22
Discovering Hematopoietic Mechanisms Through Genome-Wide Analysis of GATA Factor Chromatin Occupancy Tohru Fujiwara 1,2 , Henriette O'Geen 1,4 , Sunduz Keles 1,3 , Kimberly Blahnik 4 , Amelia K. Linnemann 2 , Yoon-A Kang 2 , Kyunghee Choi 5 , Peggy J. Farnham 4 , and Emery H. Bresnick 2,* 2 Department of Pharmacology, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706 3 Department of Statistics and Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706 4 Genome Center, University of California - Davis, Davis, CA 95616 5 Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110 SUMMARY GATA factors interact with simple DNA motifs (WGATAR) to regulate critical processes, including hematopoiesis, but very few WGATAR motifs are occupied in genomes. Given the rudimentary knowledge of mechanisms underlying this restriction, and how GATA factors establish genetic networks, we used ChIP-seq to define GATA-1 and GATA-2 occupancy genome-wide in erythroid cells. Coupled with genetic complementation analysis and transcriptional profiling, these studies revealed a rich collection of targets containing a characteristic binding motif of greater complexity than WGATAR. GATA factors occupied loci encoding multiple components of the Scl/TAL1 complex, a master regulator of hematopoiesis and leukemogenic target. Mechanistic analyses provided evidence for cross-regulatory and autoregulatory interactions among components of this complex, including GATA-2 induction of the hematopoietic corepressor ETO-2 and an ETO-2 negative autoregulatory loop. These results establish fundamental principles underlying GATA factor mechanisms in chromatin and illustrate a complex network of considerable importance for the control of hematopoiesis. INTRODUCTION Master regulators of development are commonly transcription factors that instigate complex genetic networks. Mechanisms underlying the function of these regulators are highly stringent, as deviations in their expression, chromatin site selection and protein-protein interactions elicit catastrophic phenotypes. In the context of hematopoiesis, defective genetic networks cause anemias, leukemias and lymphomas. Given the essential role of GATA factors in controlling © 2009 Elsevier Inc. All rights reserved. *Corresponding author: Mailing address: University of Wisconsin School of Medicine, Department of Pharmacology, 1300 University Avenue, Madison, WI 53706. Phone: (608) 265-6446. Fax: (608) 262-1257. [email protected]. 1 These authors contributed equally Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. NIH Public Access Author Manuscript Mol Cell. Author manuscript; available in PMC 2010 November 25. Published in final edited form as: Mol Cell. 2009 November 25; 36(4): 667–681. doi:10.1016/j.molcel.2009.11.001. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Discovering Hematopoietic Mechanisms through Genome-wide Analysis of GATA Factor Chromatin Occupancy

  • Upload
    wisc

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Discovering Hematopoietic Mechanisms Through Genome-WideAnalysis of GATA Factor Chromatin Occupancy

Tohru Fujiwara1,2, Henriette O'Geen1,4, Sunduz Keles1,3, Kimberly Blahnik4, Amelia K.Linnemann2, Yoon-A Kang2, Kyunghee Choi5, Peggy J. Farnham4, and Emery H.Bresnick2,*2Department of Pharmacology, University of Wisconsin School of Medicine and Public Health, 1300University Avenue, Madison, WI 537063Department of Statistics and Biostatistics and Medical Informatics, University of Wisconsin Schoolof Medicine and Public Health, 1300 University Avenue, Madison, WI 537064Genome Center, University of California - Davis, Davis, CA 956165Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis,MO 63110

SUMMARYGATA factors interact with simple DNA motifs (WGATAR) to regulate critical processes, includinghematopoiesis, but very few WGATAR motifs are occupied in genomes. Given the rudimentaryknowledge of mechanisms underlying this restriction, and how GATA factors establish geneticnetworks, we used ChIP-seq to define GATA-1 and GATA-2 occupancy genome-wide in erythroidcells. Coupled with genetic complementation analysis and transcriptional profiling, these studiesrevealed a rich collection of targets containing a characteristic binding motif of greater complexitythan WGATAR. GATA factors occupied loci encoding multiple components of the Scl/TAL1complex, a master regulator of hematopoiesis and leukemogenic target. Mechanistic analysesprovided evidence for cross-regulatory and autoregulatory interactions among components of thiscomplex, including GATA-2 induction of the hematopoietic corepressor ETO-2 and an ETO-2negative autoregulatory loop. These results establish fundamental principles underlying GATAfactor mechanisms in chromatin and illustrate a complex network of considerable importance for thecontrol of hematopoiesis.

INTRODUCTIONMaster regulators of development are commonly transcription factors that instigate complexgenetic networks. Mechanisms underlying the function of these regulators are highly stringent,as deviations in their expression, chromatin site selection and protein-protein interactions elicitcatastrophic phenotypes. In the context of hematopoiesis, defective genetic networks causeanemias, leukemias and lymphomas. Given the essential role of GATA factors in controlling

© 2009 Elsevier Inc. All rights reserved.*Corresponding author: Mailing address: University of Wisconsin School of Medicine, Department of Pharmacology, 1300 UniversityAvenue, Madison, WI 53706. Phone: (608) 265-6446. Fax: (608) 262-1257. [email protected] authors contributed equallyPublisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customerswe are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resultingproof before it is published in its final citable form. Please note that during the production process errors may be discovered which couldaffect the content, and all legal disclaimers that apply to the journal pertain.

NIH Public AccessAuthor ManuscriptMol Cell. Author manuscript; available in PMC 2010 November 25.

Published in final edited form as:Mol Cell. 2009 November 25; 36(4): 667–681. doi:10.1016/j.molcel.2009.11.001.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

hematopoiesis (Cantor and Orkin, 2002) and mutations in human leukemias (Crispino, 2005),it is crucial to elucidate their mechanisms, with perhaps the most rudimentary goal to establishthe ensemble of target genes genome-wide.

GATA-2 expression occurs early in hematopoiesis and is required for the maintenance andexpansion of hematopoietic stem cells and/or multipotent progenitors (Tsai et al., 1994).GATA-1 expression is induced subsequent to GATA-2 and is essential for the development oferythrocytes (Pevny et al., 1991; Simon et al., 1992), megakaryocytes (Shivdasani et al.,1997), eosinophils (Yu et al., 2002) and mast cells (Migliaccio et al., 2003). Whereas GATA-1and GATA-2 bind DNA with a similar specificity (Ko and Engel, 1993; Merika and Orkin,1993), and function redundantly to promote primitive erythroblast development (Fujiwara etal., 2004), they also exert distinct functions. GATA factors activate and repress genes, with orwithout the coregulator Friend of GATA-1 (FOG-1) (Crispino et al., 1999; Johnson et al.,2007; Tsang et al., 1997). FOG-1-dependent activation involves facilitation of GATA-1chromatin occupancy (Letting et al., 2004; Pal et al., 2004a) and GATA-2 displacement fromtarget sites (Pal et al., 2004a). FOG-1-dependent repression can be accompanied by broadhistone deacetylation (Grass et al., 2003), and FOG-1 binds two corepressors, NuRD (Honget al., 2005) and CtBP (Turner and Crossley, 1998).

Analyses at several loci suggest that GATA-1 and GATA-2 occupy a small fraction of theabundant WGATAR motif (Grass et al., 2003; Grass et al., 2006; Im et al., 2005; Johnson etal., 2002; Pal et al., 2004b). Even the presence of a conserved motif appears to be insufficientfor implicating a GATA factor in regulation. Given that GATA factor DNA bindingspecificities were defined with naked DNA, nucleotides flanking WGATAR or cis-elementsnear WGATAR may mediate occupancy in vivo, or WGATAR might not be critical inchromatin.

GATA-1 and GATA-2 function cooperatively with the master regulator of hematopoiesis Scl/TAL1 on E-box (CANNTG)-WGATAR-containing composite elements (Lahlil et al., 2004;Vyas et al., 1999; Wadman et al., 1997; Wozniak et al., 2007; Xu et al., 2003). GATA-1 (Tripicet al., 2008) and GATA-2 (Wozniak et al., 2008) co-localize on chromatin sites with Scl/TAL1.Scl/TAL1 assembles a multimeric complex containing E2A, LMO2, Ldb1 and GATA-1(Gottgens et al., 2002; Lahlil et al., 2004; Lecuyer et al., 2002; Wadman et al., 1997; Xu et al.,2003). Another critical factor that binds the Scl/TAL1 complex is the corepressor ETO-2(Amann et al., 2001; Schuh et al., 2005), which like Scl/TAL1 (Aplan et al., 1992) and LMO2(Hacein-Bey-Abina et al., 2003), is disrupted in leukemia (Gamou et al., 1998). Targeteddisruption of Cbfa2t3, which encodes ETO-2, revealed an ETO-2 requirement forhematopoietic progenitor fate decisions, proliferation, and stress-dependent hematopoiesis(Chyla et al., 2008). Although other studies implicated ETO-2 in controlling erythropoiesis(Goardon et al., 2006) and megakaryopoiesis (Hamlett et al., 2008), little is known about itsfunction in GATA-2-expressing cells.

GATA-2 and Scl/TAL1 co-localize at chromatin sites containing E-box-WGATAR motifs(Wozniak et al., 2008). As only a small fraction of E-box-WGATAR motifs are occupied inchromatin, resembling that of WGATAR motifs (Wozniak et al., 2008), the composite motifdoes not appear to confer a major advantage for chromatin occupancy vs. WGATAR.

Since GATA factor DNA binding specificities have been deduced through in vitro analyses,it is critical to generate and validate datasets of GATA factor occupancy in vivo. The magnitudeand qualitative features of GATA factor target gene ensembles is unclear. We describe ChIP-seq analysis, in conjunction with expression profiling, target validation in primary cells, andcomputational mining, which yielded principles governing GATA factor-chromatininteractions and a genetic network of considerable importance for controlling hematopoiesis.

Fujiwara et al. Page 2

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

RESULTSChIP-Seq and Expression Profiling Reveal a Rich Set of GATA Factor Targets

ChIP-seq was conducted with human K562 erythroleukemia cells that express GATA-1 (Tsaiet al., 1989) and GATA-2 (Dorfman et al., 1992) and are studied intensively in the ENCODEproject (http://genome.ucsc.edu/ENCODE). The ChIP assay was validated by measuringGATA-1 occupancy at β-globin LCR HS2 (Johnson et al., 2002). Immunoprecipitated DNAfrom two biological replicates was used to prepare libraries for deep-sequencing. Sequenceswere mapped to the UCSC Human Genome assembly. Replicates A and B yielded 4.4 millionand 10.3 million uniquely mapped sequences, respectively. Using a false discovery rate of0.001, we identified 1,536 and 6,104 GATA-1 binding sites (peaks) in replicates A and Brespectively. To assess reproducibility, peak overlap was determined. 90% of the peaks (1,380)from replicate A were present in replicate B. All reads for both replicates were merged to yield5,749 sites (Suppl. Table 1) corresponding to 4,061 genes. The peak heights ranged from20-259 sequence reads, with an average height of 38 and width of 327 bp, and peak numberdid not correlate with chromosome size (Suppl. Table 2). The analysis revealed establishedand new GATA-1 targets, including hematopoietic transcription factors, signaling molecules,and red cell cytoskeletal components (Fig. 1).

K562 cells are transformed and have a primitive erythroid phenotype (Lozzio et al., 1979),whereas murine G1E cells are untransformed and resemble normal definitive proerythroblasts(Weiss et al., 1997; Welch et al., 2004). Given these differences, its target sites might differgreatly in the two cell types. Quantitative ChIP analysis was conducted with untreated and β-estradiol-treated G1E cells stably expressing ER-GATA-1 (G1E-ER-GATA-1) to test whetherER-GATA-1 and GATA-2 occupy sites containing conserved WGATAR motifs that weredetected by ChIP-seq. Although the absolute levels of occupancy were higher for peaks withhigh vs. low peak values, ER-GATA-1 and GATA-2 occupancy were detected in 10/13 (Fig.2A) and 8/13 (Fig. 2B) of the peaks, respectively.

Computational Mining of ChIP-seq and Expression Profiling DatasetsPrior work defined GATA-1 and GATA-2 occupancy at dispersed regions of several loci(Grass et al., 2003; Grass et al., 2006; Im et al., 2005; Johnson et al., 2007; Johnson et al.,2002; Martowicz et al., 2005; Munugalavadla et al., 2005; Pal et al., 2004b; Scherzer et al.,2008; Wozniak et al., 2007). At the β-globin locus, GATA-1 instigates chromatin looping(Vakoc et al., 2005), which brings dispersed complexes in close proximity to each other. SinceGATA-1 occupancy at very few loci were analyzed, it is unclear whether occupancy occursmainly at promoters or other regions. Location analysis with the 5,749 peaks, using the Cis-regulatory Element Annotation System (http://ceas.cbi.pku.edu.cn/), revealed occupancypredominantly within introns (37%) and >1 kb away from RefSeq genes (“enhancer”) (47%)(Fig. 3A). The highest frequency of peaks was at −10 to −100 kb and +10 kb to +100 kb (Suppl.Fig. 1). Only 10% of the sites reside in proximal promoters (<1 kb upstream of RefSeq 5′ start).

GATA-1 preferentially binds WGATAR-containing DNA (Evans et al., 1988). In a siteselection analysis, recombinant chicken GATA-1 preferentially bound NNAGATAANN (Koand Engel, 1993). Site selection with recombinant mouse GATA-1 (Merika and Orkin, 1993)and Mouse Erythroleukemia Cell (MEL) extracts (Wadman et al., 1997) yielded consensussequences (G/C/A)NGAT(A/G/T)G(GCT) and CGATAA, respectively.

Based on the highly restricted occupancy of WGATAR-containing sites in cells, sequencespreferred in vitro might not be an obligate requirement in chromatin. DNA binding-defectivemutants of Scl/TAL1 (Porcher et al., 1999) and the glucocorticoid receptor (Reichardt et al.,

Fujiwara et al. Page 3

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

1998) can function in vivo, presumably through binding DNA-bound activators. Thus,GATA-1 tethered to DNA-bound factors might crosslink to sites lacking WGATAR motifs.

De novo motif finding with Cosmo (Bembom et al., 2007) and Meme (Bailey and Elkan,1995) was used to ascertain the percent of targets containing specific cis-elements. Of the 5,749ChIP-seq peaks, 5,051 (88%), 155 (3%), 193 (3%), 79 (1%) contained WGATAR, WGATA,GATAR, and WGATA + GATAR motifs (Fig. 3B), respectively, which were conservedsimilarly from mouse to man. Of the 6,976,111 WGATAR motifs in the human genome, 9,741(0.14%) reside within the 5,051 peaks (0.07-0.14% occupancy). A small subset of these motifs(6.0%) reside in E-box-WGATAR motifs. De novo motif finding using peaks containing atleast one WGATAR identified a highly significant (E-value = 2.8e−3685) position weight matrixwith the consensus (C/G)(A/T)GATAA(G/A/C)(G/A/C) (Fig. 3C, left), which occurs 297,124times in the human genome. Of the 5,051 WGATAR-containing peaks, 3,165 (63%) containthis consensus. Based on 3,165 peaks containing ≥1 copy of the consensus, 0.71% wereoccupied by GATA-1, an order of magnitude higher than the 0.07% occupancy of WGATAR(p < 0.0001). As the three extended positions (2, 9, 10) exhibit compositions that deviate greatlyfrom the equal probability of bases and the [(A, T): 0.3] and [(C, G):0.2] configuration (p <1e−100), we further evaluated the significance of this composition by randomly drawing thesame number of WGATAR occurrences from the genome and constructing a position weightmatrix with WGATAR and its first left and two right flanking positions. We repeated thisprocess 1000 times, and the flanking region information contents were significantly smallerthan that of the position weight matrix constructed from WGATAR-containing peaks (p = 0based on 1000 randomization experiments).

Of the 62,412 E-box-WGATAR composite elements in the human genome, 307 reside within304 peaks (0.49% occupancy). Analysis of the 301 peaks containing one E-boxWGATARelement revealed the logo in Fig. 3C, right. Positions 16, 23, and 24 resemble the respectivepositions of the more complex WGATAR motif (Fig. 3C) and deviate from random nucleotides(p < 1e−90). The analysis also revealed unexpected information content in the NN residues ofCANNTG (Fig. 3C, right). GATA-1-occupied composite elements had a similar probabilityof having G or T in the 1st N position and C, A, or G in the 2nd N position (Fig. 3C). Site-selection analysis with recombinant Scl/TAL1-E2A heterodimers identified the consensusAACAGATGGT, and 86-100% of bound sequences had GA in the NN positions (Hsu et al.,1994). The in vitro preference for AA and GT at the 5′ and 3' ends deviated from occupiedchromatin sites, which had no sequence preference at the 5′ end and either C, G, or T followingTG. Site-selection studies with MEL cell extracts identified E-box-GATA elements with theconsensus CAGGTG(N)9GATA (Wadman et al., 1997). The GG sequence in the NN positionsdiffered from GATA-1-occupied E-box-WGATAR elements in chromatin.

De novo motif finding on the 698 peaks lacking WGATAR identified GGAATGGAATG asoverrepresented in this group. This sequence appears 3 and 64 times in WGATAR-containingand -lacking peaks, respectively (p < 2.2e−16). GGAATGGAATG resides in microsatelliterepeats (Gangwal et al., 2008) and contains the Ets binding motif GGAA (Sharrocks, 2001).The oncogenic Ewings Sarcoma protein fusion to FLI functions through this sequence(Gangwal et al., 2008). GAATGGAATGGAAT-containing GATA-1 occupancy sites definedby ChIP-seq lack WGATAR motifs. As GATA factors physically associate with Ets factors(Rekhtman et al., 1999), a DNA-bound Ets factor might tether GATA-1 to chromatin at thisclass of sites.

To assess whether the ChIP-seq peaks pinpoint GATA-1-regulated genes, gene expression wasprofiled in untreated and β-estradiol-treated G1E-ER-GATA-1 cells (Suppl. Table 3). ER-GATA-1 induced and repressed 1,166 and 1,010 genes, respectively, >1.5 fold. Merging ChIP-seq and profiling datasets revealed 142 and 154 activated and repressed genes, respectively,

Fujiwara et al. Page 4

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

which were GATA-1-occupied (the top 60 are shown in Fig. 3D). These genes include knownGATA-1 targets, such as Slc4a1 and Epb4.9 (Kim et al., 2007) encoding red cell cytoskeletalproteins (Mohandas and Gallagher, 2008), yet many had not been implicated in GATA-1function or hematopoiesis. 44% of the genes (Fig. 3D) were not described in a prior profilinganalysis in this system (Welch et al., 2004). In primary Ter119+ bone marrow erythroblasts,GATA-1 occupied 32/36 of the sites significantly higher than the negative control Ey promoterand 14/36 higher than the positive control β-globin HS2, respectively (p < 0.05) (Fig. 3E).Using tiled microarrays containing sequences from 120 genes of Fig. 3D (with 120,000 bp ofupstream and downstream sequence), ChIP-chip analysis in primary Ter119+ bone marrowerythroblasts revealed significant GATA-1 occupancy at 90% of the targets (98% and 82% ofactivated and repressed targets, respectively) (Fig. 3F, Suppl. Table 4).

Our previous comparison of GATA-1 and GATA-2 occupancy at several loci in G1E and G1E-ER-GATA-1 cells revealed that almost all of the GATA-1-occupied sites were occupied byGATA-2 in uninduced G1E-ER-GATA-1 cells and G1E cells. To assess the extent of overlapin a cell expressing endogenous GATA-1 and GATA-2, we analyzed GATA-2 occupancy byChIP-seq in K562 cells. Analysis of two biological replicates (21,167 peaks) (Suppl. Table 5)revealed major overlap (Fig. 4A, B). Since the GATA-1 and GATA-2 samples analyzed byChIP-seq were isolated on different days, we conducted quantitative ChIP analysis for GATA-1and GATA-2 at the same time. Sampling representative GATA-1- and GATA-2-unique peaksidentified GATA-1- and GATA-2-selective targets (Fig. 4C, D). The extensive sharing of sitesby GATA-1 and GATA-2 provides insights into the finding that GATA-1 and GATA-2function redundantly to generate primitive erythroblasts (Fujiwara et al., 2004). Our analysisalso revealed GATA factor-selective targets, including: a kinase critical for controllinghematopoiesis (AK2) (Lagresle-Peyrou et al., 2009); a cell type-specific component of theMediator complex (MED10) that regulates Wnt and Nodal signaling (Lin et al., 2007b); a Hoxgene (HOXB9) induced by Wnt signaling (Nguyen et al., 2009); a Forkhead transcription factor(FOXK2); a factor (BST2) that suppresses HIV-1 release from the cell surface (Goffinet et al.,2009) and is downregulated by Kaposi's sarcoma herpesvirus (Mansouri et al., 2009); a Setdomain-containing histone H3K4 methyltransferase (SMYD3) (Nguyen et al., 2009); and aputative RNA binding protein (RBM15) that regulates Notch signaling (Ma et al., 2007),controls hematopoiesis (Raffel et al., 2007) and is implicated in acute megakaryoblasticleukemia (Ma et al., 2001). As GATA-1 mutations are linked to acute megakaryoblasticleukemia (Wechsler et al., 2002), and GATA-1 represses GATA2 transcription (Grass et al.,2003), our discovery of RBM15 as a GATA-2-selective target is intriguing.

Linking GATA-2 and Scl/TAL1 to Regulation of the Hematopoietic Corepressor ETO-2, aComponent of GATA Factor and Scl/TAL1 Complexes

ChIP-seq identified five GATA factor-bound sites at CBFA2T3 (Fig. 4B), which encodesETO-2, a co-repressor that controls hematopoiesis. We tested whether GATA factors occupyall or a subset of conserved WGATAR motifs at and near Cbfa2t3. Conserved WGATARmotifs reside at −37.3, −35.7, −21.7, −21.6, −21.6, −21.5, −13.3, −1.9, −0.2, +13.3 and +15.1kb relative to the start site (Fig. 5A). Conservation of the immediate region was high (>75%)at −35.7, −21.7, −21.6, −21.6, −21.5, −13.3, −1.9 and −0.2 kb, intermediate (>50%) at +13.3kb, and low (<50%) at −37.3 and +15.1 kb (Fig. 5A). Among five ChIP-seq peaks at humanCBFA2T3 (Fig. 4B), −27.3, −15.6, −2.1, and −0.1 kb peaks corresponded to −21.6, −13.3, −1.9kb, and the promoter of murine Cbfa2t3 (Fig. 5A). GATA-2 occupied −37.3, −21.6, −13.3,−1.9, −0.2, and +13.3 kb sites (Suppl. Fig. 2A).

Scl/TAL1 forms a complex with E2A, LMO2, and Ldb1 (Lahlil et al, 2004; Wadman et al.,1997), which co-localizes with GATA-2 (Wozniak et al., 2008) and GATA-1 (Anguita et al.,2004; Lahlil et al., 2004; Tripic et al., 2008; Xu et al., 2003) in chromatin. We tested whether

Fujiwara et al. Page 5

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

this complex resides at GATA-2-occupied regions of Cbfa2t3. Scl/TAL1 occupied all exceptthe −37.3, −35.7, and +15.1 kb sites (Suppl. Fig. 2B). The Scl/TAL1-interacting factor ETO-2(Schuh et al., 2005) occupied only the Scl/TAL1-bound sites (Suppl. Fig. 2C). The occupancyof each factor correlated with the others (Suppl. Fig. 2D).

Regulatory Interactions Among Components of Complexes Containing Master Regulatorsof Hematopoiesis and Instigators of Leukemogenesis

To test whether ETO-2 occupancy at Cbfa2t3 (Fig. 5A, Suppl. Fig. 2C) reflects negative orpositive autoregulation, ETO-2 was knocked-down in G1E cells with Cbfa2t3 siRNA.Cbfa2t3 primary transcripts were quantitated as a metric of transcription. Western blotting andreal-time RT-PCR revealed a knockdown of ETO-2 protein and Cbfa2t3 mRNA, respectively(Fig. 5B), and ETO-2 occupancy at −21.6 kb was reduced by 74% (p = 0.002) (Fig. 5C). Theknockdown increased Cbfa2t3 primary transcripts (58%, p = 0.01; 71%; p = 0.00009) (Fig.5D), suggesting that ETO-2 represses Cbfa2t3. To further evaluate negative autoregulation,we measured Pol II and Ser 5-phosphorylated Pol II (P-Ser5-Pol II) at the Cbfa2t3 promoter.The knockdown increased Pol II and P-Ser5-Pol II occupancy at two sites (~2.5 and ~2 fold,respectively, p < 0.05), without affecting occupancy at the RPII215 promoter (Fig. 5E). AsETO-2 interacts with class I HDACs (Amann et al., 2001), we asked whether knocking-downETO-2 affects histone acetylation at Cbfa2t3. Acetylated histone H3 increased at −21.6 kb (p= 0.015) and the promoter (p = 0.016), but not at the RPII215 promoter (Fig. 5F). These resultsestablish an ETO-2 negative autoregulatory loop.

ETO-2-mediated negative autoregulation might reflect a non-redundant repressor function atall of its target genes. We asked whether ETO-2 occupied and regulated other GATA factortargets (Fig. 6A), including Scl/TAL1 (Lugus et al., 2007) and Lmo2 (Landry et al., 2009).ETO-2 occupied these sites with a level comparable to that at Cbfa2t3 in G1E cells (Fig. 6B,Suppl. Fig. 2C). ETO-2 knockdown induced expression of certain GATA factor targets (Icam4,Epb4.9, Slc4a1, μmajor, Eraf, and Alas2), while others were unaffected (Fig. 6C). Similarly,ETO-2 knockdown facilitated ER-GATA-1-mediated activation of certain, but not all, targets(Fig. 6D). The knockdown might not reduce ETO-2 below a threshold at which occupancy atall targets would be impaired. If ETO-2 interacts with targets in different chromatinenvironments with distinct affinities, this could explain the differential sensitivities. Thus,knocking-down ETO-2 would reduce its concentration sufficiently to impair occupancy andregulation at sites with the lowest apparent affinities. However, the knockdown significantlydecreased ETO-2 occupancy at sensitive and resistant targets (Fig. 6B), inconsistent with thispossibility.

Since Slc4a1, which is strongly induced by the ETO-2 knockdown, is bound by GATA-2 inthe repressed state, ETO-2 loss might suffice for induction, or might be inextricably coupledto GATA-2 loss (Fig. 6E). The ETO-2 knockdown induced ETO-2, but not GATA-2, loss fromthe Slc4a1 promoter (Fig. 6F), indicating that transcriptional activation solely requires ETO-2eviction.

Despite the lack of GATA-1 in G1E cells, knocking-down ETO-2 induced certain GATA-1targets. GATA-1-mediated activation might therefore require ETO-2 displacement. However,GATA-1 activation of Slc4a1 in G1E-ER-GATA-1 cells is associated with increased ETO-2occupancy at its promoter (data not shown). Thus, GATA-1-associated coactivators dominateover ETO-2 corepressor activity, negating the need to evict ETO-2, or ETO-2 has dualcorepressor/coactivator activities.

GATA-1, GATA-2, and Scl/TAL1 occupancy at Cbfa2t3 suggested that these factors regulateCbfa2t3 transcription. Interactions among components of GATA factor – Scl/TAL1 complexesinclude GATA-1 repression of Gata2 (Grass et al., 2003), Scl/TAL1 induction of Gata2 (Lugus

Fujiwara et al. Page 6

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

et al., 2007), and GATA-2 induction of Scl/TAL1 (Chan et al., 2007; Lugus et al., 2007;Wozniak et al., 2008). We used knockout and conditional overexpression approaches in EScells to examine all possible regulatory influences of GATA-1, GATA-2 and Scl/TAL1 onGata1, Gata2, Scl/TAL1, Lmo2, Ldb1 and Cbfa2t3 expression. Gene expression was analyzedupon conditional expression of GATA-2 and Scl/TAL1 during ES cell differentiation (Luguset al., 2007) and in Gata2−/− and Scl/TAL1−/− vs. wild-type EBs. Dox-mediated induction ofGATA-2 induced Gata1, Cbfa2t3, and Lmo2 (383, 6.2, and 5.1-fold; p = 0.003, 0.03, and 0.002,respectively) (Fig. 7A). Previously, we demonstrated that GATA-2 increases Scl/TAL1expression in this system 12-fold (Wozniak et al., 2008). Gata1, Cbfa2t3, Scl/TAL1, andLmo2 were weakly to modestly downregulated in Gata2-null EBs (p = 0.0055, 0.09, 0.20 and0.58, respectively) (Fig. 7A). As GATA-3 can compensate for GATA-2 (Kobayashi-Osaki etal., 2005), this might limit the downregulation. In Gata2-null cells, reduced Cbfa2t3 expressionwould abrogate the negative autoregulatory loop. The altered Cbfa2t3 expression in GATA-2-expressed and Gata2-null ES cells, as well as in G1E-ERGATA-1 cells, establishes Cbfa2t3as a GATA factor target. Dox-mediated induction of Scl/TAL1 increased Gata2 (p < 0.001)and Lmo2 expression (p < 0.0001), and their expression, as well as that of Gata1, wasdownregulated in Scl/TAL1-null EBs (p = 0.0044, 0.0089, and 0.04 respectively). Cbfa2t3 wasrepressed in Scl-null EBs (p = 0.025), and Scl/TAL1 induction in iSCL EBs repressed itsexpression (-2.2 fold, p < 0.0001) (Fig. 7A).

DISCUSSIONGATA Factor Chromatin Occupancy Rules

The genome-wide analysis of GATA-1 chromatin occupancy identified a consensus elementas a hallmark of the repertoire of GATA-1 chromatin occupancy sites, which changes theparadigm of how GATA-1 selects sequences at target genes. Contrasting with naked DNAbinding in which WGATAR is believed to be sufficient, specific nucleotides at the 5′ and 3′flanks of WGATAR were preferred, and A dominated in the R position, both with WGATARalone and E-box-WGATAR elements, yielding the chromatin occupancy consensus (C/G)(A/T)GATAA(G/A/C)(G/A/C) (Fig. 3C). Within GATA-1-occupied E-box-WGATAR elements,the NN residues of the CANNTG E-box consensus exhibited significant sequence preferences.As FOG-1 facilitates GATA-1 chromatin occupancy (Letting et al., 2004;Pal et al., 2004a),and FoxA1 stabilizes GATA-4 chromatin complexes (Sekiya et al., 2009), protein-proteininteractions are also important determinants. An obvious candidate for controlling GATAfactor occupancy at E-box-WGATAR composite elements is the E-box, although GATA-1occupancy was only slightly greater at composite elements vs. WGATAR motifs.

The localization of 90% of the occupied sites away from promoters (Fig. 3A) indicates that acanonical mode of GATA factor function involves long-range control. GATA-1 induceslooping at the β-globin locus (Kim et al., 2007;Kim et al., 2009;Vakoc et al., 2005) and altersa pre-existing loop at c-kit (Jing et al., 2008), but whether looping is common or infrequent forGATA factors was unknown. Estrogen receptor-α induces looping (Carroll et al., 2005), andgenome-wide analyses of estrogen receptor-α chromatin occupancy (Lin et al., 2007a) revealedabundant non-promoter sites. By contrast, >80% of E2F1 targets are promoters (Bieda et al.,2006). While GATA-1 (Blobel et al., 1998) and E2F1 (Fry et al., 1999;Trouche and Kouzarides,1996) utilize CBP/p300 to regulate transcription, GATA-1 uniquely utilizes FOG-1 (Crispinoet al., 1999) and MED1 (Stumpf et al., 2006). The primary sequence determinants andtopographic constraints constitute chromatin occupancy rules that govern how GATA-1establishes genetic networks that control critical processes.

Analysis of histone modification patterns in K562 cells generated by the ENCODE projectwith the identical line of K562 cells used in our ChIP-seq analysis (UCSC Genome Browser)revealed GATA-1 occupancy in introns that was often mutually exclusive with histone H3K36

Fujiwara et al. Page 7

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

trimethylation (Suppl. Fig. 3). Trimethylated H3K36 marks open reading frames, facilitatesHDAC recruitment, and prevents aberrant transcription initiation (Carrozza et al., 2005; Li etal., 2007). Acetylated H3K9, dimethylated H3K4, and monomethylated K3K4 were commonlyenriched at GATA-1 occupancy sites (Suppl. Fig. 3). This result suggests that H3K36trimethylation is incompatible with GATA-1 function at intronic sites. Since H3K36trimethylation mediates HDAC recruitment (Li et al., 2007), H3K36 trimethylation-dependentdeacetylation at GATA-1-bound introns might oppose GATA-1-induced looping and/orintergenic transcription (Kim et al., 2009). Alternatively, H3K36 trimethylation or theassociated HDAC recruitment might disfavor the assembly of functional GATA factorcomplexes within introns. As mutually exclusive factor occupancy and H3K36 trimethylationhad not been described, it will be important to assess whether this is unique to GATA factorsor can be applied in a broader context.

Biological Insights Derived from the Genomic ScreenMining the consolidated ChIP-seq and transcriptional profiling dataset revealed a rich set ofGATA factor-shared and -selective targets, which were validated with high fidelity in primaryerythroblasts. Many of these targets had not been implicated in GATA factor function orhematopoiesis, while others, such as RBM15, had been implicated in hematopoiesis andleukemogenesis, but were not known to function in GATA factor pathways. Our GATA-1 andGATA-2 ChIP-seq data provides a important resource for elucidating hematopoietic regulatorymechanisms.

Using rigorous computational and mechanistic analyses, our results establish a conceptualframework for understanding how the actions of GATA factors, Scl/TAL1, and ETO-2 areintegrated to establish a genetic network that controls hematopoiesis and instigatesleukemogenesis. GATA-1 directly represses Gata2 by displacing GATA-2 from this locus(Grass et al., 2003; Grass et al., 2006; Martowicz et al., 2005). Given the short t1/2 of GATA-2(Lurie et al., 2008), GATA switches rapidly yield cells solely expressing GATA-1. By contrastto GATA-2 and GATA-1, which are expressed early and late in hematopoiesis, respectively,Scl/TAL1 is expressed in both stages (Begley et al., 1989; Lecuyer et al., 2002; Porcher et al.,1996; Schuh et al., 2005; Shivdasani et al., 1995). Though the mechanism by which LDB1 andLmo2 mediate Scl/TAL1 function is unclear, ETO-2 is an attractive candidate for differentiallycontrolling Scl/TAL1 activity during hematopoiesis. Reduced ETO-2 expression duringerythropoiesis (Goardon et al., 2006) can be explained by our discovery that GATA-2 directlyinduces ETO-2, which is followed by ETO-2 negative autoregulation and GATA switch-mediated ETO-2 repression (Fig. 7B,C).

Our loss-of-function and gain-of-function studies define fundamental insights into how celltype-specific trans-acting factors function combinatorially to control a complex developmentalprocess. The specific interactions include: GATA-2 induction of interacting trans-actingfactors (Scl/TAL1, GATA-1, ETO-2) and an interacting co-repressor (LMO2); GATA-1repression of GATA-2; GATA-1 repression of ETO-2, and ETO-2 negative autoregulation(Fig. 7B,C). Since GATA-2 and Scl/TAL1 co-localize (Wozniak et al., 2008) on chromatin,and ETO-2 antagonizes Scl/TAL1 (Goardon et al., 2006; Schuh et al., 2005), ETO-2 indirectlyinhibits GATA-2. Accordingly, ETO-2 counteracts GATA-2-mediated induction of GATA-1,and given that GATA-1 represses ETO-2 expression, the balance between these positive andnegative interactions must be strictly managed and dynamically regulated to ensure highfidelity of hematopoiesis. The elaborate integration of the activities of GATA factors, Scl/TAL1, and ETO-2, ensures the efficient and rapid transition from a GATA-2- to a GATA-1-driven genetic network. The differential co-repressor function of ETO-2 at endogenous targets,including key regulators of erythropoiesis and erythroid cell function, suggests that minimizingETO-2 level/activity during later stages of hematopoiesis enables GATA-1 and Scl/TAL1 to

Fujiwara et al. Page 8

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

efficiently establish the genetic network of the developing red blood cell. Further mining ofour highly validated resource is expected to reveal additional fundamental insights intohematopoiesis and a broader spectrum of important biological processes.

EXPERIMENTAL PROCEDURESCell Culture

GATA-1-null G1E (Weiss et al., 1997) and G1E-ER-GATA-1 cells (Gregory et al., 1999;Johnson et al., 2002) were maintained as described (Gregory et al., 1999; Johnson et al.,2002) and in Supplementary Experimental Procedures. ES cells were cultured anddifferentiated as described (Park et al., 2004) and in Supplemental Experimental Procedures.

AntibodiesAntibodies are described in Supplementary Experimental Procedures.

Quantitative ChIP AssayQuantitative chromatin immunoprecipitation (ChIP) analysis was conducted and validated asdescribed (Im et al., 2004).

ChIP-Seq Cell and Data ProcessingChIP-Seq analysis was conducted as described in Supplementary Experimental Procedures.

siRNA-mediated KnockdownThe knockdown was conducted as described in Supplementary Experimental Procedures.

Quantitative Analysis of RNA and ProteinQuantitative RT-PCR analysis was conducted as described in Supplementary ExperimentalProcedures

Isolation of Primary Murine Bone Marrow Erythroid PrecursorsMurine bone marrow erythroblasts were separated by magnetic cell sorting system (MiltenyiBiotec) using anti-Ter119 microbeads (Miltenyi Biotec).

ChIP-chip AnalysisA tiled microarray was constructed by Nimblegen, which contains sequences from 120GATA-1 target genes (Fig. 3D), including 120,000 bp of upstream and downstream sequence,and ChIP-chip was conducted as described in Supplementary Experimental Procedures.

Supplementary MaterialRefer to Web version on PubMed Central for supplementary material.

AcknowledgmentsThis work was funded by NIH grants DK50107 (EHB), DK68634 (EHB), HG003747 (SK), HL55337 (KC) and1U54HG004558 (PJF). We thank Stuart Orkin for providing Gata2−/− and Scl−/− ES cells.

REFERENCESAmann JM, Nip J, Strom DK, Lutterbach B, Harada H, Lenny N, Downing JR, Meyers S, Hiebert SW.

ETO, a target of t(8:21) in acute leukemia, makes distinct contacts with multiple histone deacetylases

Fujiwara et al. Page 9

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

and binds Sin3A through its oligomerization domain. Mol Cell Biol 2001;21:6470–6483. [PubMed:11533236]

Anguita E, Hughes J, Heyworth C, Blobel GA, Wood WG, Higgs DR. Globin gene activation duringhematopoiesis is driven by protein complexes nucleated by GATA-1 and GATA-2. EMBO J2004;23:2841–2852. [PubMed: 15215894]

Aplan PD, Lombardi DP, Reaman GH, Sather HN, Hammond GD, Kirsch IR. Involvement of the putativehematopoietic transcription factor SCL in T- cell acute lymphoblastic leukemia. Blood 1992;79:1327–1333. [PubMed: 1311214]

Bailey TL, Elkan C. Unsupervised learning of multiple motifs in biopolymers using EM. MachineLearning 1995;21:51–80.

Begley CG, Aplan PD, Denning SM, Haynes BF, Waldmann TA, Kirsch IR. The gene SCL is expressedduring early hematopoiesis and encodes a differentiation-related DNA-binding motif. Proc Natl AcadSci U S A 1989;86:10128–10132. [PubMed: 2602361]

Bembom O, Keles S, van der Laan MJ. Supervised detection of conserved motifs in DNA sequences withcosmo. Statistical Applications in Genetics and Molecular Biology 2007;6Article 8

Bieda M, Xu X, Singer MA, Green R, Farnham PJ. Unbiased location analysis of E2F1-binding sitessuggests a widespread role for E2F1 in the human genome. Genome Res 2006;16:595–605. [PubMed:16606705]

Blobel GA, Nakajima T, Eckner R, Montminy M, Orkin SH. CREB-binding protein cooperates withtranscription factor GATA-1 and is required for erythroid differentiation. Proc Natl Acad Sci USA1998;95:2061–2066. [PubMed: 9482838]

Cantor AB, Orkin SH. Transcriptional regulation of erythropoiesis: an affair involving multiple partners.Oncogene 2002;21:3368–3376. [PubMed: 12032775]

Carroll JS, Liu XS, Brodsky AS, Li W, Meyer CA, Szary AJ, Eeckhoute J, Shao W, Hestermann EV,Geistlinger TR, et al. Chromosome mapping of estrogen receptor binding reveals long-rangeregulation requiring the forkhead protein FoxA1. Cell 2005;122:33–43. [PubMed: 16009131]

Carrozza MJ, Li B, Fiorens L, Suganuma T, Swanson SK, Lee KK, Shia WJ, Anderson S, Yates J,Washburn MP, Workman JL. Histone H3 methylation by Set2 directs deacetylation of coding regionsby Rpd3S to suppress spurious intragenic transcription. Cell 2005;123:581–592. [PubMed:16286007]

Chan WY, Follows GA, Lacaud G, Pimanda JE, Landry JR, Kinston S, Knezevic K, Piltz S, DonaldsonIJ, Gambardella L, et al. The paralogous hematopoietic regulators Lyl1 and SCL are co-regulated byEts and GATA factors yet Lyl1 cannot rescue the early SCL−/− phenotype. Blood 2007;109:1908–1916. [PubMed: 17053063]

Chyla BJ, Moreno-Miralles I, Steapleton MA, Thompson MA, Bhaskara S, Engel M, Hiebert SW.Deletion of Mtg16, a target of t(16:21), alters hematopoietic progenitor cell proliferation and lineageallocation. Mol Cell Biol 2008;28:6234–6247. [PubMed: 18710942]

Crispino JD. GATA-1 in normal and malignant hematopoiesis. Semin Cell Dev Biol 2005;16:137–147.[PubMed: 15659348]

Crispino JD, Lodish MB, MacKay JP, Orkin SH. Use of altered specificity mutants to probe a specificprotein-protein interaction in differentiation: the GATA-1:FOG complex. Mol Cell 1999;3:219–228.[PubMed: 10078204]

Dorfman DM, Wilson DB, Bruns GA, Orkin SH. Human transcription factor GATA-2. Evidence forregulation of preproendothelin-1 gene expression in endothelial cells. J Biol Chem 1992;267:1279–1285. [PubMed: 1370462]

Evans T, Reitman M, Felsenfeld G. An erythrocyte-specific DNA-binding factor recognizes a regulatorysequence common to all chicken globin genes. Proc Natl Acad Sci U S A 1988;85:5976–5980.[PubMed: 3413070]

Fry CJ, Pearson A, Malinowski E, Bartley SM, Greenblatt J, Farnham PJ. Activation of the murinedihydrofolate reductase promoter by E2F1. A requirement for CBP recruitment. J Biol Chem1999;274:15883–15891. [PubMed: 10336493]

Fujiwara Y, Chang AN, Williams AM, Orkin SH. Functional overlap of GATA-1 and GATA-2 inprimitive hematopoietic development. Blood 2004;103:583–585. [PubMed: 14504093]

Fujiwara et al. Page 10

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Gamou T, Kitamura E, Hosoda F, Shimizu K, Shinsohara K, Hayashi Y, Nagese T, Yokoyama Y, OhkiM. The partner gene of AML1 in t(16:21) myeloid malignancies is a novel member of the MTG8(ETO) family. Blood 1998;91:4028–4037. [PubMed: 9596646]

Gangwal K, Sankar S, Hollenhorst PC, Kinsey M, Haroldsen SC, Shah AA, Boucher KM, Watkins WS,Jorde LB, Graves BJ, Lessnick SL. Microsatellites as EWS/FLI response elements in Ewing'ssarcoma. Proc Natl Acad Sci U S A 2008;105:10149–10154. [PubMed: 18626011]

Goardon N, Lambert JA, Rodriguez P, Nissaire P, Herblot S, Thibault P, Dumenil D, Strouboulis J,Romeo PH, Hoang T. ETO2 coordinates cellular proliferation and differentiation duringerythropoiesis. EMBO J 2006;25:357–366. [PubMed: 16407974]

Goffinet C, Allespach I, Homann S, Tervo HM, Habermann A, Rupp D, Oberbremer L, Kern C, TibroniN, Welsch S, et al. HIV-1 antagonism of CD317 is species specific and involves Vpu-mediatedproteasomal degradation of the restriction factor. Cell Host Microbe 2009;5:285–297. [PubMed:19286137]

Gottgens B, Nastos A, Kinston S, Piltz S, Delabesse EC, Stanley M, Sanchez MJ, Ciau-Uitz A, PatientRK, Green AR. Establishing the transcriptional programme for blood: the SCL stem cell enhanceris regulated by a multiprotein complex containing Ets and GATA factors. EMBO J 2002;21:3039–3050. [PubMed: 12065417]

Grass JA, Boyer ME, Pal S, Wu J, Weiss MJ, Bresnick EH. GATA-1-dependent transcriptional repressionof GATA-2 via disruption of positive autoregulation and domain-wide chromatin remodeling. ProcNatl Acad Sci U S A 2003;100:8811–8816. [PubMed: 12857954]

Grass JA, Jing H, Kim S-I, Martowicz ML, Pal S, Blobel GA, Bresnick EH. Distinct functions of dispersedGATA factor complexes at an endogenous gene locus. Mol Cell Biol 2006;26:7056–7067. [PubMed:16980610]

Gregory T, Yu C, Ma A, Orkin SH, Blobel GA, Weiss MJ. GATA-1 and erythropoietin cooperate topromoter erythroid cell survival by regulating bcl-xl expression. Blood 1999;94:87–96. [PubMed:10381501]

Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, Lim A,Osborne CS, Pawliuk R, Morillon E, et al. LMO2-associated clonal T cell proliferation in two patientsafter gene therapy for SCID-X1. Science 2003;302:415–419. [PubMed: 14564000]

Hamlett I, Draper J, Strouboulis J, Iborra F, Porcher C, Vyas P. Characterization of megakaryocyteGATA1-interacting proteins: the corepressor ETO2 and GATA1 interact to regulate terminalmegakaryocyte maturation. Blood 2008;112:2738–2749. [PubMed: 18625887]

Hong W, Nakazawa M, Chen YY, Kori R, Vakoc CR, Rakowski C, Blobel GA. FOG-1 recruits the NuRDrepressor complex to mediate transcriptional repression by GATA-1. EMBO J 2005;24:67–78.

Hsu HL, Huang L, Tsan JT, Funk W, Wright WE, Hu JS, Kingston RE, Baer R. Preferred sequences forDNA recognition by the TAL1 helix-loop-helix proteins. Mol Cell Biol 1994;14:1256–1265.[PubMed: 8289805]

Im H, Grass JA, Johnson KD, Boyer ME, Wu J, Bresnick EH. Measurement of protein-DNA interactionsin vivo by chromatin immunoprecipitation. Methods Mol Biol 2004;284:129–146. [PubMed:15173613]

Im H, Grass JA, Johnson KD, Kim S-I, Boyer ME, Imbalzano AN, Bieker JJ, Bresnick EH. Chromatindomain activation via GATA-1 utilization of a small subset of dispersed GATA motifs within a broadchromosomal region. Proc Natl Acad Sci U S A 2005;102:17065–17070. [PubMed: 16286657]

Jing H, Vakoc CR, Ying L, Mandat S, Wang H, Zheng X, Blobel GA. Exchange of GATA factors mediatestransitions in looped chromatin organization at a developmentally regulated gene locus. Mol Cell2008;29:232–242. [PubMed: 18243117]

Johnson KD, Boyer ME, Kim S-I, Kang SY, Wickrema A, Cantor AB, Bresnick EH. Friend of GATA-1-independent transcriptional repression: a novel mode of GATA-1 function. Blood 2007;109:5230–5233. [PubMed: 17339418]

Johnson KD, Grass JD, Boyer ME, Kiekhaefer CM, Blobel GA, Weiss MJ, Bresnick EH. Cooperativeactivities of hematopoietic regulators recruit RNA polymerase II to a tissue-specific chromatindomain. Proc Natl Acad Sci USA 2002;99:11760–11765. [PubMed: 12193659]

Fujiwara et al. Page 11

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Kim S-I, Bultman SJ, Jing H, Blobel GA, Bresnick EB. Dissecting molecular steps in chromatin domainactivation during hematopoietic differentiation. Mol Cell Biol 2007;27:4551–4565. [PubMed:17438135]

Kim S-I, Bultman SJ, Kiefer CM, Dean A, Bresnick EH. BRG1 requirement for long-range interactionof a locus control region with a downstream promoter. Proc Natl Acad Sci U S A 2009;106:2259–2264. [PubMed: 19171905]

Ko LJ, Engel JD. DNA-binding specificities of the GATA transcription factor family. Mol Cell Biol1993;13:4011–4022. [PubMed: 8321208]

Kobayashi-Osaki M, Ohneda O, Suzuki N, Minegishi N, Yokomizo T, Takahashi S, Lim KC, Engel JD,Yamamoto M. GATA motifs regulate early hematopoietic lineage-specific expression of the Gata2gene. Mol Cell Biol 2005;25:7005–7020. [PubMed: 16055713]

Lagresle-Peyrou C, Six EM, Picard C, Rieux-Laucat F, Michel V, Ditadi A, Demerens-de ChappedelaineC, Morillon E, Valensi F, Simon-Stoos KL, et al. Human adenylate kinase deficiency causes aprofound hematopoietic defect associated with sensorineural deafness. Nat Genet 2009;41:106–111.[PubMed: 19043416]

Lahlil R, Lecuyer E, Herblot S, Hoang T. SCL assembles a multifactorial complex that determinesglycophorin A expression. Mol Cell Biol 2004;24:1439–1452. [PubMed: 14749362]

Landry JR, Bonadies N, Kinston S, Knezevic K, Wilson NK, Oram SH, Janes M, Piltz S, Hammett M,Carter J, et al. Expression of the leukaemia oncogene Lmo2 is controlled by an array of tissue specificelements dispersed over 100 kb and bound by Tal1/Lmo2, Ets, and GATA factors. Blood2009;113:5783–5792. [PubMed: 19171877]

Lecuyer E, Herblot S, Saint-Denis M, Martin R, Begley CG, Porcher C, Orkin SH, Hoang T. The SCLcomplex regulates c-kit expression in hematopoietic cells through functional interaction with Sp1.Blood 2002;100:2430–2440. [PubMed: 12239153]

Letting DL, Chen YY, Rakowski C, Reedy S, Blobel GA. Context-dependent regulation of GATA-1 byfriend of GATA-1. Proc Natl Acad Sci U S A 2004;101:476–481. [PubMed: 14695898]

Li B, Gogol M, Carey M, Lee D, Seidel C, Workman JL. Combined action of PHD and chromo domainsdirects the Rpd3S HDAC to transcribed chromatin. Science 2007;316:1050–1054. [PubMed:17510366]

Lin C-Y, Vega VB, Thomsen JS, Zhang T, Kong SL, Xie M, Chiu KP, Lipovich L, Barnett DH, StossiF, et al. While-genome cartography of estrogen receptor alpha binding sites. PLoS Genet 2007a;3:867–885.

Lin X, Rinaldo L, Fazly AF, Xu X. Depletion of Med10 enhances Wnt and suppresses Nodal signalingduring zebrafish embryogenesis. Dev Biol 2007b;303:536–548. [PubMed: 17208216]

Lozzio CB, Lozzio BB, Machado EA, Fuhr JE, Lair SV, Bamberger EG. Effects of sodium butyrate onhuman chronic myelogenous leukemia cell line K562. Nature 1979;281:709–710. [PubMed: 298333]

Lugus JJ, Chung YS, Mills JC, Kim SI, Grass JA, Kyba M, Doherty JM, Bresnick EH, Choi K. GATA2functions at multiple steps in hemangioblast development and differentiation. Development2007;134:393–405. [PubMed: 17166922]

Lurie LJ, Boyer ME, Grass JA, Bresnick EH. Differential GATA factor stabilities: implications forchromatin occupancy by structurally similar transcription factors. Biochem 2008;47:859–869.[PubMed: 18154321]

Ma X, Renda MJ, Wang L, Cheng EC, Niu C, Morris SW, Chi AS, Krause DS. Rbm15 modulates Notch-induced transcriptional activation and affects myeloid differentiation. Mol Cell Biol 2007;27:3056–3064. [PubMed: 17283045]

Ma Z, Morris SW, Valentine V, Li M, Herbrick JA, Cui X, Bouman D, Li Y, Mehta PK, Nizetic D, etal. Fusion of two novel genes, RBM15 and MKL1 in the t(1:22)(p13:q13) of acute megakaryoblasticleukemia. Nat Genet 2001;28:220–221. [PubMed: 11431691]

Mansouri M, Viswanathan K, Douglas JL, Hines J, Gustin J, Moses AV, Fruh K. Molecular mechanismof BST2/tetherin downregulation by K5/MIR2 of Kaposi's sarcoma herpesvirus. J Virol July 15.2009Epub ahead of print

Martowicz ML, Grass JA, Boyer ME, Guend H, Bresnick EH. Dynamic GATA factor interplay at a multi-component regulatory region of the GATA-2 locus. J Biol Chem 2005;280:1724–1732. [PubMed:15494394]

Fujiwara et al. Page 12

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Merika M, Orkin SH. DNA-binding specificity of GATA family transcription factors. Mol Cell Biol1993;13:3999–4010. [PubMed: 8321207]

Migliaccio AR, Rana RA, Sanchez M, Lorenzini R, Centurione L, Bianchi L, Vannucchi AM, MigliaccioG, Orkin SH. GATA-1 as a regulator of mast cell differentiation revealed by the phenotype of theGATA-1low mouse mutant. J Exp Med 2003;197:281–296. [PubMed: 12566412]

Mohandas N, Gallagher PG. Red cell membrae: past, present, and future. Blood 2008;112:3939–3948.[PubMed: 18988878]

Munugalavadla V, Dore LC, Tan BL, Hong L, Vishnu M, Weiss MJ, Kapur R. Repression of c-kit andits downstream substrates by GATA-1 inhibits cell proliferation during erythroid maturation. MolCell Biol 2005;25:6747–6759. [PubMed: 16024808]

Nguyen DX, Chiang AC, Zhang XH, Kim JY, Kris MG, Ladanyi M, Gerald WL, Massague J. WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis. Cell2009;138:51–62. [PubMed: 19576624]

Pal S, Cantor AB, Johnson KD, Moran T, Boyer ME, Orkin SH, Bresnick EH. Coregulator-dependentfacilitation of chromatin occupancy by GATA-1. Proc Natl Acad Sci U S A 2004a;101:980–985.[PubMed: 14715908]

Pal S, Nemeth MJ, Bodine DM, Miller JL, Svaren J, Thein SL, Lowry PJ, Bresnick EH. Neurokinin-Btranscription in erythroid cells: direct activation by the hematopoietic transcription factor GATA-1.J Biol Chem 2004b;279:31348–31356. [PubMed: 15123623]

Park C, Afrikanova I, Chung YS, Zhang WJ, Arentson E, Fong GH, Rosendahl A, Choi K. A hierarchicalorder of factors in the generation of FLK1- and SCL-expressing hematopoietic and endothelialprogenitors from embryonic stem cells. Development 2004;131:2749–2762. [PubMed: 15148304]

Pevny L, Simon MC, Robertson E, Klein WH, Tsai SF, D'Agati V, Orkin SH, Costantini F. Erythroiddifferentiation in chimaeric mice blocked by a targeted mutation in the gene for transcription factorGATA-1. Nature 1991;349:257–260. [PubMed: 1987478]

Porcher C, Liao EC, Fujiwara Y, Zon LI, Orkin SH. Specification of hematopoietic and vasculardevelopment by the bHLH transcription factor SCL without direct DNA binding. Development1999;126:4603–4615. [PubMed: 10498694]

Porcher C, Swat W, Rockwell K, Fujiwara Y, Alt FW, Orkin SH. The T cell leukemia oncoprotein SCL/tal-1 is essential for development of all hematopoietic lineages. Cell 1996;86:47–57. [PubMed:8689686]

Raffel GD, Mercher T, Shigematsu H, Williams IR, Cullen DE, Akashi K, Bernard OA, Gilliland DG.Ott1 (Rbm15) has pleitropic roles in hematopoietic development. Proc Natl Acad Sci U S A2007;104:6001–6006. [PubMed: 17376872]

Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O, Bock R, Gass P, Schmid W, HerrlichP, Angel P, Schutz G. DNA binding of the glucocorticoid receptor is not essential for survival. Cell1998;93:531–541. [PubMed: 9604929]

Rekhtman N, Radparvar F, Evans T, Skoultchi AI. Direct interaction of hematopoietic transcriptionfactors PU.1 and GATA-1: functional antagonism in erythroid cells. Genes Dev 1999;13:1398–1411.[PubMed: 10364157]

Scherzer CR, Grass JA, Liao Z, Pepivani I, Zheng B, Eklund AC, Ney PA, J N, McGoldrick M,Mollenhauer B, et al. GATA transcription factors directly regulate the Parkinson's disease-linkedgene alpha-synuclein. Proc Natl Acad Sci USA 2008;105:10907–10912. [PubMed: 18669654]

Schuh AH, Tipping AJ, Clark AJ, Hamlett I, Guyot B, Iborra FJ, Rodriguez P, Strouboulis J, Enver T,Vyas P, Porcher C. ETO-2 associates with SCL in erythroid cells and megakaryocytes and providesrepressor functions in erythropoiesis. Mol Cell Biol 2005;25:10235–10250. [PubMed: 16287841]

Sekiya T, Muthurajan UM, Luger K, Tulin AV, Zaret KS. Nucleosome-binding affinity as a primarydeterminant of the nuclear mobility of the pioneer transcription factor FoxA. Genes Dev2009;23:804–809. [PubMed: 19339686]

Sharrocks AD. The ETS-domain transcription factor family. Nat Rev Mol Cell Biol 2001;2:827–837.[PubMed: 11715049]

Shivdasani RA, Fujiwara Y, McDevitt MA, Orkin SH. A lineage-selective knockout establishes thecritical role of transcription factor GATA-1 in mekagaryocyte growth and platelet development.EMBO J 1997;16:3965–3973. [PubMed: 9233806]

Fujiwara et al. Page 13

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Shivdasani RA, Mayer EL, Orkin SH. Absence of blood formation in mice lacking the T-cell leukaemiaoncoprotein tal-1/SCL. Nature 1995;373:432–434. [PubMed: 7830794]

Simon MC, Pevny L, Wiles MV, Keller G, Costantini F, Orkin SH. Rescue of erythroid development ingene targeted GATA-1- mouse embryonic stem cells. Nat Genet 1992;1:92–98. [PubMed: 1302015]

Stumpf M, Waskow C, Krotschel M, van Essen D, Rodriguez P, Zhang X, Guyot B, Roeder RG, BorggrefeT. The mediator complex functions as a coactivator for GATA-1 in erythropoiesis via subunit Med1/TRAP220. Proc Natl Acad Sci U S A 2006;103:18504–18509. [PubMed: 17132730]

Tripic T, Deng W, Cheng Y, Vakoc CR, Gregory GD, Hardison RC, Blobel GA. SCL and associatedprotein distinguish active from repressive GATA transcription factor complexes. Blood2008;113:2191–2201. [PubMed: 19011221]

Trouche D, Kouzarides T. E2F1 and E1A(12S) have a homologous activation domain regulated by RBand CBP. Proc Natl Acad Sci U S A 1996;93:1439–1442. [PubMed: 8643650]

Tsai FY, Keller G, Kuo FC, Weiss M, Chen J, Rosenblatt M, Alt FW, Orkin SH. An early haematopoieticdefect in mice lacking the transcription factor GATA-2. Nature 1994;371:221–226. [PubMed:8078582]

Tsai SF, Martin DI, Zon LI, D'Andrea AD, Wong GG, Orkin SH. Cloning of cDNA for the major DNA-binding protein of the erythroid lineage through expression in mammalian cells. Nature1989;339:446–451. [PubMed: 2725678]

Tsang AP, Visvader JE, Turner CA, Fujuwara Y, Yu C, Weiss MJ, Crossley M, Orkin SH. FOG, amultitype zinc finger protein as a cofactor for transcription factor GATA-1 in erythroid andmegakaryocytic differentiation. Cell 1997;90:109–119. [PubMed: 9230307]

Turner J, Crossley M. Cloning and characterization of mCtBP2, a co-repressor that associates with basicKruppel-like factor and other mammalian transcriptional regulators. EMBO J 1998;17:5129–5140.[PubMed: 9724649]

Vakoc CR, Letting DL, Gheldof N, Sawado T, Bender MA, Groudine M, Weiss MJ, Dekker J, BlobelGA. Proximity among distant regulatory elements at the beta globin locus requires GATA-1 andFOG-1. Mol Cell 2005;17:453–462. [PubMed: 15694345]

Vyas P, McDevitt MA, Cantor AB, Katz SG, Fujiwara Y, Orkin SH. Different sequence requirementsfor expression in erythroid and megakaryocytic cells within a regulatory element upstream of theGATA-1 gene. Development 1999;126:2799–2811. [PubMed: 10331989]

Wadman IA, Osada H, Grutz GG, Agulnick AD, Westphal H, Forster A, Rabbitts TH. The LIM-onlyprotein Lmo2 is a bridging molecule assembling an erythroid, DNA-binding complex which includesthe TAL1, E47, GATA-1 and Ldb1/NLI proteins. EMBO J 1997;16:3145–3157. [PubMed: 9214632]

Wechsler J, Greene M, McDevitt MA, Anastasi J, J.E K, LeBeau MM, Crispino JD. Acquired mutationsin GATA-1 in the megakaryoblastic leukemia of Down syndrome. Nat Genet 2002;32:148–152.[PubMed: 12172547]

Weiss MJ, Yu C, Orkin SH. Erythroid-cell-specific properties of transcription factor GATA-1 revealedby phenotypic rescue of a gene-targeted cell line. Mol Cell Biol 1997;17:1642–1651. [PubMed:9032291]

Welch JJ, Watts JA, Vakoc CR, Yao Y, Wang H, Hardison RC, Blobel GA, Chodosh LA, Weiss MJ.Global regulation of erythroid gene expression by transcription factor GATA-1. Blood2004;104:3136–3147. [PubMed: 15297311]

Wozniak RJ, Boyer ME, Grass JA, Lee Y-S, Bresnick EH. Context-dependent GATA factor function:combinatorial requirements for transcriptional control in hematopoietic and endothelial cells. J BiolChem 2007;282:14665–14674. [PubMed: 17347142]

Wozniak RJ, Keles S, Lugus JJ, Young K, Boyer ME, Tran TT, Choi K, Bresnick EH. Molecularhallmarks of endogenous chromatin complexes containing master regulators of hematopoiesis. MolCell Biol 2008;28:6681–6694. [PubMed: 18779319]

Xu Z, Huang S, Chang LS, Agulnick AD, Brandt SJ. Identification of a TAL1 target gene reveals apositive role for the LIM domain-binding protein (Ldb1) in erythroid gene expression anddifferentiation. Mol Cell Biol 2003;23:7585–7599. [PubMed: 14560005]

Yu C, Cantor AB, Yang H, Browne C, Wells RA, Fukiwara Y, Orkin SH. Targeted deletion of a high-affinity GATA-binding site in the GATA-1 promoter leads to selective loss of the eosinophil lineagein vivo. J Exp Med 2002;195:1387–1395. [PubMed: 12045237]

Fujiwara et al. Page 14

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 1. Representative GATA-1 Targets Identified Through ChIP-seq AnalysisGATA-1 signal maps are shown for representative hematopoietic transcription factors,signaling molecules and red cell cytoskeletal proteins. Arrows, ChIP-seq peak locationsrelative to the transcription start site of the respective GATA-1 target gene (kb).

Fujiwara et al. Page 15

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 2. Validation of Human ChIP-seq Results in Murine G1E-ER-GATA-1 cellsQuantitative real-time ChIP analysis of ER-GATA-1 and GATA-2 occupancy at 13 high (A)and low (B) ChIP-seq hits containing conserved WGATAR motifs in both β-estradiol-untreated and -treated (1 μM, 24 h) G1E-ER-GATA-1 cells (mean +/− SE, 3 independentexperiments).

Fujiwara et al. Page 16

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 3. Computational/Statistical Mining of ChIP-seq Data(A, B) The 5,749 ChIP-seq peaks were classified by (A) locations relative to nearest-neighborgenes and (B) GATA motif pattern within each peak. (C) Sequence logos from 5051WGATAR-containing (left) and 301 E-box-WGATAR-containing (right) peaks. The logo onthe left was obtained by de novo motif finding using MEME, while the logo on the right wasgenerated by aligning E-box-WGATAR sequences within peaks. Information content wasmeasured in bits (ranging from 0 to 2 for a given position of sequence). A position in the motifat which all nucleotides occur with equal probability has a value of 0, while a position at whichonly a single nucleotide can occur has a value of 2. (D) Merged ChIP-seq and Illuminaexpression profiling results. ChIP-seq peaks were merged with array data profiling expressionin untreated and β-estradiol-treated (1 μM, 24 h) G1E-ER-GATA-1 cells (2 independentexperiments). Among 296 genes shared by both datasets, the top 60 activated and repressedgenes are shown. Asterisk, gene demonstrated previously to be differentially expressed in G1E-ER-GATA-1 cells (Welch et al., 2004). (E) Quantitative ChIP analysis of GATA-1 occupancyin primary murine Ter119+ bone marrow cells (mean +/− SE, 2 independent experiments).Asterisk, significantly greater GATA-1 occupancy compared to the Ey promoter (p < 0.05).Preimmune signals were analyzed with all primer sets and did not exceed 0.0025.

Fujiwara et al. Page 17

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 4. Comparison of GATA-1 and GATA-2 Chromatin Occupancy Genome-Wide by ChIP-seq(A) Peaks were called on the merged GATA-1 and merged GATA-2 replicate datasets (9,042and 21,167 for GATA-1 and GATA-2, respectively). GATA-2 peaks were ranked andtruncated to the size of the GATA-1 peak list. A comparison of the 9,042 GATA-1 and GATA-2peaks revealed 65% overlap. Using the ENCODE overlap rule, the top 40% of each peak filewas compared to the entire set of 9,042 peaks for the other factor. A 90-91% overlap wasobserved, indicating that the majority of the highest ranked peaks for each factor are containedwithin the peak set for the other factor. When the ENCODE overlap rule is applied to replicatedatasets of the same factor, high quality datasets often overlap by 80-90%. (B) ChIP-seq signal

Fujiwara et al. Page 18

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

maps for GATA-1/GATA-2-shared targets. Arrows, ChIP-seq peak locations relative to thetranscription start site (kb). (C) Quantitative ChIP analysis of GATA factor occupancy atGATA-1- and GATA-2-selective targets in K562 cells (mean +/− SE, 3 independentexperiments). (D) ChIP-seq signal maps for GATA-1- and GATA-2-selective targets. Arrows,ChIP-seq peak locations relative to the transcription start site (kb).

Fujiwara et al. Page 19

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 5. ETO-2 Negative Autoregulatory Loop(A) Cbfa2t3 locus organization (UCSC genome assembly: uc009ntn.1). Open and filled boxes,noncoding and coding exons, respectively. Asterisk, locations of conserved E-boxWGATARmotif; downward pointing arrows, WGATAR motifs conserved from mouse to human. FourWGATAR motifs located at −21.7, −21.6, −21.6, and −21.5 kb were analyzed as a cluster. TheVISTA plot depicts sequence identity between mouse and human, using mouse as a reference.(B) Quantitative RT-PCR analysis of Cbfa2t3 mRNA (left) (mean +/− SE, 4 independentexperiments) and anti-ETO-2 Western blot of whole cell extracts (right) from G1E cellstransfected with siRNA against mouse Cbfa2t3 or control siRNA. Asterisk, cross-reactiveband. (C) Analysis of ETO-2 occupancy at the Cbfa2t3 locus (−21.6 kb) in ETO-2-knockdownand control G1E cells (mean +/− SE, 2 independent experiments). (D) Cbfa2t3 primarytranscripts were quantitated by real-time RT-PCR analysis in control and ETO-2-knockdownG1E cells (mean +/− SE, 4 independent experiments). Two primer sets (Intron1/Intron1 andIntron11/Exon12) were used. Gapdh mRNA was quantitated as a control. (E) Analysis of PolII and P-Ser5-Pol II occupancy at Cbfa2t3 (left and middle) and RPII215 (right) promoters inETO-2-knockdown and control G1E cells (mean +/− SE, 2 independent experiments). Twoprimer sets were used to analyze the Cbfa2t3 promoter. (F) Analysis of AcH3 at the Cbfa2t3−21.6 kb (left), Cbfa2t3 promoter-B (middle), and RPII215 promoter (right) in ETO-2-knockdown and control G1E cells (mean +/− SE, 2 independent experiments).

Fujiwara et al. Page 20

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 6. Context-Dependent ETO-2 Corepressor Function at Endogenous Loci(A) The diagram depicts the location of the ChIP-seq peaks analyzed. Asterisk, location of thecorresponding peak in mouse; shaded boxes, coding regions. (B) Analysis of ETO-2 occupancyat GATA target genes in G1E cells transfected with control or Cbfa2t3 siRNA (mean +/− SE,2 independent experiments). Asterisk, p < 0.05. (C, D) Real-time RT-PCR analysis of GATAtarget genes in G1E cells (C) and G1E-ER-GATA1 cells (D) transfected with control orCbfa2t3 siRNA. For G1E-ER-GATA1 cells, β-estradiol was added 24 h after the initial siRNAtransfection and cells were cultured for 24 h. mRNA levels were normalized to Gapdh mRNA(mean +/− SE, 3 and 4 independent experiments for Fig. 6C and 6D, respectively). The relativetranscript level for control G1E-ER-GATA-1 cells was designated as 1. Asterisk, p < 0.05. (E)Models for ETO-2-dependent repression of Slc4a1. GATA-2 occupies the repressed Slc4a1promoter (left), and ETO-2 loss suffices for induction (right, upper) or is coupled to GATA-2loss (right, lower). (F) Analysis of GATA-2 occupancy at the Slc4a1 promoter in G1E cellstransfected with control or Cbfa2t3 siRNA (mean +/− SE, 2 independent experiments).

Fujiwara et al. Page 21

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 7. Regulatory Interactions Among Components of Complexes Containing MasterRegulators of Hematopoiesis and Leukemogenic Factors(A) The table summarizes changes in the expression of genes in day 3/4, 6 and 8 EBs derivedfrom mouse ES cells following Dox-mediated GATA-2 induction (mean +/− SE, 9 independentexperiments for day 3/4 and day 6 EBs; 6 independent experiments for day 8 EBs), day 4 EBsfrom mouse ES cells following Dox-mediated Scl/TAL1 induction (mean +/− SE, 3independent experiments), and G1E-ER-GATA1 cells after β-estradiol-treatment (1 μM for 3,8 and 24 h; mean +/− SE, 2 independent experiments). For EBs derived from Gata2−/− andScl/TAL1−/− ES cells, values are expressed relative to that of wild-type EBs, which has a valueof 1.0. mRNA levels were quantitated by real-time RT-PCR, and the expression level wasdivided by that of corresponding control cells. (B) The model summarizes cross-regulatoryinteractions among Scl/TAL1 complex components. (C) Model demonstrating GATA-2activation of Cbfa2t3 transcription, ETO-2 negative autoregulatory loop, and GATA switch-mediated Cbfa2t3 repression.

Fujiwara et al. Page 22

Mol Cell. Author manuscript; available in PMC 2010 November 25.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript