derived in vitro.Sequence frequencies are compiled (top) from all 27 in vitro–derived ETS-binding sites (62) or (bottom) fromthe most overrepresented sequence in regions co-occupied in cells by GABPA, ETS1, and ELF1 (62, 65,136). Frequencies are shown in logo form, which depicts base pair (A:T, T:A, G:C, C:G) frequency asrelative heights of A, T, G, and C, respectively. (b) Redundant occupancy is observed for even the mostdivergent family members and between species. The relative distribution of specific and overlapping boundregions are shown for GABPA (136) and ELF1 (62) in human Jurkat T cells and SPI1 in mouse macrophages(138) by numbers of bound regions within a Venn diagram display. Analysis of SPI1 mouse data was limitedto promoter regions, which could be compared with orthologous human regions using the LiftOver tool athttp://genome.UCSC.edu (235). Data were reprocessed for this comparison, as previously reported (65).(c,d ) Serum response factor (SRF) and RUNX preferentially co-occupy regions bound specifically by ELK1and ETS1, respectively. Diagrams (c,d ) illustrate data from Boros et al. (134) and Hollenhorst et al. (65),respectively. See Supplemental Table 4 for database sources of all genomics data.

its occurrence near transcription start sitessuggest a common role for ETS factors in pro-moter proximal regions, such as transcriptioninitiation, promoter clearance, or maintenanceof active chromatin. Functional studies will benecessary to resolve whether individual ETS

proteins have distinct activities at these sitesor whether a redundant function could beexecuted by alternative family members.

Redundancy within subfamilies has not beenextensively investigated by genome-wide tech-niques. However, such redundancy may be

452 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 17: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

expected and has been investigated in a detailedanalysis of the EGR1 promoter. This promoteris regulated by the TCF subfamily, in part byrecruitment of Mediator, as mentioned above.Direct ChIP experiments for ELK1, ELK3, andELK4 demonstrated alternate occupancy thatappeared to correspond to relative abundanceof the factors in different cell types (121). Inthis case, findings suggest the TCF proteinsdiffered in Mediator binding, thus providingdistinct EGR1 expression and responsiveness tosignaling in different cell types.

Specific Binding of ETS Proteins inLiving Cells

Specific binding by an ETS factor is defined asthe binding of only one or possibly only highlyrelated subfamily members. Because compar-isons of ETS transcription factor genomicoccupancy data reveal a significant amount ofredundant binding, it can be difficult to differ-entiate specific binding sites from false-negativeor false-positive results. Furthermore, untilall ETS family members expressed in a singlecell are assayed by ChIP, specificity cannot bedefinitively concluded. Despite these limita-tions, current genome-wide studies have identi-fied specific genomic occupancy that correlateswith predicted ETS protein roles. For example,in hematopoietic stem cells, the closely relatedETS proteins ERG and FLI1 bind to a largelyredundant set of targets, whereas targets of themore divergent ETS protein SPI1 have muchless overlap (29). However, the mechanism ofspecificity is yet to be determined.

In some cases the subtle differences inDNA-sequence preference observed in vitroare supported by genomic studies. Regionsbound specifically by ETS1 are found in Jurkatcells and have a higher proportion of sites witha thymine at position 7 compared with regionsco-occupied by ETS1 and GABPA (65).This observation may be due to the relativetolerance of this change by ETS1 comparedwith other ETS proteins (147). Genomicoccupancy data for SPI1 in myeloid cells andSPDEF in prostate cells enabled bioinformatics

AR AR

FLI1

EWS

FLI1

EWS

ETS1

ELK1

GABPA

ELF1

Housekeeping genes

T-cellactivation

genesRUNX

Serumresponsive

genesSRF SRF

Prostatedifferentiation

genesERG

Ewing’s sarcoma

genes

CCGGAAGT

AGGATGTGG

GGAAGGAAGGAAGGAA

CC(A/T)6GG

ELK1

CCGGAAGT

GNACANNNTGTNCC AGGAAG

ETS1

Figure 7Partnerships provide a route to specificity in normal and disease states. Modelsdepict ETS protein interactions with other DNA-binding proteins at thepromoters or enhancers of distinct target classes. ETS proteins exhibitredundant occupancy of a consensus ETS-binding site in housekeepingpromoters (132, 134). In contrast, ETS proteins bind specifically near genesthat agree with predicted biological functions in either normal or cancer cells(65, 133–134, 139). Specific interactions are associated with activation ( greenarrows) or repression (red bar) of target genes. Specific targets are highlightedby co-occupancy of multiple transcription factors [RUNX, serum responsefactor (SRF), androgen receptor (AR)] implicating partnerships as drivers ofspecificity. EWS-FLI1 tandem occupancy is inferred from functional andbiochemical analyses of binding to GGAA microsatellite repeats (133, 137,187). See Supplemental Table 4 for database sources of all genomics data.

analyses for these divergent family members(62). Position-weight matrices derived fromunbiased searches of top-scoring regions showsimilarities to the subtly different sequencesderived from the high-throughput proteomicsapproaches that are discussed above. However,analysis of ELK1, SPI1, GABPA, ERG, FLI1,and ETV1 by other groups report no distinctETS motifs (29, 134, 136, 138–140). Moreparsing of bound regions into different classes,such as redundant proximal promoter versusspecific distal enhancers, may be necessary toascertain signatures of specificity determinants.

One sequence change that is often seenin specific binding sites is the presence of anadenine instead of a cytosine at position 3

www.annualreviews.org • ETS Family of Transcription Factors 453

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 18: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

(ACAGGAAGT). This change may influencespecificity in some cases. However, it mayalso be a result of selection against a CpG,which would be vulnerable to DNA methy-lation and, thus, inhibition of ETS-proteinbinding (148, 149). This selection would benecessary at enhancers, where ERG (62) andETS1 specificity (65) is observed, but not atredundant promoter-binding sites where theETS-binding sequence can be protected fromDNA methylation as a result of proximity to,or inclusion in, hypomethylated CpG islands.

In conclusion, genomic-binding studieshave revealed both redundant and specific occu-pancy by ETS factors. The power of genomicsto detect redundancy contrasts with the limits ofgenetic analyses in which such redundancy mayeasily be masked. However, the relevance of allgenomic occupancy awaits more detailed func-tional tests, especially ones that can distinguishroles of distinct ETS factors. In addition, manyETS sites predicted on the basis of biochemicalunderstanding, and bioinformatics tools are notoccupied in vivo, and conversely, many actualsites of genomic occupancy are not predicted.These discrepancies are consistent with stud-ies in other transcription factor families (130,150) and likely reflect in vivo complexities suchas competition with chromatin packaging andother DNA-binding proteins. Thus, identifica-tion of other chromatin-associated factors andspecificity partnerships, as described below, willprovide a necessary backdrop to understandETS-protein binding detected in living cells bygenomics approaches.

COOPERATIVE PARTNERSHIPSPROVIDE ETS FAMILYSPECIFICITY: GENOMIC ANDBIOCHEMICAL PERSPECTIVES

A well-established theme in the regulationof eukaryotic gene expression is combinato-rial assembly of multiple factors within thesame control elements (e.g., a promoter orenhancer). This phenomenon can provideadded specificity and thus synergistic controlif there is positive or negative cooperative

DNA binding between juxtaposed factors orjoint recruitment of additional components ofthe transcriptional machinery. ETS proteinsdisplay a variety of such cooperative part-nerships, as previously reviewed (1, 2, 151,152). To provide a mechanistic frameworkfor understanding this route to specificity, wefocus on the high-resolution structural modelsof ETS partnerships that delineate underlyingprotein-protein interactions (Figure 4; Sup-plemental Table 2) and on cases supported byunbiased identification of composite elementsthrough genome-wide analysis (Figure 7).

Ternary Complex Factors/SerumResponse Factor

Serum response elements that mediate acti-vation of immediate-early genes such as c-FOS include both ETS- and SRF-binding sites.The cooperative binding interactions betweenSRF and the TCF subfamily of ETS tran-scription factors, including ELK1, ELK3, andELK4, have been well characterized (11; re-viewed in Reference 89). Crystallographic stud-ies demonstrate that DNA binding is enhancedby a direct protein contact between ELK4 andthe SRF MADS domain (44). The partnershipalso depends on the TCF B-box, a 21-residueregion that adopts a helix-sheet-helix motifwhen bound to the MADS domain of SRF. Thisconserved box is limited to the TCF subfamilyand, thus, illustrates a route to both redundancywithin a subfamily and specificity within the en-tire ETS family (Figures 1a and 4b).

Chromatin occupancy studies that relied onChIP-chip reveal co-occupancy of ELK1 andSRF at more than 200 promoters (Figure 6c)(134, 153). These co-occupied regions are morelikely than other sites to be bound specificallyby ELK1 and not by another ETS protein.This specific binding mode occurs at only 22%of ELK1-bound regions, whereas the redun-dant binding mode, characterized by a strongconsensus ETS-binding sequence and overlap-ping occupancy with other ETS transcriptionfactors, is observed more frequently (50%).Thus, similar to ETS1, the TCF subfamily can

454 Hollenhorst · McIntosh · Graves

Supplemental Material

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 19: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

undergo both specific and redundant binding(Figure 7).

SPI1/IRF4

The ETS factor SPI1 partners with theinterferon regulatory family transcriptionfactor IRF4 at immunoglobulin light-chaingene enhancers. The low affinity of the wingedhelix-turn-helix DNA-binding domain of IRF4for the λB enhancer is increased 20–40-fold bythe presence of SPI1 at this site (154, 155). Thecrystallographically determined structure ofthe two DNA-binding domains on a compositeenhancer sequence reveals that this coopera-tivity arises from both direct protein-proteincontacts across the central minor grooveand induced DNA conformational changes(Figure 4e) (46). More dramatically, however,the phosphorylated PEST sequence of SPI1interacts with an autoinhibitory region of IRF4to enhance DNA binding by almost threeorders of magnitude (154). This enhancedinteraction awaits structural characterization.

ETS1/RUNX1 and Autoinhibition

ETS1 and RUNX1 form a partnership that iswell characterized by biochemical and genomicapproaches (156–158). The level of cooperativ-ity of ETS1 and RUNX1 on ETS sites sim-ilar to that in the Moloney murine leukemiavirus enhancer approximates the 10-fold re-pression of ETS1 DNA binding by autoinhibi-tion. Also, ETS1 variants with a deleted or dis-rupted inhibitory module no longer show suchenhancement. These observations suggest thatcooperativity results from a relief of autoinhi-bition. Intriguingly, the effect also requires au-toinhibitory sequences in RUNX1 (157). At thetime of writing this review, molecular coordi-nates for several ternary complexes of ETS1 andRUNX1 are on hold in the Protein Data Bank.Thus, we can anticipate informative structuraldata on this partnership in the near future.

Both redundant and specific occupancywere characterized for ETS1 in Jurkat Tcells with more than 14,000 bound regions

detected (65). Specific ETS1 occupancy oc-curs primarily in regions relatively distal totranscription start sites that have enhancercharacteristics, such as DNase I sensitivityand histone H3K4 mono-methylation (142,143). More than 1000 of these specificallybound regions detected by ChIP-Seq displaythe sequence AGGA(A/T)GTGG. This motifhas been characterized in the T-cell receptorenhancers as an ETS1/RUNX1 cooperativebinding element with the RUNX1 site com-prising the (A/T)GTGG (Figure 7). Indeed,co-occupancy by RUNX factors at many ofthe ETS1-specific regions was verified byChIP-Seq (Figure 6d ). More than 30-foldcooperative binding is detected with compositesequences bearing this close spacing (156);thus, there may be additional mechanisms atplay beyond simply counteracting the 10-foldeffect of autoinhibition. Ontology analysis ofgenes with the composite motif included com-ponents of the T-cell activation machinery,including ZAP70, LCK, CXCR3, CXCR4, andCD38. Thus, the ETS/RUNX sequence motifis predictive of the function of nearby genes(65). These sites also recruit the transcrip-tional coactivator CBP, which adds furtherevidence for their functional relevance. Thesegenomic studies are consistent with geneticsexperiments in which mice with a deletionof ETS1 have defects in T-cell activation(159).

Despite the evidence that ETS1 regulatesT-cell genes via ETS/RUNX-binding sites, it isnot clear that these sites are completely specificfor ETS1. Currently, only GABPA and ELF1ChIP-Seq data are available in Jurkat T cells tocompare with ETS1 (62, 136). However, otherETS proteins such as ELF2, ELF4, and FLI1have been reported to interact with RUNXproteins (160, 161). Furthermore, ChIP-Seqin hematopoietic stem cells identifies extensiveco-occupancy of RUNX1 with FLI1 and ERG(29). Thus, it is possible that ETS/RUNX bind-ing sites have only partial specificity for sub-sets of ETS proteins and that combinations ofETS and RUNX factors may vary with cell type.The forthcoming structural analysis of ETS and

www.annualreviews.org • ETS Family of Transcription Factors 455

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 20: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

RUNX combinations will be important for de-ciphering rules of their partnering.

ETS1/PAX5

ETS1 and PAX5 bind cooperatively to a com-posite sequence located in the promoter ofmb-1, a gene expressed in early B cell differenti-ation (Figure 4c) (162). This ETS-binding sitedisplays a variant GGAG core. Crystallo-graphic studies of this cooperative partnershipdemonstrate a protein contact that leads to analtered conformation of a tyrosine conserved inmost ETS domains (Tyr395 in ETS1). Gln22,found in the β-hairpin of the PAX5 paired do-main, hydrogen bonds to Tyr395 in the recog-nition helix H3 of ETS1 to stabilize this alteredconformation. This enables new DNA contactsfor ETS1, while avoiding unfavorable interac-tions with the variant GC bp (43). Interest-ingly, this same conserved tyrosine is involvedin ELK4’s ability to bind the variant GGATcore (41).

This partnership also provides insights intothe linkage of protein interactions and ETS1autoinhibition. The PAX5 paired domain en-hances the binding of both the autoinhibitedand uninhibited variants of ETS1 to the sameaffinity (163). Thus, the PAX5 partnership ap-pears to counteract the effects of autoinhibi-tion, possibly through the ETS domain ratherthan direct interactions with inhibitory ele-ments. Gln22 of the paired domain also con-tacts Gln336 near the N terminus of helix H1 ofthe ETS domain (43), providing a conduit intothe dynamic core of the ETS domain. There isspecificity of this PAX5 partnership for ETS1rather than other family members, thus illus-trating the use of protein interactions to selectindividual ETS proteins (162). However, thebiochemical basis for this selectivity is not clear,as the key residues Tyr395 and Gln336, whichcontact PAX5, are present in other family mem-bers. Furthermore, there are no genomic datain the B-cell system to ascertain the frequencyand specificity of this partnership in cells or thecommon occurrence of a PAX5-ETS compos-ite element.

ETS/FOX

The major historical route to discovery of part-nerships in the ETS family, including thosementioned above, has been mapping of tissue-specific enhancer regions that leads to the iden-tification of a functional ETS-binding site andneighboring synergistic elements. A number ofadditional ETS partnerships have been impli-cated at this level, but they are not the focus ofthis review. However, one is mentioned to il-lustrate emerging topics and critical directionsfor investigation of partnerships. The ETS andFOX families converge, as illustrated by the roleof ETV2 and FOXC2 in the endothelial-cellgene-expression program (31, 32). The part-nership is characterized by a composite motif,detected in genome-wide bioinformatics anal-yses, which can be co-occupied by ETV2 andFOXC2 in DNA-binding assays. Endothelial-cell-specific genes, including FLK1, TAL1,NOTCH4, and VE-CADHERIN, display themotif. Importantly, a genome-wide search withthis motif identified endothelial-specific genes.A combination of cell-based transcription as-says, mouse transgenics, and zebrafish reversegenetics demonstrates the function of this part-nership. The basis of transcriptional synergy isnot yet established, and thus it will be importantto perform biochemical and structural studieson an ETS-FOX ternary complex.

ETV6 Self-Associationand Autoinhibition

In contrast to most ETS proteins, ETV6 isa transcriptional repressor. ETV6 monomersbind DNA extremely weakly owing to au-toinhibition of the ETS domain, as discussedabove (50). In contrast, an engineered ETV6dimer shows higher affinity, but only onmultiple ETS-binding motifs. It is proposedthat self-association via the PNT domain (seeFigure 4f ) mediates cooperative DNA bind-ing, which compensates for the low affinity ofthe monomeric ETS domain. Furthermore,this cooperativity is very tolerant of the orien-tation and spacing (within five helical turns) of

456 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 21: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

the multiple ETS-binding sites, suggesting asubstantial degree of flexibility of the middle re-gion of ETV6 (∼200 residues), which links theETS and PNT domains (Figure 1a). Collec-tively, these results suggest that autoinhibitionand polymerization could define the specificityof ETV6 by targeting promoters bearingmultiple binding sites. Such sites need not havea well-ordered array of GGA core sequences.Furthermore, polymerization of ETV6 isspeculated to create an extended complex,which, along with recruitment of corepressors,may facilitate transcriptional repression (93).

Reports on Drosophila Yan, the apparent or-tholog of ETV6, strongly suggest that self-association is a key to the function of this classof ETS factors (26). Biological relevance isalso drawn from the chromosome rearrange-ments of the ETV6 locus that encode onco-genic fusion proteins associated with B cellleukemias. The fusions include the PNT do-main of ETV6 and the DNA-binding domainof either RUNX1 (Figure 1a) (164) or PAX5(165, 166). We speculate that the rules re-garding DNA binding learned from the nativeETV6 polymer, as modeled by the dimer, mayalso apply to these oncogenic fusion proteins.Genomic and additional biochemical data willbe helpful to understand fully how the biophys-ical properties of ETV6 impact target selectionof both native and oncogenic versions of ETV6.

Other Dimeric Configurations

In addition to ETV6, several ETS proteinshave been implicated in dimeric interactions.ETS1 binds cooperatively as a homodimer totandem ETS motifs, such as that found inthe MMP3 promoter. The cooperativity ofdimerization is the same 10-fold effect as au-toinhibition, suggesting that the dimer config-uration may counteract autoinhibition (167).The crystallography-based model of the ternarycomplex reveals that residues in the loop be-tween inhibitory helix HI-2 and helix H1 ofone monomer interact with those in the turnbetween helices H2 and H3 of the ETS do-main in the other monomer (Figure 4d ) (49).

An alternatively spliced isoform of ETS1, lack-ing inhibitory elements N terminal to theETS domain, fails to dimerize, thus implicat-ing the contact made by the inhibitory elementsin the cooperativity mechanism (168). How-ever, as with ETS1/PAX5 and ETS1/RUNX1partnerships, a complete mechanism for linkingpartnerships and autoinhibition is still lacking.

Other ETS proteins possibly use multi-ple ETS-binding motifs. On the basis of thecrystallographic structure of a single ELF3ETS domain bound to the B site of a tan-dem type II TGF-B receptor promoter, en-hanced binding of a second ELF3 to the ad-jacent A site was proposed to result fromprotein-induced DNA conformational changes(51). This configuration has not been investi-gated by either biochemical or genomics ap-proaches. In addition, GABPA has two alter-native non-DNA-binding partners GABPB1and GABPB2, which enhance its DNA-bindingaffinity and provide a homodimeric interfaceto form GABPA-(GABPB)2-GABPA heterote-tramers on DNA (40). The spacing require-ment for the tetrameric GABPA/B complex hasnot been determined, and genomic data forGABPA have not yet elucidated whether suchcomplexes distinguish specific sites in promot-ers. Note that most of the GABPA sites are alsoused by monomeric ETS1 or other ETS pro-teins (65), thus implicating no GABPA speci-ficity determinants.

The ability of ETS factors to use multi-ple ETS motifs either by ETV6 polymers orother dimeric configurations begs the questionof whether clusters of ETS consensus sites oc-cur in the genome. We found no evidence foruse of tandem ETS sites in Jurkat cells withthe dimeric spacing of the MMP3 promoter(P.C. Hollenhorst, unpublished data). How-ever, a high density of many transcription factorbinding motifs, including ETS sites, is notedin promoter and enhancer regions (134, 136,169). Testing of ETS-factor polymeric bindingin cells awaits discovery of additional authenticETS targets.

In conclusion, combinatorial control oftranscription employs multiple DNA-binding

www.annualreviews.org • ETS Family of Transcription Factors 457

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 22: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

AR: androgenreceptor

transcription factors in close proximity. Al-though the DNA recognition sequences forETS proteins and their partners may be closelyjuxtaposed or even overlapping, the resultingprotein-protein interfaces are often small andthe effects on their DNA binding rather subtle.Nevertheless, the thermodynamic result of thedirect protein-protein interactions and/or theindirect changes in protein-DNA interactionsis net-positive cooperativity relative to theindependent binding of each transcription fac-tor. The added specificity of larger compositeDNA-binding sites, which is gained throughthe cooperative binding of two factors, mayconstitute the greatest biological impact ofsuch partnerships. Extrapolating from the de-tailed mechanism of ETS1 autoinhibition, thedynamic character of the ETS domain couldbe impacted within the context of a cooperativepartnership. It remains to be determined howmany of these ETS factors and their respectivepartnerships tap into the potential to regulatean ETS domain by autoinhibition and/or byaffecting protein dynamics.

GENOMIC INSIGHTS INTOONCOGENIC ETS FACTORS

ERG and Androgen Receptor inProstate Cancer

Rearrangements of ETS loci are a hallmark ofprostate cancer with ∼50% of tumors show-ing alterations at an ETS gene locus (36). Inthe most common scenario, a promoter regionfrom an androgen-responsive, prostate-specificgene, TMPRSS2, is attached to the ERG locusto drive aberrantly regulated ERG expressionin prostate cells. Chromosomal rearrangementsthat result in the overexpression of members ofthe PEA3 subfamily (ETV1, ETV4, or ETV5)are found less frequently (36, 170, 171). Thesegenetic changes are implicated in tumorigene-sis (170, 172–176). ChIP-Seq of ERG in bothcell lines and tumors that overexpress this tran-scription factor identifies a striking level of co-occupancy (44%) with the androgen receptor(AR) (62, 139). The involvement of the AR

in normal prostate development, coupled withthe prevalence of ETS-binding sequences atAR-bound regions (177), implies a coopera-tive interaction that could play a role in ETS-mediated prostate oncogenesis (Figure 7).Expression studies indicate that a potentialmechanism of oncogenic ETS proteins, suchas ERG and ETV5, at these sites is antagonismof normal AR function (139, 178, 179). Sup-porting this idea, tumor-suppressor ETS pro-teins, such as SPDEF, synergize with the AR(66). However, the genetic finding that ETSand AR genes cooperate in oncogenesis (175)indicates that this mechanism is more complexthan our current understanding and may differon a target-by-target basis (180).

The biochemical mechanism for interac-tions between ETS factors and the AR is alsonot known. There is some evidence that theAR can physically interact with ERG (139),ETV5 (179), SPDEF (66), ETV1 (181), andETS1 (177) through their ETS domains, evenin the absence of DNA. However, whetherthis interaction has any ETS family specificityor how it impacts ETS function remainsunknown. DNA-binding assays have nottested cooperative binding, and bioinformaticsanalyses have not identified an overrepre-sented spacing of ETS- and AR-binding sitesfunctionally analogous to that observed withthe ETS1/RUNX partnership. Therefore, theinteraction between ETS proteins and the ARmay lead to specificity of function, but notnecessarily cooperative DNA binding. Thus,ETS proteins may influence cancer via ARtarget genes, but the mechanism of this effectremains to be discovered.

EWS-FLI1 in Ewing’s Sarcoma

Ewing’s sarcoma, a common pediatric cancer,is invariably associated with ETS gene translo-cations (182) that result in a fusion oncogeneconsisting of the 5′ end of EWS and the 3′ endof FLI1, or less frequently ERG, ETV1, ETV4,or FEV (Figure 1a) (183, 184). The strongtransactivation activity in the N terminusof EWS contributes to the function of the

458 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 23: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

Microsatellite: agenomic region with10 to 100 tandemrepeats of a 2–6-bpsequence

resulting oncogenic fusion protein (185). How-ever, insights from genomic studies suggestthat the fusion protein also directs regulationof new transcriptional target genes. ChIP-chipand ChIP-Seq have identified the genomicregions bound by the most common ETS fu-sion proteins in Ewing’s sarcoma, EWS-FLI1and EWS-ERG (62, 133, 137). Two overrep-resented DNA sequences are found in theseregions: a consensus ETS-binding site that issimilar to promoters discussed above, whichhave redundant occupancy of ETS factors, andmicrosatellites that bear tandem repeats of thesequence GGAA. Of these two classes of targetsequences, only genes near the microsatelliterepeats correlate with expression changes asso-ciated with transformation, including NROB1(133) and GSTM4 (186). Thus, GGAA repeattargets, but not redundant promoter targets,mediate the oncogenic role of these fusions.

GGAA microsatellites, which can havemore than 10 copies of the tetrameric repeat,constitute arrays of ETS-binding sites thatcould mediate cooperative binding betweenneighboring factors. In fact, four or morerepeats are necessary for EWS-FLI1 transcrip-tional activation in reporter assays, and twoEWS-FLI1 monomers bind to this four-motifarray in biochemical assays (Figure 7) (133,137, 187). However, whether cooperativity isat play is unclear. A recent study revealed thatthe EWS portion of the fusion oncoproteincontributes only to the transcriptional acti-vation, not the DNA-binding activity. Thisimplicates the ETS-domain component of thefusion protein in microsatellite binding (187).Indeed, multiple ETS proteins, includingexamples not involved in Ewing’s sarcoma, canbind to GGAA microsatellites. Furthermore, asdiscussed above, ETS proteins ETV6, ETV7,

GABPA, and ELF3 bind as homo-oligomersto neighboring ETS-binding sites. Whetherthe GGAA microsatellites that are involved inEwing’s sarcoma are the natural targets of anyof these ETS proteins remains to be tested.

In conclusion, the oncogenic potential ofETS factors is likely derived from theirtranscriptional properties. In addition to thechromosome rearrangements described above,there is mounting evidence for an oncogenicrole for dysregulation of ETS genes in prostate(188, 188a), melanoma (189), and gastroin-testinal stromal tumors (140). Alterations inany ETS protein can have multiple down-stream effects, amplifying the primary geneticdefect. Thus, a potentially effective interven-tion in ETS-driven cancers would be a dis-ruption in the function of the oncogenic ETSprotein. The range of macromolecular inter-actions, which include DNA-protein and var-ious homo- or hetero-typic protein-proteincomplexes, all serve as potential interfaces fortargeted therapeutics.

In considering the route to oncogenesis asa window into the specificity question of theETS family, it is noteworthy that multiple fam-ily members can be involved in sarcoma andprostate cancer with an interesting overlap be-tween the two diseases. There are no substantialdata to indicate that the various family memberscause different disease progression or outcome.Future questions include whether these familymembers are selected as a result of susceptibilityof the genetic locus to rearrangements or dis-tinct properties of these, but not other, ETSfamily members. Finally, continued genomicand biochemical analysis of the oncogenic ETSfactors can help elucidate oncogenesis pathwaysas well as shed light on specificity routes withinthe family.

SUMMARY POINTS

1. Conservation of the ETS domain leads to similar DNA interactions among all ETSfactors as detected by in vitro biochemical assays. However, deviation from these highest-affinity sites is encountered in biological settings.

www.annualreviews.org • ETS Family of Transcription Factors 459

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 24: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

2. The PNT domain provides diversity to ETS proteins, both by its variable presence anddiffering functions, ranging from kinase and coactivator binding to self-association.

3. The DNA-binding affinity of many, if not most, ETS proteins is tempered by autoin-hibition. Specific DNA motifs used in cells can diverge from the optimal consensus,thus lowering affinity further. This combination of circumstances poises an ETS fac-tor to participate in partnerships with other factors that have additional preferencesfor DNA sequence and architecture, thus enhancing specificity for genomic locations.High-resolution molecular models demonstrate the relatively small protein-interactionsurfaces that can mediate such cooperativity.

4. Protein dynamics underlie phosphorylation-dependent regulatory processes, includingautoinhibition of the ETS domain and enhanced binding of CBP by the PNT domain.Flexible regions, which are likely sites of modification, function in concert with morestructured regions, which also exhibit dynamic conformational equilibria.

5. ETS binding sites displaying a strong match to the highest-affinity consensus motifs arecommon in CpG island-containing housekeeping promoters. These sequences serve asinterchangeable binding sites for many ETS proteins. Every cell type expresses multipleETS factors that could potentially participate in this redundant occupancy.

6. Genome-wide occupancy studies identify many (hundreds to thousands) binding sitesfor a single ETS protein in a particular cell type. Although the functional relevance ofmany of these sites awaits further analysis, bioinformatics approaches have identifiedETS consensus motifs and composite elements that are beginning to define genomicdeterminants for specificity and cooperative partnerships.

7. Oncogenic ETS proteins alter numerous transcriptional targets within cells that expressmany other ETS factors. Selection of oncogenic target genes is likely achieved by altereduse of the specificity mechanisms for normal cells described in this review.

FUTURE ISSUES

1. Dynamics are increasingly implicated as a powerful regulatory mechanism in proteinfunction. Studying how intra- and intermolecular interactions establish the amplitudeand timescale of these dynamics—and, thus potentially, the DNA-binding properties oftranscription factors—is a new avenue for understanding the regulation of gene expres-sion. Biophysical studies of increasing sophistication will help define the physicochemicalbases for ETS autoinhibition and dissect the role of dynamics in protein and target DNArecognition by the PNT and ETS domains, respectively.

2. A high level of redundant genomic occupancy in the ETS family was detected in promoterproximal regions. Further work will determine whether ETS proteins play a redundantmechanistic role in transcription regulation at these targets and whether these sites havefunctions distinct from more distal enhancer elements.

3. As genome-wide binding data become available for more ETS proteins, as well as othertranscription factors, comparisons will allow classification of redundant, specific, andpartially specific binding sites. This categorization will improve the robustness of thenew data sets to investigate mechanisms that enable specificity within the ETS family,

460 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 25: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

including composite elements for cooperative binding partnerships and other routes tosynergistic transcriptional regulation. Redundancy within subfamilies versus the entirefamily needs to be clarified.

4. The ETS genes that are involved in chromosomal abnormalities in solid tumors arelimited largely to those in ERG and PEA3 subfamilies. It is not known to what degree theoncogenic functions of these genes overlap. Determining the specific roles of these twosubfamilies, compared with the rest of the ETS family, will be important to understandingthe mechanisms of ETS-mediated oncogenesis.

5. The development of therapeutic strategies, including small-molecule modulators of pro-tein interactions, to regulate ETS proteins could have high clinical impact. Currentlyavailable and new structural and biochemical information about ETS proteins will bevaluable in achieving this challenging research goal.

DISCLOSURE STATEMENT

The authors are not aware of any affiliations, memberships, funding or financial holding thatmight be perceived as affecting the objectivity of this review.

ACKNOWLEDGMENTS

We thank Tim Parnell and Krista Meyer for critical comments on the manuscript, David Nixand Katherine Chandler for help with data analysis, and Carlos Escalante and Aneel Aggarwalfor providing the coordinates of the SPI1/IRF4/DNA complex. This work was supported by theNational Institutes of Health through awards GM38663 (to B.J.G.) and CA42014 (to the Hunts-man Cancer Institute for support of shared resources) as well as the Canadian Cancer Society Re-search Institute (017308 to L.P.M.). Huntsman Cancer Institute/Huntsman Cancer Foundationfunding (to B.J.G.); Indiana University School of Medicine support (to P.C.H.); and instrumentsupport from the Canadian Institutes for Health Research, the Canadian Foundation for Inno-vation, the British Columbia Knowledge Development Fund, the Michael Smith Foundation forHealth Research, and the UBC Blusson Fund (to L.P.M.) are gratefully acknowledged.

LITERATURE CITED

1. Graves BJ, Petersen JM. 1998. Specificity within the ETS family of transcription factors. Adv. CancerRes. 75:1–55

2. Sharrocks AD. 2001. The ETS-domain transcription factor family. Nat. Rev. Mol. Cell Biol. 2:827–373. Oikawa T, Yamada T. 2003. Molecular biology of the ETS family of transcription factors. Gene 303:11–

344. Hsu T, Trojanowska M, Watson DK. 2004. ETS proteins in biological control and cancer. J. Cell.

Biochem. 91:896–9035. Gutierrez-Hartmann A, Duval DL, Bradford AP. 2007. ETS transcription factors in endocrine systems.

Trends Endocrinol. Metab. 18:150–586. Kumar-Sinha C, Tomlins SA, Chinnaiyan AM. 2008. Recurrent gene fusions in prostate cancer.

Nat. Rev. Cancer 8:497–5117. Degnan BM, Degnan SM, Naganuma T, Morse DE. 1993. The ETS multigene family is conserved

throughout the Metazoa. Nucleic Acids Res. 21:3479–84

www.annualreviews.org • ETS Family of Transcription Factors 461

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 26: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

8. Wang Z, Zhang Q. 2009. Genome-wide identification and evolutionary analysis of the animal specificETS transcription factor family. Evol. Bioinform. Online 5:119–31

9. Laudet V, Hanni C, Stehelin D, Duterque-Coquillaud M. 1999. Molecular phylogeny of the ETS genefamily. Oncogene 18:1351–59

10. Kang HS, Nelson ML, Mackereth CD, Scharpf M, Graves BJ, McIntosh LP. 2008. Identification andstructural characterization of a CBP/p300-binding domain from the ETS family transcription factorGABP alpha. J. Mol. Biol. 377:636–46

11. Demonstrates thestructural role of theB-box in an ELK4/SRFDNA ternary complexformation.

11. Hassler M, Richmond TJ. 2001. The B-box dominates SAP-1-SRF interactions in the structureof the ternary complex. EMBO J. 20:3018–28

12. Bojovic BB, Hassell JA. 2001. The PEA3 ETS transcription factor comprises multiple domains thatregulate transactivation and DNA binding. J. Biol. Chem. 276:4509–21

13. Augustijn KD, Duval DL, Wechselberger R, Kaptein R, Gutierrez-Hartmann A, van der Vliet PC.2002. Structural characterization of the PIT-1/ETS-1 interaction: PIT-1 phosphorylation regulatesPIT-1/ETS-1 binding. Proc. Natl. Acad. Sci. USA 99:12657–62

14. Macauley MS, Errington WJ, Scharpf M, Mackereth CD, Blaszczak AG, et al. 2006. Beads-on-a-string,characterization of ETS-1 sumoylated within its flexible N-terminal sequence. J. Biol. Chem. 281:4164–72

15. Lens Z, Dewitte F, Monte D, Baert JL, Bompard C, et al. 2010. Solution structure of the N-terminaltransactivation domain of ERM modified by SUMO-1. Biochem. Biophys. Res. Commun. 399:104–10

16. Dyson HJ, Wright PE. 2005. Intrinsically unstructured proteins and their functions. Nat. Rev. Mol. CellBiol. 6:197–208

17. Tootle TL, Rebay I. 2005. Post-translational modifications influence transcription factor activity: aview from the ETS superfamily. Bioessays 27:285–98

18. Charlot C, Dubois-Pot H, Serchov T, Tourrette Y, Wasylyk B. 2010. A review of post-translationalmodifications and subcellular localization of ETS transcription factors: possible connection with cancerand involvement in the hypoxic response. Methods Mol. Biol. 647:3–30

19. Hart AH, Reventar R, Bernstein A. 2000. Genetic analysis of ETS genes in C. elegans. Oncogene 19:6400–8

20. Hsu T, Schulz RA. 2000. Sequence and functional properties of Ets genes in the model organismDrosophila. Oncogene 19:6409–16

21. Schmid C, Schwarz V, Hutter H. 2006. AST-1, a novel ETS-box transcription factor, controls axonguidance and pharynx development in C. elegans. Dev. Biol. 293:403–13

22. Miley GR, Fantz D, Glossip D, Lu X, Saito RM, et al. 2004. Identification of residues of the Caenorhab-ditis elegans LIN-1 ETS domain that are necessary for DNA binding and regulation of vulval cell fates.Genetics 167:1697–709

23. Thyagarajan B, Blaszczak AG, Chandler KJ, Watts JL, Johnson WE, Graves BJ. 2010. ETS-4 is atranscriptional regulator of life span in Caenorhabditis elegans. PLoS Genet. 6:e1001125

24. Brunner D, Ducker K, Oellers N, Hafen E, Scholz H, Klambt C. 1994. The ETS domain proteinpointed-P2 is a target of MAP kinase in the sevenless signal transduction pathway. Nature 370:386–89

25. Fletcher JC, Burtis KC, Hogness DS, Thummel CS. 1995. The Drosophila E74 gene is required formetamorphosis and plays a role in the polytene chromosome puffing response to ecdysone. Development121:1455–65

26. Zhang J, Graham TGW, Vivekanand P, Cote L, Cetera M, Rebay I. 2010. Sterile alpha motif domain-mediated self-association plays an essential role in modulating the activity of the Drosophila ETS familytranscriptional repressor Yan. Mol. Cell. Biol. 30:1158–70

27. Ciau-Uitz A, Pinheiro P, Gupta R, Enver T, Patient R. 2010. Tel1/ETV6 specifies blood stem cellsthrough the agency of VEGF signaling. Dev. Cell 18:569–78

28. Kruse EA, Loughran SJ, Baldwin TM, Josefsson EC, Ellis S, et al. 2009. Dual requirement for the Etstranscription factors Fli-1 and Erg in hematopoietic stem cells and the megakaryocyte lineage. Proc.Natl. Acad. Sci. USA 106:13814–19

29. Wilson NK, Foster SD, Wang X, Knezevic K, Schutte J, et al. 2010. Combinatorial transcriptionalcontrol in blood stem/progenitor cells: genome-wide analysis of ten major transcriptional regulators.Cell Stem Cell 7:532–44

462 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 27: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

30. Wei G, Srinivasan R, Cantemir-Stone CZ, Sharma SM, Santhanam R, et al. 2009. Ets1 and Ets2 arerequired for endothelial cell survival during embryonic angiogenesis. Blood 114:1123–30

31. De Val S, Chi NC, Meadows SM, Minovitsky S, Anderson JP, et al. 2008. Combinatorial regulation ofendothelial gene expression by Ets and Forkhead transcription factors. Cell 135:1053–64

32. De Val S, Black BL. 2009. Transcriptional control of endothelial cell development. Dev. Cell 16:180–9533. Costello P, Nicolas R, Willoughby J, Wasylyk B, Nordheim A, Treisman R. 2010. Ternary complex

factors SAP-1 and Elk-1, but not Net, are functionally equivalent in thymocyte development. J. Immunol.185:1082–92

34. Hollenhorst PC, Jones DA, Graves BJ. 2004. Expression profiles frame the promoter specificity dilemmaof the Ets family of transcription factors. Nucleic Acids Res. 32:5693–702

35. Galang CK, Muller WJ, Foos G, Oshima RG, Hauser CA. 2004. Changes in the expression of manyEts family transcription factors and of potential target genes in normal mammary tissue and tumors.J. Biol. Chem. 279:11281–92

36. Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, et al. 2005. Recurrent fusion ofTMPRSS2 and Ets transcription factor genes in prostate cancer. Science 310:644–8

37. Liang H, Mao XH, Olejniczak ET, Nettesheim DG, Yu LP, et al. 1994. Solution structure of the Etsdomain of Fli-1 when bound to DNA. Nat. Struct. Biol. 1:871–6

38. Kodandapani R, Pio F, Ni CZ, Piccialli G, Klemsz M, et al. 1996. A new pattern for helix-turn-helixrecognition revealed by the PU.1 ETS-domain-DNA complex. Nature 380:456–60

39. Werner MH, Clore GM, Fisher CL, Fisher RJ, Trinh L, et al. 1997. Correction of the NMR structureof the ETS1/DNA complex. J. Biomol. NMR 10:317–28

40. Batchelor AH, Piper DE, de la Brousse FC, McKnight SL, Wolberger C. 1998. The structure of GABPalpha/beta: An Ets domain ankyrin repeat heterodimer bound to DNA. Science 279:1037–41

41. Mo Y, Vaessen B, Johnston K, Marmorstein R. 1998. Structures of SAP-1 bound to DNA targets fromthe E74 and c-fos promoters: Insights into DNA sequence discrimination by Ets proteins. Mol. Cell2:201–12

42. Mo Y, Vaessen B, Johnston K, Marmorstein R. 2000. Structure of the Elk-1-DNA complex reveals howDNA-distal residues affect Ets domain recognition of DNA. Nat. Struct. Biol. 7:292–7

43. Garvie CW, Hagman J, Wolberger C. 2001. Structural studies of Ets-1/Pax5 complex formation onDNA. Mol. Cell. Biol. 8:1267–76

44. Mo Y, Ho W, Johnston K, Marmorstein R. 2001. Crystal structure of a ternary SAP-1/SRF/c-fos SIREDNA complex. J. Mol. Biol. 314:495–506

45. Garvie CW, Pufall MA, Graves BJ, Wolberger C. 2002. Structural analysis of the autoinhibition ofEts-1 and its role in protein partnerships. J. Biol. Chem. 277:45529–36

46. Escalante CR, Brass AL, Pongubala JM, Shatova E, Shen L, et al. 2002. Crystal structure of PU.1/IRF-4/DNA ternary complex. Mol. Cell 10:1097–105

47. Wang YZ, Feng L, Said M, Balderman S, Fayazi Z, et al. 2005. Analysis of the 2.0 angstrom crystalstructure of the protein-DNA complex of the human PDEF Ets domain bound to the prostate specificantigen regulatory site. Biochemistry 44:7095–106

48. Lee GM, Donaldson LW, Pufall MA, Kang HS, Pot I, et al. 2005. The structural and dynamic basis ofEts-1 DNA binding autoinhibition. J. Biol. Chem. 280:7088–99

49. Lamber EP, Vanhille L, Textor LC, Kachalova GS, Sieweke MH, Wilmanns M. 2008. Regulation ofthe transcription factor Ets-1 by DNA-mediated homo-dimerization. EMBO J. 27:2006–17

50. Green SM, Coyne HJ, McIntosh LP, Graves BJ. 2010. DNA binding by the Ets protein TEL (ETV6)is regulated by autoinhibition and self-association. J. Biol. Chem. 285:18496–504

51. Agarkar VB, Babayeva ND, Wilder PJ, Rizzino A, Tahirov TH. 2010. Crystal structure of mouse Elf3C-terminal DNA-binding domain in complex with Type II TGF-beta receptor promoter DNA. J. Mol.Biol. 397:278–89

52. Gajiwala KS, Burley SK. 2000. Winged helix proteins. Curr. Opin. Struct. Biol. 10:110–653. Szymczyna BR, Arrowsmith CH. 2000. DNA binding specificity studies of four Ets proteins support

an indirect read-out mechanism of protein-DNA recognition. J. Biol. Chem. 275:28363–7054. Poon GMK, Macgregor RB. 2003. Base coupling in sequence-specific site recognition by the Ets domain

of murine PU.1. J. Mol. Biol. 328:805–19

www.annualreviews.org • ETS Family of Transcription Factors 463

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 28: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

55. Brown TA, McKnight SL. 1992. Specificities of protein-protein and protein-DNA interaction of GABPalpha and two newly defined ets-related proteins. Genes Dev. 6:2502–12

56. Nye JA, Petersen JM, Gunther CV, Jonsen MD, Graves BJ. 1992. Interaction of murine Ets-1 withGGA-binding sites establishes the Ets domain as a new DNA-binding motif. Genes Dev. 6:975–90

57. Ray-Gallet D, Mao C, Tavitian A, Moreau-Gachelin F. 1995. DNA binding specificities of Spi-1/PU.1and Spi-B transcription factors and identification of a Spi-1/Spi-B binding site in the c-fes/c-fps pro-moter. Oncogene 11:303–13

58. Shore P, Sharrocks AD. 1995. The Ets-domain transcription factors Elk-1 and SAP-1 exhibit differentialDNA binding specificities. Nucleic Acids Res. 23:4698–706

59. John S, Marais R, Child R, Light Y, Leonard WJ. 1996. Importance of low affinity Elf-1 sites in theregulation of lymphoid-specific inducible gene expression. J. Exp. Med. 183:743–50

60. Choi YS, Sinha S. 2006. Determination of the consensus DNA-binding sequence and a transcriptionalactivation domain for ESE-2. Biochem. J. 398:497–507

61. Mao X, Miesfeldt S, Yang H, Leiden JM, Thompson CB. 1994. The FLI-1 and chimeric EWS-FLI-1oncoproteins display similar DNA binding specificities. J. Biol. Chem. 269:18216–22

62. Provides acomprehensive analysisof in vitro derived DNAbinding sequences forvertebrate ETSproteins.

62. Wei GH, Badis G, Berger MF, Kivioja T, Palin K, et al. 2010. Genome-wide analysis of ETS-family DNA-binding in vitro and in vivo. EMBO J. 29:2147–60

63. Wang CY, Petryniak B, Ho IC, Thompson CB, Leiden JM. 1992. Evolutionarily conserved Ets familymembers display distinct DNA binding specificities. J. Exp. Med. 175:1391–9

64. Bosselut R, Levin J, Adjadj E, Ghysdael J. 1993. A single amino-acid substitution in the Ets domainalters core DNA binding specificity of Ets1 to that of the related transcription factors Elf1 and E74.Nucleic Acids Res. 21:5184–91

65. Shows redundantETS protein binding athousekeepingpromoters andgenome-wide evidencefor ETS1/RUNXpartnership.

65. Hollenhorst PC, Chandler KJ, Poulsen RL, Johnson WE, Speck NA, Graves BJ. 2009. DNAspecificity determinants associate with distinct transcription factor functions. PLoS Genet.

5:e100077866. Oettgen P, Finger E, Sun Z, Akbarali Y, Thamrongsak U, et al. 2000. PDEF, a novel prostate epithelium-

specific Ets transcription factor, interacts with the androgen receptor and activates prostate-specificantigen gene expression. J. Biol. Chem. 275:1216–25

67. Donaldson LW, Petersen JM, Graves BJ, McIntosh LP. 1996. Solution structure of the ETS domainfrom murine Ets-1: a winged helix-turn-helix DNA binding motif. EMBO J. 15:125–34

68. Wasylyk C, Bradford AP, Gutierrez-Hartmann A, Wasylyk B. 1997. Conserved mechanisms of Rasregulation of evolutionary related transcription factors, Ets1 and Pointed P2. Oncogene 14:899–913

69. Pufall MA, Graves BJ. 2002. Autoinhibitory domains: modular effectors of cellular regulation. Annu.Rev. Cell Dev. Biol. 18:421–62

70. Hagman J, Grosschedl R. 1992. An inhibitory carboxyl-terminal domain in Ets-1 and Ets-2 mediatesdifferential binding of ETS family factors to promoter sequences of the mb-1 gene. Proc. Natl. Acad.Sci. USA 89:8889–93

71. Wasylyk C, Kerckaert JP, Wasylyk B. 1992. A novel modulator domain of Ets transcription factors.Genes Dev. 6:965–74

72. Lim F, Kraut N, Framptom J, Graf T. 1992. DNA binding by c-Ets-1, but not v-Ets, is repressed byan intramolecular mechanism. EMBO J. 11:643–52

73. Lionneton F, Lelievre E, Baillat D, Stehelin D, Soncin F. 2003. Characterization and functional analysisof the p42Ets-1 variant of the mouse Ets-1 transcription factor. Oncogene 22:9156–64

74. Rabault B, Ghysdael J. 1994. Calcium-induced phosphorylation of ETS1 inhibits its specific DNAbinding activity. J. Biol. Chem. 269:28143–51

75. Cowley DO, Graves BJ. 2000. Phosphorylation represses Ets-1 DNA binding by reinforcing autoinhi-bition. Genes Dev. 14:366–76

76. Petersen JM, Skalicky JJ, Donaldson LW, Mcintosh LP, Alber T, Graves BJ. 1995. Modulation of tran-scription factor ETS-1 DNA-binding—DNA-induced unfolding of an alpha-helix. Science 269:1866–69

77. Kalodimos CG, Biris N, Bonvin AM, Levandoski MM, Guennuegues M, et al. 2004. Structure andflexibility adaptation in nonspecific and specific protein-DNA complexes. Science 305:386–89

464 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 29: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

78. Kalodimos CG, Boelens R, Kaptein R. 2004. Toward an integrated model of protein-DNA recognitionas inferred from NMR studies on the Lac repressor system. Chem. Rev. 104:3567–86

79. Demonstrates roleof dynamics inregulation of ETS1DNA binding by aphosphorylatedunstructured region.

79. Pufall MA, Lee GM, Nelson ML, Kang HS, Velyvis A, et al. 2005. Variable control of Ets-1DNA binding by multiple phosphates in an unstructured region. Science 309:142–45

80. Lee GM, Pufall MA, Meeker CA, Kang HS, Graves BJ, McIntosh LP. 2008. The affinity of Ets-1 forDNA is modulated by phosphorylation through transient interactions of an unstructured region. J. Mol.Biol. 382:1014–30

81. Liu H, Grundstrom T. 2002. Calcium regulation of GM-CSF by calmodulin-dependent kinase IIphosphorylation of Ets1. Mol. Biol. Cell 13:4497–507

82. Gardner KH, Montminy M. 2005. Can you hear me now? Regulating transcriptional activators byphosphorylation. Sci. STKE 2005:pe44

83. Kamberaj H, van der Vaart A. 2009. Correlated motions and interactions at the onset of the DNA-induced partial unfolding of Ets-1. Biophys. J. 96:1307–17

84. Greenall A, Willingham N, Cheung E, Boam DS, Sharrocks AD. 2001. DNA binding by the ETS-domain transcription factor PEA3 is regulated by intramolecular and intermolecular protein-proteininteractions. J. Biol. Chem. 276:16207–15

85. de Launoit Y, Baert JL, Chotteau A, Monte D, Defossez PA, et al. 1997. Structure-function relationshipsof the PEA3 group of Ets-related transcription factors. Biochem. Mol. Med. 61:127–35

86. Kopp JL, Wilder PJ, Desler M, Kinarsky L, Rizzino A. 2007. Different domains of the transcriptionfactor ELF3 are required in a promoter-specific manner and multiple domains control its binding toDNA. J. Biol. Chem. 282:3027–41

87. Oettgen P, Kas K, Dube A, Gu X, Grall F, et al. 1999. Characterization of ESE-2, a novel ESE-1-relatedEts transcription factor that is restricted to glandular epithelium and differentiated keratinocytes. J. Biol.Chem. 274:29439–52

88. Sharrocks AD. 2002. Complexities in ETS-domain transcription factor function and regulation: lessonsfrom the TCF (ternary complex factor) subfamily. The Colworth Medal Lecture. Biochem. Soc. Trans.30:1–9

89. Buchwalter G, Gross C, Wasylyk B. 2004. Ets ternary complex transcription factors. Gene 324:1–1490. Stinson J, Inoue T, Yates P, Clancy A, Norton JD, Sharrocks AD. 2003. Regulation of TCF ETS-

domain transcription factors by helix-loop-helix motifs. Nucleic Acids Res. 31:4717–2891. Yang SH, Shore P, Willingham N, Lakey JH, Sharrocks AD. 1999. The mechanism of phosphorylation-

inducible activation of the ETS-domain transcription factor Elk-1. EMBO J. 18:5666–7492. Slupsky CM, Gentile LN, Donaldson LW, Mackereth CD, Seidel JJ, et al. 1998. Structure of the Ets-1

pointed domain and mitogen-activated protein kinase phosphorylation site. Proc. Natl. Acad. Sci. USA95:12129–34

93. Discovery of thepolymerizationproperties of the PNTdomain of ETV6.

93. Kim CA, Phillips ML, Kim W, Gingery M, Tran HH, et al. 2001. Polymerization of the SAMdomain of TEL in leukemogenesis and transcriptional repression. EMBO J. 20:4173–82

94. Tran HH, Kim CA, Faham S, Siddall M-C, Bowie JU. 2002. Native interface of the SAM domainpolymer of TEL. BMC Struct. Biol. 2:5

95. Mackereth CD, Scharpf M, Gentile LN, MacIntosh SE, Slupsky CM, McIntosh LP. 2004. Diversityin structure and function of the Ets family PNT domains. J. Biol. Mol. 342:1249–64

96. Qiao F, Song HY, Kim CA, Sawaya MR, Hunter JB, et al. 2004. Derepression by depolymerization:Structural insights into the regulation of Yan by Mae. Cell 118:163–73

97. Demonstratesmechanism forphosphorylation-dependent binding ofCBP and ETS1 viacharge complementarityand conformationaldynamics.

97. Nelson ML, Kang HS, Lee GM, Blaszczak AG, Lau DKW, et al. 2010. Ras signaling requiresdynamic properties of Ets1 for phosphorylation-enhanced binding to coactivator CBP. Proc.

Natl. Acad. Sci. USA 107:10026–3198. Kim CA, Bowie JU. 2003. SAM domains: uniform structure, diversity of function. Trends Biochem. Sci.

28:625–2899. Klambt C. 1993. The Drosophila gene pointed encodes two ETS-like proteins which are involved in the

development of the midline glial cells. Development 117:163–76100. Seidel JJ, Graves BJ. 2002. An ERK2 docking site in the Pointed domain distinguishes a subset of ETS

transcription factors. Genes Dev. 16:127–37

www.annualreviews.org • ETS Family of Transcription Factors 465

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 30: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

101. Waas WF, Dalby KN. 2002. Transient protein-protein interactions and a random-ordered kineticmechanism for the phosphorylation of a transcription factor by extracellular-regulated protein kinase2. J. Biol. Chem. 277:12532–40

102. Qiao F, Harada B, Song HY, Whitelegge J, Courey AJ, Bowie JU. 2006. Mae inhibits Pointed-P2transcriptional activity by blocking its MAPK docking site. EMBO J. 25:70–79

103. Biondi RM, Nebreda AR. 2003. Signalling specificity of Ser/Thr protein kinases through docking-site-mediated interactions. Biochem. J. 372:1–13

104. Sharrocks AD, Yang SH, Galanis A. 2000. Docking domains and substrate-specificity determinationfor MAP kinases. Trends Biochem. Sci. 25:448–53

105. Rainey MA, Callaway K, Barnes R, Wilson B, Dalby KN. 2005. Proximity-induced catalysis by theprotein kinase ERK2. J. Am. Chem. Soc. 127:10494–95

106. Goldsmith EJ, Akella R, Min X, Zhou T, Humphreys JM. 2007. Substrate and docking interactions inserine/threonine protein kinases. Chem. Rev. 107:5065–81

107. Akella R, Moon TM, Goldsmith EJ. 2008. Unique MAP Kinase binding sites. Biochim. Biophys. Acta1784:48–55

108. Presents a kineticand structuralmechanism for thedocking of ETS1 andMAP kinase ERK2.

108. Callaway K, Waas WF, Rainey MA, Ren PY, Dalby KN. 2010. Phosphorylation of the tran-scription factor ETS-1 by ERK2: rapid dissociation of ADP and phospho-ETS-1. Biochemistry

49:3619–30109. Callaway KA, Rainey MA, Riggs AF, Abramczyk O, Dalby KN. 2006. Properties and regulation of a

transiently assembled ERK2·Ets-1 signaling complex. Biochemistry 45:13719–33110. Abramczyk O, Rainey MA, Barnes R, Martin L, Dalby KN. 2007. Expanding the repertoire of an ERK2

recruitment site: cysteine footprinting identifies the D-recruitment site as a mediator of Ets-1 binding.Biochemistry 46:9174–86

111. Yang SH, Sharrocks AD, Whitmarsh AJ. 2003. Transcriptional regulation by the MAP kinase signalingcascades. Gene 320:3–21

112. Foulds CE, Nelson ML, Blaszczak AG, Graves BJ. 2004. Ras/mitogen-activated protein kinase signalingactivates Ets-1 and Ets-2 by CBP/p300 recruitment. Mol. Cell. Biol. 24:10954–64

113. Wojciak JM, Martinez-Yamout MA, Dyson HJ, Wright PE. 2009. Structural basis for recruitment ofCBP/p300 coactivators by STAT1 and STAT2 transactivation domains. EMBO J. 28:948–58

114. Brown LA, Yang SH, Hair A, Galanis A, Sharrocks AD. 1999. Molecular characterization of a zebrafishTCF ETS-domain transcription factor. Oncogene 18:7985–93

115. Li QJ, Yang SH, Maeda Y, Sladek FM, Sharrocks AD, Martins-Green M. 2003. MAP kinasephosphorylation-dependent activation of Elk-1 leads to activation of the co-activator p300. EMBOJ. 22:281–91

116. Yang SH, Jaffray E, Hay RT, Sharrocks AD. 2003. Dynamic interplay of the SUMO and ERK pathwaysin regulating Elk-1 transcriptional activity. Mol. Cell 12:63–74

117. Bai Y, Srinivasan L, Perkins L, Atchison ML. 2005. Protein acetylation regulates both PU.1 transacti-vation and Ig kappa 3′ enhancer activity. J. Immunol. 175:5160–69

118. Aikawa Y, Katsumoto T, Zhang P, Shima H, Shino M, et al. 2010. PU.1-mediated upregulation ofCSF1R is crucial for leukemia stem cell potential induced by MOZ-TIF2. Nat. Med. 16:580–85

119. Goel A, Janknecht R. 2004. Concerted activation of ETS protein ER81 by p160 coactivators, theacetyltransferase p300 and the receptor tyrosine kinase HER2/Neu. J. Biol. Chem. 279:14909–16

120. Papoutsopoulou S, Janknecht R. 2000. Phosphorylation of ETS transcription factor ER81 in a complexwith its coactivators CREB-binding protein and p300. Mol. Cell. Biol. 20:7300–10

121. Balamotis MA, Pennella MA, Stevens JL, Wasylyk B, Belmont AS, Berk AJ. 2009. Complexity intranscription control at the activation domain-mediator interface. Sci. Signal. 2:RA20

122. Cantin GT, Stevens JL, Berk AJ. 2003. Activation domain-mediator interactions promote transcriptionpreinitiation complex assembly on promoter DNA. Proc. Natl. Acad. Sci. USA 100:12003–8

123. Stevens JL, Cantin GT, Wang G, Shevchenko A, Berk AJ. 2002. Transcription control by E1A andMAP kinase pathway via Sur2 Mediator subunit. Science 296:755–58

124. Wang W, Huang L, Huang Y, Yin JW, Berk AJ, et al. 2009. Mediator MED23 links insulin signalingto the adipogenesis transcription cascade. Dev. Cell 16:764–71

466 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 31: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

125. Potter MD, Buijs A, Kreider B, van Rompaey L, Grosveld GC. 2000. Identification and characterizationof a new human ETS-family transcription factor, TEL2, that is expressed in hematopoietic tissues andcan associate with TEL1/ETV6. Blood 95:3341–48

126. Meruelo AD, Bowie JU. 2009. Identifying polymer-forming SAM domains. Proteins 74:1–5127. Vivekanand P, Rebay I. 2006. Intersection of signal transduction pathways and development. Annu.

Rev. Genet. 40:139–57128. Baker DA, Mille-Baker B, Wainwright SM, Ish-Horowicz D, Dibb NJ. 2001. Mae mediates MAP

kinase phosphorylation of Ets transcription factors in Drosophila. Nature 411:330–34129. Tootle TL, Lee PS, Rebay I. 2003. CRM1-mediated nuclear export and regulated activity of the receptor

tyrosine kinase antagonist YAN require specific interactions with MAE. Development 130:845–57130. Farnham PJ. 2009. Insights from genomic profiling of transcription factors. Nat. Rev. Genet. 10:605–16131. Barski A, Zhao K. 2009. Genomic location analysis by ChIP-Seq. J. Cell. Biochem. 107:11–18132. Hollenhorst PC, Shah AA, Hopkins C, Graves BJ. 2007. Genome-wide analyses reveal properties of

redundant and specific promoter occupancy within the ETS gene family. Genes Dev. 21:1882–94

133. Genome-wideEWS-FLI1 occupancyshows route tospecificity for oncogenicETS protein via use ofmicrosatellites.133. Gangwal K, Sankar S, Hollenhorst PC, Kinsey M, Haroldsen SC, et al. 2008. Microsatellites as

EWS/FLI response elements in Ewing’s sarcoma. Proc. Natl. Acad. Sci. USA 105:10149–54134. Genome-wideELK1 occupancydisplays redundant andspecific binding patternsand evidence forELK1/SRF partnership.

134. Boros J, Donaldson IJ, O’Donnell A, Odrowaz ZA, Zeef L, et al. 2009. Elucidation of the ELK1target gene network reveals a role in the coordinate regulation of core components of the generegulation machinery. Genome Res. 19:1963–73

135. Kubosaki A, Lindgren G, Tagami M, Simon C, Tomaru Y, et al. 2010. The combination of geneperturbation assay and ChIP-chip reveals functional direct target genes for IRF8 in THP-1 cells.Mol. Immunol. 47:2295–302

136. Valouev A, Johnson DS, Sundquist A, Medina C, Anton E, et al. 2008. Genome-wide analysis oftranscription factor binding sites based on ChIP-Seq data. Nat. Methods 5:829–34

137. Guillon N, Tirode F, Boeva V, Zynovyev A, Barillot E, Delattre O. 2009. The oncogenic EWS-FLI1protein binds in vivo GGAA microsatellite sequences with potential transcriptional activation function.PLoS One 4:e4932

138. Ghisletti S, Barozzi I, Mietton F, Polletti S, De Santa F, et al. 2010. Identification and characterization ofenhancers controlling the inflammatory gene expression program in macrophages. Immunity 32:317–28

139. Proposes amechanistic route forERG-drivenoncogenesis througheffects on anepigenetics program.

139. Yu J, Yu J, Mani RS, Cao Q, Brenner CJ, et al. 2010. An integrated network of androgenreceptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer

Cell 17:443–54140. Chi P, Chen Y, Zhang L, Guo X, Wongvipat J, et al. 2010. ETV1 is a lineage survival factor that

cooperates with KIT in gastrointestinal stromal tumours. Nature 467:849–53141. Boros J, O’Donnell A, Donaldson IJ, Kasza A, Zeef L, Sharrocks AD. 2009. Overlapping promoter

targeting by Elk-1 and other divergent ETS-domain transcription factor family members. Nucleic AcidsRes. 37:7368–80

142. Heintzman ND, Stuart RK, Hon G, Fu Y, Ching CW, et al. 2007. Distinct and predictive chromatinsignatures of transcriptional promoters and enhancers in the human genome. Nat. Genet. 39:311–18

143. Boyle AP, Davis S, Shulha HP, Meltzer P, Margulies EH, et al. 2008. High-resolution mapping andcharacterization of open chromatin across the genome. Cell 132:311–22

144. Fitzgerald PC, Shlyakhtenko A, Mir AA, Vinson C. 2004. Clustering of DNA sequences in humanpromoters. Genome Res. 14:1562–74

145. Bina M, Wyss P, Ren W, Szpankowski W, Thomas E, et al. 2004. Exploring the characteristics ofsequence elements in proximal promoters of human genes. Genomics 84:929–40

146. Xie X, Lu J, Kulbokas EJ, Golub TR, Mootha V, et al. 2005. Systematic discovery of regulatory motifsin human promoters and 3′ UTRs by comparison of several mammals. Nature 434:338–45

147. Pio F, Assa-Munt N, Yguerabide J, Maki RA. 1999. Mutants of ETS domain PU.1 and GGAA/Trecognition: free energies and kinetics. Protein Sci. 8:2098–109

148. Yokomori N, Kobayashi R, Moore R, Sueyoshi T, Negishi M. 1995. A DNA methylation site in themale-specific P450 (Cyp 2 d-9) promoter and binding of the heteromeric transcription factor GABP.Mol. Cell. Biol. 15:5355–62

www.annualreviews.org • ETS Family of Transcription Factors 467

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 32: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

149. Gaston K, Fried M. 1995. CpG methylation has differential effects on the binding of YY1 and ETSproteins to the bi-directional promoter of the Surf-1 and Surf-2 genes. Nucleic Acids Res. 23:901–9

150. Massie CE, Mills IG. 2009. Chromatin immunoprecipitation (ChIP) methodology and readouts.Methods Mol. Biol. 505:123–37

151. Li R, Pei H, Watson DK. 2000. Regulation of Ets function by protein-protein interactions. Oncogene19:6514–23

152. Verger A, Duterque-Coquillaud M. 2002. When Ets transcription factors meet their partners. Bioessays24:362–70

153. Cooper SJ, Trinklein ND, Nguyen L, Myers RM. 2007. Serum response factor binding sites differ inthree human cell types. Genome Res. 17:136–44

154. Brass AL, Zhu AQ, Singh H. 1999. Assembly requirements of PU.1-Pip (IRF-4) activator complexes:inhibiting function in vivo using fused dimers. EMBO J. 18:977–91

155. Yee AA, Yin P, Siderovski DP, Mak TW, Litchfield DW, Arrowsmith CH. 1998. Cooperative inter-action between the DNA-binding domains of PU.1 and IRF4. J. Mol. Biol. 279:1075–83

156. Goetz TL, Gu TL, Speck NA, Graves BJ. 2000. Auto-inhibition of Ets-1 is counteracted by DNAbinding cooperativity with core-binding factor alpha 2. Mol. Cell. Biol. 20:81–90

157. Gu TL, Goetz TL, Graves BJ, Speck NA. 2000. Auto-inhibition and partner proteins, core-bindingfactor beta (CBF beta) and Ets-1, modulate DNA finding by CBF alpha 2 (AML1). Mol. Cell. Biol.20:91–103

158. Wotton D, Ghysdael J, Wang S, Speck NA, Owen MJ. 1994. Cooperative binding of Ets-1 and corebinding factor to DNA. Mol. Cell. Biol. 14:840–50

159. Muthusamy N, Barton K, Leiden JM. 1995. Defective activation and survival of T cells lacking theEts-1 transcription factor. Nature 377:639–42

160. Mao S, Frank RC, Zhang J, Miyazaki Y, Nimer SD. 1999. Functional and physical interactions be-tween AML1 proteins and an ETS protein, MEF: implications for the pathogenesis of t(8;21)-positiveleukemias. Mol. Cell. Biol. 19:3635–44

161. Huang H, Yu M, Akie TE, Moran TB, Woo AJ, et al. 2009. Differentiation-dependent interactionsbetween RUNX-1 and FLI-1 during megakaryocyte development. Mol. Cell. Biol. 29:4103–15

162. Fitzsimmons D, Lutz R, Wheat W, Chamberlin HM, Hagman J. 2001. Highly conserved amino acidsin Pax and Ets proteins are required for DNA binding and ternary complex assembly. Nucleic Acids Res.29:4154–65

163. Fitzsimmons D, Lukin K, Lutz R, Garvie CW, Wolberger C, Hagman J. 2009. Highly cooperativerecruitment of Ets-1 and release of autoinhibition by Pax5. J. Mol. Biol. 392:452–64

164. Golub TR, Barker GF, Stegmaier K, Gilliland DG. 1997. The TEL gene contributes to the pathogenesisof myeloid and lymphoid leukemias by diverse molecular genetic mechanisms. Curr. Top. Microbiol.Immunol. 220:67–79

165. Cazzaniga G, Daniotti M, Tosi S, Giudici G, Aloisi A, et al. 2001. The paired box domain gene PAX5is fused to ETV6/TEL in an acute lymphoblastic leukemia case. Cancer Res. 61:4666–70

166. Fazio G, Palmi C, Rolink A, Biondi A, Cazzaniga G. 2008. PAX5/TEL acts as a transcriptional repressorcausing down-modulation of CD19, enhances migration to CXCL12, and confers survival advantagein pre-BI cells. Cancer Res. 68:181–89

167. Baillat D, Begue A, Stehelin D, Aumercier M. 2002. ETS-1 transcription factor binds cooperativelyto the palindromic head to head ETS-binding sites of the stromelysin-1 promoter by counteractingautoinhibition. J. Biol. Chem. 277:29386–98

168. Leprivier G, Baillat D, Begue A, Hartmann B, Aumercier M. 2009. Ets-1 p51 and p42 isoforms differ-entially modulate Stromelysin-1 promoter according to induced DNA bend orientation. Nucleic AcidsRes. 37:4341–52

169. Gotea V, Visel A, Westlund JM, Nobrega MA, Pennacchio LA, Ovcharenko I. 2010. Homotypic clustersof transcription factor binding sites are a key component of human promoters and enhancers. GenomeRes. 20:565–77

170. Helgeson BE, Tomlins SA, Shah N, Laxman B, Cao Q, et al. 2008. Characterization of TM-PRSS2:ETV5 and SLC45A3:ETV5 gene fusions in prostate cancer. Cancer Res. 68:73–80

468 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 33: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

171. Tomlins SA, Mehra R, Rhodes DR, Smith LR, Roulston D, et al. 2006. TMPRSS2:ETV4 gene fusionsdefine a third molecular subtype of prostate cancer. Cancer Res. 66:3396–400

172. Tomlins SA, Laxman B, Dhanasekaran SM, Helgeson BE, Cao X, et al. 2007. Distinct classes ofchromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature 448:595–99

173. Cai C, Hsieh CL, Omwancha J, Zheng Z, Chen SY, et al. 2007. ETV1 is a novel androgen receptor-regulated gene that mediates prostate cancer cell invasion. Mol. Endocrinol. 21:1835–46

174. Tomlins SA, Laxman B, Varambally S, Cao X, Yu J, et al. 2008. Role of the TMPRSS2-ERG genefusion in prostate cancer. Neoplasia 10:177–88

175. Zong Y, Xin L, Goldstein AS, Lawson DA, Teitell MA, Witte ON. 2009. ETS family transcriptionfactors collaborate with alternative signaling pathways to induce carcinoma from adult murine prostatecells. Proc. Natl. Acad. Sci. USA 106:12465–70

176. Goldstein AS, Huang J, Guo C, Garraway IP, Witte ON. 2010. Identification of a cell of origin forhuman prostate cancer. Science 329:568–71

177. Massie CE, Adryan B, Barbosa-Morais NL, Lynch AG, Tran MG, et al. 2007. New androgen receptorgenomic targets show an interaction with the ETS1 transcription factor. EMBO Rep. 8:871–78

178. Zhang M, Magit D, Sager R. 1997. Expression of maspin in prostate cells is regulated by a positive Etselement and a negative hormonal responsive element site recognized by androgen receptor. Proc. Natl.Acad. Sci. USA 94:5673–78

179. Schneikert J, Peterziel H, Defossez PA, Klocker H, Launoit Y, Cato AC. 1996. Androgen receptor–Etsprotein interaction is a novel mechanism for steroid hormone-mediated down-modulation of matrixmetalloproteinase expression. J. Biol. Chem. 271:23907–13

180. Rickman DS, Chen YB, Banerjee S, Pan Y, Yu J, et al. 2010. ERG cooperates with androgen receptorin regulating trefoil factor 3 in prostate cancer disease progression. Neoplasia 12:1031–40

181. Shin S, Kim TD, Jin F, van Deursen JM, Dehm SM, et al. 2009. Induction of prostatic intraepithelialneoplasia and modulation of androgen receptor by ETS variant 1/ETS-related protein 81. Cancer Res.69:8102–10

182. May WA, Denny CT. 1997. Biology of EWS/FLI and related fusion genes in Ewing’s sarcoma andprimitive neuroectodermal tumor. Curr. Top. Microbiol. Immunol. 220:143–50

183. Delattre O, Zucman J, Plougastel B, Desmaze C, Melot T, et al. 1992. Gene fusion with an ETSDNA-binding domain caused by chromosome translocation in human tumours. Nature 359:162–65

184. Janknecht R. 2005. EWS-ETS oncoproteins: the linchpins of Ewing tumors. Gene 363:1–14185. May WA, Lessnick SL, Braun BS, Klemsz M, Lewis BC, et al. 1993. The Ewing’s sarcoma EWS/FLI-1

fusion gene encodes a more potent transcriptional activator and is a more powerful transforming genethan FLI-1. Mol. Cell. Biol. 13:7393–98

186. Luo W, Gangwal K, Sankar S, Boucher KM, Thomas D, Lessnick SL. 2009. GSTM4 is a microsatellite-containing EWS/FLI target involved in Ewing’s sarcoma oncogenesis and therapeutic resistance.Oncogene 28:4126–32

187. Gangwal K, Close D, Enriquez CA, Hill CP, Lessnick SL. 2010. Emergent properties of EWS/FLIregulation via GGAA microsatellites in Ewing’s sarcoma. Genes Cancer 1:177–87

188. Kunderfranco P, Mello-Grand M, Cangemi R, Pellini S, Mensah A, et al. 2010. ETS transcriptionfactors control transcription of EZH2 and epigenetic silencing of the tumor suppressor gene Nkx3.1in prostate cancer. PLoS One 5:e10547

188a. Hollenhorst PC, Paul L, Ferris MW, Graves BJ. 2011. The ETS gene ETV4 is required for anchorage-independent growth and a cell proliferation gene expression program in PC3 prostate cells. Genes Cancer.1:1044–52

189. Jane-Valbuena J, Widlund HR, Perner S, Johnson LA, Dibner AC, et al. 2010. An oncogenic role forETV1 in melanoma. Cancer Res. 70:2075–84

190. Jothi R, Cuddapah S, Barski A, Cui K, Zhao K. 2008. Genome-wide identification of in vivo protein-DNA binding sites from ChIP-Seq data. Nucleic Acids Res. 36:5221–31

191. Wain HM, Bruford EA, Lovering RC, Lush MJ, Wright MW, Povey S. 2002. Guidelines for humangene nomenclature. Genomics 79:464–70

www.annualreviews.org • ETS Family of Transcription Factors 469

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 34: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

192. Thompson JD, Higgins DG, Gibson TJ. 1994. CLUSTAL W: improving the sensitivity of progressivemultiple sequence alignment through sequence weighting, position-specific gap penalties and weightmatrix choice. Nucleic Acids Res. 22:4673–80

193. Saitou N, Nei M. 1987. The neighbor-joining method: a new method for reconstructing phylogenetictrees. Mol. Biol. Evol. 4:406–25

194. Loughran SJ, Kruse EA, Hacking DF, de Graaf CA, Hyland CD, et al. 2008. The transcription factorErg is essential for definitive hematopoiesis and the function of adult hematopoietic stem cells. Nat.Immunol. 9:810–19

195. Garrett-Sinha LA, Su GH, Rao S, Kabak S, Hao Z, et al. 1999. PU.1 and Spi-B are required for normalB cell receptor-mediated signal transduction. Immunity 10:399–408

196. Zhang Z, Verheyden JM, Hassell JA, Sun X. 2009. FGF-regulated Etv genes are essential for repressingShh expression in mouse limb buds. Dev. Cell 16:607–13

197. Iwasaki H, Somoza C, Shigematsu H, Duprez EA, Iwasaki-Arai J, et al. 2005. Distinctive and in-dispensable roles of PU.1 in maintenance of hematopoietic stem cells and their differentiation. Blood106:1590–600

198. Kamath MB, Houston IB, Janovski AJ, Zhu X, Gowrisankar S, et al. 2008. Dose-dependent repression ofT-cell and natural killer cell genes by PU.1 enforces myeloid and B-cell identity. Leukemia 22:1214–25

199. Rosenbauer F, Owens BM, Yu L, Tumang JR, Steidl U, et al. 2006. Lymphoid cell growth and trans-formation are suppressed by a key regulatory element of the gene encoding PU.1. Nat. Genet. 38:27–37

200. Dakic A, Metcalf D, Di Rago L, Mifsud S, Wu L, Nutt SL. 2005. PU.1 regulates the commitment ofadult hematopoietic progenitors and restricts granulopoiesis. J. Exp. Med. 201:1487–502

201. McKercher SR, Torbett BE, Anderson KL, Henkel GW, Vestal DJ, et al. 1996. Targeted disruptionof the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J. 15:5647–58

202. Houston IB, Kamath MB, Schweitzer BL, Chlon TM, DeKoter RP. 2007. Reduction in PU.1 activityresults in a block to B-cell development, abnormal myeloid proliferation, and neonatal lethality. Exp.Hematol. 35:1056–68

203. Scott EW, Simon MC, Anastasi J, Singh H. 1994. Requirement of transcription factor PU.1 in thedevelopment of multiple hematopoietic lineages. Science 265:1573–77

204. Su GH, Chen HM, Muthusamy N, Garrett-Sinha LA, Baunoch D, et al. 1997. Defective B cell receptor-mediated responses in mice lacking the Ets protein, Spi-B. EMBO J. 16:7118–29

205. Kohyama M, Ise W, Edelson BT, Wilker PR, Hildner K, et al. 2009. Role for Spi-C in the developmentof red pulp macrophages and splenic iron homeostasis. Nature 457:318–21

206. Wang LC, Kuo F, Fujiwara Y, Gilliland DG, Golub TR, Orkin SH. 1997. Yolk sac angiogenic defectand intra-embryonic apoptosis in mice lacking the Ets-related factor TEL. EMBO J. 16:4374–83

207. Hock H, Meade E, Medeiros S, Schindler JW, Valk PJ, et al. 2004. Tel/Etv6 is an essential and selectiveregulator of adult hematopoietic stem cell survival. Genes Dev. 18:2336–41

208. Ng AY, Waring P, Ristevski S, Wang C, Wilson T, et al. 2002. Inactivation of the transcriptionfactor Elf3 in mice results in dysmorphogenesis and altered differentiation of intestinal epithelium.Gastroenterology 122:1455–66

209. Zhou J, Chehab R, Tkalcevic J, Naylor MJ, Harris J, et al. 2005. Elf5 is essential for early embryogenesisand mammary gland development during pregnancy and lactation. EMBO J. 24:635–44

210. Donnison M, Beaton A, Davey HW, Broadhurst R, L’Huillier P, Pfeffer PL. 2005. Loss of the extraem-bryonic ectoderm in Elf5 mutants leads to defects in embryonic patterning. Development 132:2299–308

211. Lacorazza HD, Miyazaki Y, Di Cristofano A, Deblasio A, Hedvat C, et al. 2002. The ETS protein MEFplays a critical role in perforin gene expression and the development of natural killer and NK-T cells.Immunity 17:437–49

212. Garrett-Sinha LA, Dahl R, Rao S, Barton KP, Simon MC. 2001. PU.1 exhibits partial functionalredundancy with Spi-B, but not with Ets-1 or Elf-1. Blood 97:2908–12

213. Chen G, Korfhagen TR, Xu Y, Kitzmiller J, Wert SE, et al. 2009. SPDEF is required for mousepulmonary goblet cell differentiation and regulates a network of genes associated with mucus production.J. Clin. Investig. 119:2914–24

470 Hollenhorst · McIntosh · Graves

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 35: Genomic and Biochemical Insights into the Specificity of ...

BI80CH20-Graves ARI 20 May 2011 13:19

214. Gregorieff A, Stange DE, Kujala P, Begthel H, van den Born M, et al. 2009. The Ets-domain tran-scription factor SPDEF promotes maturation of goblet and paneth cells in the intestinal epithelium.Gastroenterology 137:1333–45, e1–3

215. Arber S, Ladle DR, Lin JH, Frank E, Jessell TM. 2000. ETS gene Er81 controls the formation offunctional connections between group Ia sensory afferents and motor neurons. Cell 101:485–98

216. Chen C, Ouyang W, Grigura V, Zhou Q, Carnes K, et al. 2005. ERM is required for transcriptionalcontrol of the spermatogonial stem cell niche. Nature 436:1030–34

217. Livet J, Sigrist M, Stroebel S, De Paola V, Price SR, et al. 2002. ETS gene Pea3 controls the centralposition and terminal arborization of specific motor neuron pools. Neuron 35:877–92

218. Laing MA, Coonrod S, Hinton BT, Downie JW, Tozer R, et al. 2000. Male sexual dysfunction in micebearing targeted mutant alleles of the PEA3 Ets gene. Mol. Cell. Biol. 20:9337–45

219. Ayadi A, Zheng H, Sobieszczuk P, Buchwalter G, Moerman P, et al. 2001. Net-targeted mutant micedevelop a vascular phenotype and up-regulate Egr-1. EMBO J. 20:5139–52

220. Costello PS, Nicolas RH, Watanabe Y, Rosewell I, Treisman R. 2004. Ternary complex factor SAP-1is required for Erk-mediated thymocyte positive selection. Nat. Immunol. 5:289–98

221. Cesari F, Brecht S, Vintersten K, Vuong LG, Hofmann M, et al. 2004. Mice deficient for the Etstranscription factor Elk-1 show normal immune responses and mildly impaired neuronal gene activation.Mol. Cell Biol. 24:294–305

222. Bories JC, Willerford DM, Grevin D, Davidson L, Camus A, et al. 1995. Increased T-cell apoptosis andterminal B-cell differentiation induced by inactivation of the Ets-1 proto-oncogene. Nature 377:635–38

223. Barton K, Muthusamy N, Fischer C, Ting CN, Walunas TL, et al. 1998. The Ets-1 transcription factoris required for the development of natural killer cells in mice. Immunity 9:555–63

224. Yamamoto H, Flannery ML, Kupriyanov S, Pearce J, McKercher SR, et al. 1998. Defective trophoblastfunction in mice with a targeted mutation of Ets2. Genes Dev. 12:1315–26

225. Lee D, Park C, Lee H, Lugus JJ, Kim SH, et al. 2008. ER71 acts downstream of BMP, Notch, andWnt signaling in blood and vessel progenitor specification. Cell Stem Cell 2:497–507

226. Ferdous A, Caprioli A, Iacovino M, Martin CM, Morris J, et al. 2009. Nkx2–5 transactivates the Ets-related protein 71 gene and specifies an endothelial/endocardial fate in the developing embryo. Proc.Natl. Acad. Sci. USA 106:814–19

227. Papadaki C, Alexiou M, Cecena G, Verykokakis M, Bilitou A, et al. 2007. Transcriptional repressor Erfdetermines extraembryonic ectoderm differentiation. Mol. Cell. Biol. 27:5201–13

228. Zhang XK, Moussa O, LaRue A, Bradshaw S, Molano I, et al. 2008. The transcription factor Fli-1modulates marginal zone and follicular B cell development in mice. J. Immunol. 181:1644–54

229. Hart A, Melet F, Grossfeld P, Chien K, Jones C, et al. 2000. Fli-1 is required for murine vascular andmegakaryocytic development and is hemizygously deleted in patients with thrombocytopenia. Immunity13:167–77

230. Melet F, Motro B, Rossi DJ, Zhang L, Bernstein A. 1996. Generation of a novel Fli-1 protein by genetargeting leads to a defect in thymus development and a delay in Friend virus-induced erythroleukemia.Mol. Cell. Biol. 16:2708–18

231. Spyropoulos DD, Pharr PN, Lavenburg KR, Jackers P, Papas TS, et al. 2000. Hemorrhage, impairedhematopoiesis, and lethality in mouse embryos carrying a targeted disruption of the Fli1 transcriptionfactor. Mol. Cell. Biol. 20:5643–52

232. Hendricks TJ, Fyodorov DV, Wegman LJ, Lelutiu NB, Pehek EA, et al. 2003. Pet-1 ETS gene playsa critical role in 5-HT neuron development and is required for normal anxiety-like and aggressivebehavior. Neuron 37:233–47

233. Liu C, Maejima T, Wyler SC, Casadesus G, Herlitze S, Deneris ES. 2010. Pet-1 is required acrossdifferent stages of life to regulate serotonergic function. Nat. Neurosci. 13:1190–98

234. Ristevski S, O’Leary DA, Thornell AP, Owen MJ, Kola I, Hertzog PJ. 2004. The ETS transcriptionfactor GABPalpha is essential for early embryogenesis. Mol. Cell. Biol. 24:5844–49

235. Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, et al. 2002. The human genome browserat UCSC. Genome Res. 12:996–1006

www.annualreviews.org • ETS Family of Transcription Factors 471

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 36: Genomic and Biochemical Insights into the Specificity of ...

BI80-FrontMatter ARI 16 May 2011 14:13

Annual Review ofBiochemistry

Volume 80, 2011Contents

Preface

Past, Present, and Future Triumphs of BiochemistryJoAnne Stubbe � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �v

Prefatory

From Serendipity to TherapyElizabeth F. Neufeld � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 1

Journey of a Molecular BiologistMasayasu Nomura � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �16

My Life with NatureJulius Adler � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �42

Membrane Vesicle Theme

Protein Folding and Modification in the MammalianEndoplasmic ReticulumIneke Braakman and Neil J. Bulleid � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �71

Mechanisms of Membrane Curvature SensingBruno Antonny � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 101

Biogenesis and Cargo Selectivity of AutophagosomesHilla Weidberg, Elena Shvets, and Zvulun Elazar � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 125

Membrane Protein Folding and Insertion Theme

Introduction to Theme “Membrane Protein Folding and Insertion”Gunnar von Heijne � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 157

Assembly of Bacterial Inner Membrane ProteinsRoss E. Dalbey, Peng Wang, and Andreas Kuhn � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 161

β-Barrel Membrane Protein Assembly by the Bam ComplexChristine L. Hagan, Thomas J. Silhavy, and Daniel Kahne � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 189

vii

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 37: Genomic and Biochemical Insights into the Specificity of ...

BI80-FrontMatter ARI 16 May 2011 14:13

Transmembrane Communication: General Principles and Lessonsfrom the Structure and Function of the M2 Proton Channel, K+

Channels, and Integrin ReceptorsGevorg Grigoryan, David T. Moore, and William F. DeGrado � � � � � � � � � � � � � � � � � � � � � � � � 211

Biological Mass Spectrometry Theme

Mass Spectrometry in the Postgenomic EraBrian T. Chait � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 239

Advances in the Mass Spectrometry of Membrane Proteins:From Individual Proteins to Intact ComplexesNelson P. Barrera and Carol V. Robinson � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 247

Quantitative, High-Resolution Proteomics for Data-DrivenSystems BiologyJurgen Cox and Matthias Mann � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 273

Applications of Mass Spectrometry to Lipids and MembranesRichard Harkewicz and Edward A. Dennis � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 301

Cellular Imaging Theme

Emerging In Vivo Analyses of Cell Function UsingFluorescence ImagingJennifer Lippincott-Schwartz � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 327

Biochemistry of Mobile Zinc and Nitric Oxide Revealedby Fluorescent SensorsMichael D. Pluth, Elisa Tomat, and Stephen J. Lippard � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 333

Development of Probes for Cellular Functions Using FluorescentProteins and Fluorescence Resonance Energy TransferAtsushi Miyawaki � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 357

Reporting from the Field: Genetically Encoded Fluorescent ReportersUncover Signaling Dynamics in Living Biological SystemsSohum Mehta and Jin Zhang � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 375

Recent Advances in Biochemistry

DNA Replicases from a Bacterial PerspectiveCharles S. McHenry � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 403

Genomic and Biochemical Insights into the Specificity of ETSTranscription FactorsPeter C. Hollenhorst, Lawrence P. McIntosh, and Barbara J. Graves � � � � � � � � � � � � � � � � � � � 437

viii Contents

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 38: Genomic and Biochemical Insights into the Specificity of ...

BI80-FrontMatter ARI 16 May 2011 14:13

Signals and Combinatorial Functions of Histone ModificationsTamaki Suganuma and Jerry L. Workman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 473

Assembly of Bacterial RibosomesZahra Shajani, Michael T. Sykes, and James R. Williamson � � � � � � � � � � � � � � � � � � � � � � � � � � � � 501

The Mechanism of Peptidyl Transfer Catalysis by the RibosomeEdward Ki Yun Leung, Nikolai Suslov, Nicole Tuttle, Raghuvir Sengupta,

and Joseph Anthony Piccirilli � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 527

Amyloid Structure: Conformational Diversity and ConsequencesBrandon H. Toyama and Jonathan S. Weissman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 557

AAA+ Proteases: ATP-Fueled Machines of Protein DestructionRobert T. Sauer and Tania A. Baker � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 587

The Structure of the Nuclear Pore ComplexAndre Hoelz, Erik W. Debler, and Gunter Blobel � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 613

Benchmark Reaction Rates, the Stability of Biological Moleculesin Water, and the Evolution of Catalytic Power in EnzymesRichard Wolfenden � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 645

Biological Phosphoryl-Transfer Reactions: Understanding Mechanismand CatalysisJonathan K. Lassila, Jesse G. Zalatan, and Daniel Herschlag � � � � � � � � � � � � � � � � � � � � � � � � � � � 669

Enzymatic Transition States, Transition-State Analogs, Dynamics,Thermodynamics, and LifetimesVern L. Schramm � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 703

Class I Ribonucleotide Reductases: Metallocofactor Assemblyand Repair In Vitro and In VivoJoseph A. Cotruvo Jr. and JoAnne Stubbe � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 733

The Evolution of Protein Kinase Inhibitors from Antagoniststo Agonists of Cellular SignalingArvin C. Dar and Kevan M. Shokat � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 769

Glycan Microarrays for Decoding the GlycomeCory D. Rillahan and James C. Paulson � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 797

Cross Talk Between O-GlcNAcylation and Phosphorylation:Roles in Signaling, Transcription, and Chronic DiseaseGerald W. Hart, Chad Slawson, Genaro Ramirez-Correa, and Olof Lagerlof � � � � � � � � � 825

Regulation of Phospholipid Synthesis in the YeastSaccharomyces cerevisiaeGeorge M. Carman and Gil-Soo Han � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 859

Contents ix

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.

Page 39: Genomic and Biochemical Insights into the Specificity of ...

BI80-FrontMatter ARI 16 May 2011 14:13

Sterol Regulation of Metabolism, Homeostasis, and DevelopmentJoshua Wollam and Adam Antebi � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 885

Structural Biology of the Toll-Like Receptor FamilyJin Young Kang and Jie-Oh Lee � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 917

Structure-Function Relationships of the G Domain, a CanonicalSwitch MotifAlfred Wittinghofer and Ingrid R. Vetter � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 943

STIM Proteins and the Endoplasmic Reticulum-PlasmaMembrane JunctionsSilvia Carrasco and Tobias Meyer � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 973

Amino Acid Signaling in TOR ActivationJoungmok Kim and Kun-Liang Guan � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �1001

Mitochondrial tRNA Import and Its Consequencesfor Mitochondrial TranslationAndre Schneider � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �1033

Caspase Substrates and Cellular RemodelingEmily D. Crawford and James A. Wells � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �1055

Regulation of HSF1 Function in the Heat Stress Response:Implications in Aging and DiseaseJulius Anckar and Lea Sistonen � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �1089

Indexes

Cumulative Index of Contributing Authors, Volumes 76–80 � � � � � � � � � � � � � � � � � � � � � � � � � �1117

Cumulative Index of Chapter Titles, Volumes 76–80 � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �1121

Errata

An online log of corrections to Annual Review of Biochemistry articles may be found athttp://biochem.annualreviews.org/errata.shtml

x Contents

Ann

u. R

ev. B

ioch

em. 2

011.

80:4

37-4

71. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Bri

tish

Col

umbi

a on

09/

16/1

1. F

or p

erso

nal u

se o

nly.