Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem...

10
Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumar a,b , Kirby D. Johnson a,b , Xin Gao a,b , Meghan E. Boyer a,b , Yuan-I Chang b,c , Kyle J. Hewitt a,b , Jing Zhang b,c , and Emery H. Bresnick a,b,1 a Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705; c McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706; and b University of WisconsinMadison Blood Research Program, Madison, WI 53705 Edited by Stuart H. Orkin, Childrens Hospital and the DanaFarber Cancer Institute, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, and approved February 11, 2014 (received for review January 2, 2014) The unremitting demand to replenish differentiated cells in tissues requires efficient mechanisms to generate and regulate stem and progenitor cells. Although master regulatory transcription factors, including GATA binding protein-2 (GATA-2), have crucial roles in these mechanisms, how such factors are controlled in developmentally dy- namic systems is poorly understood. Previously, we described five dispersed Gata2 locus sequences, termed the -77, -3.9, -2.8, -1.8, and +9.5 GATA switch sites, which contain evolutionarily conserved GATA motifs occupied by GATA-2 and GATA-1 in hematopoietic precursors and erythroid cells, respectively. Despite common attributes of transcriptional enhancers, targeted deletions of the -2.8, -1.8, and +9.5 sites revealed distinct and unpredictable contributions to Gata2 expression and hematopoiesis. Herein, we describe the targeted deletion of the -3.9 site and mechanistically compare the -3.9 site with other GATA switch sites. The -3.9 -/- mice were viable and exhibited normal Gata2 expression and steady-state hematopoiesis in the embryo and adult. We established a Gata2 repression/reactivation assay, which revealed unique +9.5 site activity to mediate GATA factor-dependent chromatin structural transitions. Loss-of-function analyses provided evidence for a mechanism in which a mediator of long-range transcriptional con- trol [LIM domain binding 1 (LDB1)] and a chromatin remodeler [Brahma related gene 1 (BRG1)] synergize through the +9.5 site, conferring expression of GATA-2, which is known to promote the genesis and survival of hematopoietic stem cells. cis element | HSCs W hereas proximal promoter sequences assemble the basal transcriptional machinery and RNA polymerase, distant cis-regulatory elements often confer tissue-specific or context- dependent transcriptional regulation. Enhancer elements reside many kilobases upstream or downstream of a promoter or within introns, and extensive efforts have focused on elucidating ac- tion-at-a-distancemechanisms (1). Long-range transcriptional control involves physical interactions between proteins bound at distal regions and promoter sequences and higher order struc- tural transitions, including subnuclear relocalization of target loci (24). Given the high frequency of long-range mechanisms at mammalian loci and the mutations that disrupt the function of such elements in pathological conditions, elucidating the un- derlying mechanisms in development, tissue homeostasis, and disease is critically important. In the context of the essential process of hematopoiesis, we have been dissecting long-range mechanisms controlling the expression and function of the mas- ter regulator GATA binding protein-2 (GATA-2) (512). The dual zinc finger transcription factor GATA-2 is expressed in hematopoietic stem cells (HSCs), select hematopoietic pro- genitors, endothelial cells, neurons, and additional specialized cell types (1318). Targeted deletion of Gata2 revealed its es- sential function for hematopoiesis. Gata2-nullizygous mouse embryos die from severe anemia at embryonic day (E) 10.5 (13, 15), and Gata2 +/- HSCs have reduced activity in competitive transplants (19, 20). Heterozygous mutations of GATA2 underlie the development of a human immunodeficiency syndrome, mon- ocytopenia and mycobacterial infection (MonoMAC), and related disorders, which are accompanied by myelodysplastic syndrome and acute myeloid leukemia (2123). Although the critical role of GATA-2 in hematopoietic stem/progenitor biology has been established through rigorous genetic studies, many questions remain unanswered regarding mechanisms underlying Gata2 ex- pression and regulation. Studies in cultured and primary erythroid cells revealed five GATA-1and GATA-2occupied upstream (-77, -3.9, -2.8, and -1.8 kb) and intronic (+9.5 kb) sites of the Gata2 locus (10). Because GATA-2 occupies these prospective regulatory sites in erythroid precursor cells lacking GATA-1, we proposed that this reflects GATA-2mediated positive autoregulation (10). Be- cause GATA-1 is expressed during erythropoiesis, it displaces GATA-2, instigating Gata2 repression (24). GATA-1mediated displacement of GATA-2 from chromatin is termed GATA switching, and the GATA factor-occupied sites are deemed GATA switch sites (10, 24). Despite the compelling biochemical and molecular attributes of the GATA switch sites, targeted deletion of the -1.8 and -2.8 sites individually in the mouse revealed only minor roles in max- imizing Gata2 expression in hematopoietic precursors (6, 7). The -1.8 -/- and -2.8 -/- mice were born at normal Mendelian ra- tios, and hematopoiesis was largely normal in steady-state and stress contexts. The -1.8 element is uniquely required to maintain, but Significance The continuous replenishment of differentiated cells, for example, those constituting the blood, involves proteins that control the generation and function of stem and progenitor cells. Although master regulatorsare implicated in these processes, many ques- tions remain unanswered regarding how their synthesis and activities are regulated. We describe a mechanism that con- trols the production of the master regulator GATA binding protein-2 (GATA-2) in the context of blood stem and progenitor cells. Thousands of GATA-2 binding sites exist in the genome, and genetic analyses indicate that they differ greatly and un- predictably in functional importance. The parameters involved in endowing sites with functional activity are not established. We describe unique insights into ascertaining functionally im- portant GATA-2 binding sites within chromosomes. Author contributions: R.S., K.D.J., X.G., and E.H.B. designed research; R.S., K.D.J., X.G., M.E.B., and Y.-I.C. performed research; K.D.J. contributed new reagents/analytic tools; R.S., K.D.J., X.G., K.J.H., J.Z., and E.H.B. analyzed data; and R.S., K.D.J., and E.H.B. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. 1 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1400065111/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1400065111 PNAS | Published online March 10, 2014 | E1091E1100 GENETICS PNAS PLUS Downloaded by guest on November 24, 2020

Transcript of Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem...

Page 1: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

Mechanism governing a stem cell-generatingcis-regulatory elementRajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin Gaoa,b, Meghan E. Boyera,b, Yuan-I Changb,c, Kyle J. Hewitta,b,Jing Zhangb,c, and Emery H. Bresnicka,b,1

aDepartment of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, Carbone Cancer Center, University of Wisconsin School of Medicineand Public Health, Madison, WI 53705; cMcArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI53706; and bUniversity of Wisconsin–Madison Blood Research Program, Madison, WI 53705

Edited by Stuart H. Orkin, Children’s Hospital and the Dana–Farber Cancer Institute, Harvard Medical School and Howard Hughes Medical Institute, Boston,MA, and approved February 11, 2014 (received for review January 2, 2014)

The unremitting demand to replenish differentiated cells in tissuesrequires efficient mechanisms to generate and regulate stem andprogenitor cells. Although master regulatory transcription factors,including GATA binding protein-2 (GATA-2), have crucial roles in thesemechanisms, how such factors are controlled in developmentally dy-namic systems is poorly understood. Previously, we described fivedispersed Gata2 locus sequences, termed the −77, −3.9, −2.8, −1.8,and +9.5 GATA switch sites, which contain evolutionarily conservedGATA motifs occupied by GATA-2 and GATA-1 in hematopoieticprecursors and erythroid cells, respectively. Despite commonattributes of transcriptional enhancers, targeted deletions ofthe −2.8, −1.8, and +9.5 sites revealed distinct and unpredictablecontributions to Gata2 expression and hematopoiesis. Herein, wedescribe the targeted deletion of the −3.9 site and mechanisticallycompare the−3.9 site with other GATA switch sites. The−3.9−/− micewere viable and exhibited normal Gata2 expression and steady-statehematopoiesis in the embryo and adult. We established a Gata2repression/reactivation assay, which revealed unique +9.5 siteactivity to mediate GATA factor-dependent chromatin structuraltransitions. Loss-of-function analyses provided evidence for amechanism in which a mediator of long-range transcriptional con-trol [LIM domain binding 1 (LDB1)] and a chromatin remodeler[Brahma related gene 1 (BRG1)] synergize through the +9.5 site,conferring expression of GATA-2, which is known to promote thegenesis and survival of hematopoietic stem cells.

cis element | HSCs

Whereas proximal promoter sequences assemble the basaltranscriptional machinery and RNA polymerase, distant

cis-regulatory elements often confer tissue-specific or context-dependent transcriptional regulation. Enhancer elements residemany kilobases upstream or downstream of a promoter or withinintrons, and extensive efforts have focused on elucidating “ac-tion-at-a-distance” mechanisms (1). Long-range transcriptionalcontrol involves physical interactions between proteins bound atdistal regions and promoter sequences and higher order struc-tural transitions, including subnuclear relocalization of targetloci (2–4). Given the high frequency of long-range mechanismsat mammalian loci and the mutations that disrupt the function ofsuch elements in pathological conditions, elucidating the un-derlying mechanisms in development, tissue homeostasis, anddisease is critically important. In the context of the essentialprocess of hematopoiesis, we have been dissecting long-rangemechanisms controlling the expression and function of the mas-ter regulator GATA binding protein-2 (GATA-2) (5–12).The dual zinc finger transcription factor GATA-2 is expressed

in hematopoietic stem cells (HSCs), select hematopoietic pro-genitors, endothelial cells, neurons, and additional specializedcell types (13–18). Targeted deletion of Gata2 revealed its es-sential function for hematopoiesis. Gata2-nullizygous mouseembryos die from severe anemia at embryonic day (E) 10.5 (13,15), and Gata2+/− HSCs have reduced activity in competitive

transplants (19, 20). Heterozygous mutations of GATA2 underliethe development of a human immunodeficiency syndrome, mon-ocytopenia and mycobacterial infection (MonoMAC), and relateddisorders, which are accompanied by myelodysplastic syndromeand acute myeloid leukemia (21–23). Although the critical roleof GATA-2 in hematopoietic stem/progenitor biology has beenestablished through rigorous genetic studies, many questionsremain unanswered regarding mechanisms underlying Gata2 ex-pression and regulation.Studies in cultured and primary erythroid cells revealed five

GATA-1– and GATA-2–occupied upstream (−77, −3.9, −2.8,and −1.8 kb) and intronic (+9.5 kb) sites of the Gata2 locus (10).Because GATA-2 occupies these prospective regulatory sites inerythroid precursor cells lacking GATA-1, we proposed that thisreflects GATA-2–mediated positive autoregulation (10). Be-cause GATA-1 is expressed during erythropoiesis, it displacesGATA-2, instigating Gata2 repression (24). GATA-1–mediateddisplacement of GATA-2 from chromatin is termed GATAswitching, and the GATA factor-occupied sites are deemedGATA switch sites (10, 24).Despite the compelling biochemical and molecular attributes

of the GATA switch sites, targeted deletion of the −1.8 and −2.8sites individually in the mouse revealed only minor roles in max-imizing Gata2 expression in hematopoietic precursors (6, 7).The −1.8−/− and −2.8−/− mice were born at normal Mendelian ra-tios, and hematopoiesis was largely normal in steady-state and stresscontexts. The −1.8 element is uniquely required to maintain, but

Significance

The continuous replenishment of differentiated cells, for example,those constituting the blood, involves proteins that control thegeneration and function of stem and progenitor cells. Although“master regulators” are implicated in these processes, many ques-tions remain unanswered regarding how their synthesis andactivities are regulated. We describe a mechanism that con-trols the production of the master regulator GATA bindingprotein-2 (GATA-2) in the context of blood stem and progenitorcells. Thousands of GATA-2 binding sites exist in the genome,and genetic analyses indicate that they differ greatly and un-predictably in functional importance. The parameters involvedin endowing sites with functional activity are not established.We describe unique insights into ascertaining functionally im-portant GATA-2 binding sites within chromosomes.

Author contributions: R.S., K.D.J., X.G., and E.H.B. designed research; R.S., K.D.J., X.G.,M.E.B., and Y.-I.C. performed research; K.D.J. contributed new reagents/analytic tools;R.S., K.D.J., X.G., K.J.H., J.Z., and E.H.B. analyzed data; and R.S., K.D.J., and E.H.B. wrotethe paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.1To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1400065111/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1400065111 PNAS | Published online March 10, 2014 | E1091–E1100

GEN

ETICS

PNASPL

US

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 2: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

not to initiate,Gata2 repression in late-stage erythroblasts, but thismolecular defect was not coupled to major functional deficits (6).In contrast to the −1.8 and −2.8 site deletions, targeted deletionof the +9.5 intronic site is embryonically lethal at E13.5–E14.5(5). The +9.5 site is essential for GATA-2 expression in hema-topoietic stem and progenitor cells (HSPCs) and in endotheliumduring embryogenesis (5, 9, 25, 26). Definitive hematopoiesis isseverely impaired in +9.5−/− mice due to defective HSC produc-tion, as demonstrated by competitive transplants and imaging ofHSC genesis from hemogenic endothelium in the dorsal aorta (25).The +9.5 site contains an E-box–GATA composite element,

which mediates assembly of a complex containing GATA-1 orGATA-2, T-cell acute lymphocytic leukemia 1 (TAL1), LIMdomain binding 1 (LDB1), and LIM domain only 2 (LMO2).The GATA and E-box motifs, and the spacing between the

motifs, are essential for +9.5 site enhancer activity in reporterassays (11). The E-box binding protein TAL1 cooperates withGATA factors in the assembly of a multicomponent complex onE-box–GATA composite elements at genes important for bloodcell development and function (27–33). The TAL1-interactingproteins LDB1 and LMO2 control the development and functionof HSPCs (22, 34–38). In addition to binding sites containingGATA–E-box composite elements, like the +9.5 site, TAL1 occu-pies GATA motif-containing sites lacking a consensus E-box, pre-sumably via recruitment by the GATA factor (28). The LIMdomain binding-1 coregulator LDB1 promotes chromatin loop-ing (39, 40) and facilitates HSC maintenance, primitive hema-topoietic progenitor generation, and erythroid differentiation(37, 41–44). Certain patients with MonoMAC who lack GATA2coding region mutations harbored deletion or point mutations in

Fig. 1. The −3.9 GATA switch site bears hallmarks of an important cis-regulatory element. (A) Sequence alignment of the −3.9 site demonstrates conser-vation among vertebrates. The WGATAR motifs and intervening sequence that were removed by homologous recombination are indicated. (B) ChIP-sequencing profiles for factor occupancy and histone modifications at the Gata2 locus mined from existing datasets (37, 69–72). BM, bone marrow;H3K27a, acetylation of H3 at lysine 27; H3K4m1, monomethylation of histone H3 at lysine 4; MEL, murine erythroleukemia cells. (C) Strategy for targeteddeletion of the −3.9 site. Following NeoR excision, the targeted allele has a 126-bp Xba I-to-Not I fragment containing a single LoxP site substituted for theGATA motifs and intervening sequence. Arrowheads indicate positions of primers used for genotype determination. (D) Representative gel shows PCR-basedstrategy to distinguish WT and targeted alleles following NeoR excision. (E) Genotypes of viable pups from mating −3.9+/− males and females determined atthe time of weaning. Expected numbers of pups based on Mendelian ratios are shown in parenthesis. (F) Representative −3.9+/+ and −3.9−/− embryos at E12.5.

E1092 | www.pnas.org/cgi/doi/10.1073/pnas.1400065111 Sanalkumar et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 3: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

or near the +9.5 site (5, 45). Thus, mutations of the +9.5 element,an essential mediator of definitive hematopoiesis in the mouse,underlie human hematopoietic pathology.The contributions of the −77 and −3.9 GATA switch sites to

Gata2 regulation in vivo have not been reported. The −3.9 siteharbors two inverted GATA motifs and contains canonicalattributes of cis-regulatory elements, including DNase I hyper-sensitivity and GATA site-dependent enhancer activity in atransfection assay (9, 12). Because these attributes are sharedwith one or more of the −2.8, −1.8, and +9.5 sites, which differgreatly in their importance in vivo, the contribution of the −3.9site can only be ascertained by disruption of this site at the en-dogenous locus. Herein, we describe the consequences of a −3.9site deletion from the endogenous Gata2 locus and a mecha-nistic comparison of the −3.9 site with the −2.8, −1.8, and +9.5sites in biologically relevant contexts. These studies led to a modelto explain the unique importance of the +9.5 site.

ResultsSequence Conservation and Transcription Factor Occupancy Do NotPredict Gata2 Cis-Element Function in Vivo. The −3.9 site containsinverted WGATAR motifs that are well conserved among verte-brates (Fig. 1A). GATA-2 occupancy of the −3.9 site, first de-scribed in GATA-1–null G1E cells (12), occurs in multiple celllines and primary cells, including lineage negative (Lin−) he-matopoietic progenitors from bone marrow (Fig. 1B). Featurescommonly associated with enhancers [DNase hypersensitivity,monomethylation of histone H3 at lysine 4, acetylation of H3 atlysine 27, and p300 occupancy (46, 47)] characterize the −3.9 siteand other GATA switch sites. To assess −3.9 site function, we

deleted 27 nucleotides encompassing the GATA motifs andintervening sequence by homologous recombination and excisedthe NeoR gene (Fig. 1C). Targeted and WT alleles were distin-guishable by PCR with primers flanking the −3.9 site (Fig. 1D).Like −1.8 and −2.8 mice, genotypes of −3.9−/− and −3.9+/− mutantsconformed to Mendelian genetics, indicating the −3.9 site is dis-pensable for viability (Fig. 1E). The −3.9−/− embryos wereindistinguishable from WT littermates and did not exhibit he-matopoietic or vascular defects characteristic of +9.5−/− embryos(Fig. 1F).Although a singleGata2 allele is sufficient to confer viability in

Gata2+/− and +9.5+/− mouse strains, heterozygosity in thesemice reduces Gata2 expression, as well as HSC generation andfunction (5, 19, 20, 25). To assess the influence of the −3.9mutation on HSC genesis, we conducted whole-mount 3D em-bryo imaging to visualize c-Kit+ hematopoietic clusters con-taining HSCs in E10.5 aorta gonad mesonephros (AGM) of−3.9+/+ and −3.9−/− littermates. Whereas +9.5−/− AGM is al-most devoid of c-Kit staining (25), −3.9−/− AGM hematopoieticclusters were indistinguishable in size and number vs. those ofWT littermates (Fig. 2A). Quantitation of Gata2 mRNA levels inE13.5 fetal livers and brains revealed no differences between−3.9+/+, −3.9+/−, and −3.9−/− littermates (Fig. 2B).In adult −3.9+/+, −3.9+/−, and −3.9−/− mice, complete blood

cell count (CBC) measurements were compared at 2 and 6 mo ofage (Tables S1 and S2). The quantities of circulating blood celltypes were indistinguishable. Because GATA-2 haploinsufficiencyincreases quiescence and apoptosis in primitive bone marrow cellswithout changes to circulating blood cells (19), the −3.9 mutationmight alter GATA-2 levels in HSPCs without significantly affect-

Fetal Liver Fetal Brain

BA

0

0.5

1.0

Gat

a2 m

RN

A L

evel

s(R

elat

ive

Un

its)

-RT+/-+/+ -/- -RT+/-+/+ -/-

CD31c-Kit

DA

ADAD

-3.9+/+ -3.9-/-

+9.5-/-

CD31c-Kit CD31c-Kit

0

100

200

300

400

500

c-K

it

cells

per

DA

+* **

-3.9+/+ -3.9-/- +9.5-/-

0

0.2

0.4

0.6

0.8

1.0Mpl Hbb-b1

0

0.08

0.16

0.24

0.32

Lin- Ter119+ -RT0

0.05

0.10

0.15

+/+-/-

+/+ -/-

Gat

a2 m

RN

A L

evel

s (R

elat

ive

Un

its)

Gat

a2 m

RN

A L

evel

s (R

elat

ive

Un

its)

Sca

-1

c-Kit

103

104

105

+/+

0 103

104

105

103

104

105

-/-

+ Sca Kit+

mR

NA

Lev

els

(Rel

ativ

e U

nit

s)

+ Sca Kit+ + Lin Sca Kit+ - D F

Lin

-

EC

Ter1

19+

Ter1

19-

Lin

+

-RT

Lin

-

Ter1

19+

Ter1

19-

Lin

+

-RT

Fig. 2. The −3.9 site is dispensable for Gata2 expression during hematopoiesis. (A) Whole-mount immunostaining of CD31+ cells (magenta) and c-Kit+ cells(green) within the aorta region of E10.5 embryos. The −3.9−/− embryos were compared with WT littermates and with +9.5−/− embryos that almost entirely lackc-Kit+ HSCs (25). (Scale bars, 100 μM.) Quantitation of the number of c-Kit+ cells per dorsal aorta (DA) (four embryos each for −3.9+/+ and −3.9−/−; two embryosfor +9.5−/−) (mean ± SEM). (B) Quantitative analysis of Gata2mRNA in E13.5 livers [six litters: −3.9+/+ (n = 12), −3.9+/− (n = 20), −3.9−/− (n = 18)] and brains [fourlitters: −3.9+/+ (n = 9), −3.9+/− (n = 14), −3.9−/− (n = 11)] (mean ± SEM). −RT, no reverse transcriptase. (C) Ter119+ and Lin− populations were sequentiallyisolated from bone marrow via magnetic bead separation. Enrichment of the distinct populations was confirmed in −3.9+/+ bone marrow samples bymeasuring the expression of the lineage-restricted genesMpl (Lin−) and Hbb-b1 (Ter119+). (D) Comparison of Gata2mRNA expression in Lin− and Ter119+ cellsfrom three independent isolations (mean ± SEM). Fluorescence-activated cell sorting of Sca-1+ and c-Kit+ double-positive cells from Lin− cells of −3.9+/+ and−3.9−/− mice (E) and comparison of Gata2 expression in Lin−Sca+Kit+ cells from two independent biological replicates (mean ± SD) (F) are shown. ***P < 0.001(two-tailed unpaired Student t test).

Sanalkumar et al. PNAS | Published online March 10, 2014 | E1093

GEN

ETICS

PNASPL

US

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 4: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

ing differentiated cell types. Gata2 mRNA levels were quantitatedin Lin− progenitors and Ter119+ erythroid cells isolated frombone marrow of −3.9+/+ and −3.9−/− mice. The purity of eachpopulation was confirmed by quantitating expression of lineage-specific markers Mpl and Hbb-b1 (Fig. 2C). Although highlyexpressed in the Lin− population, Gata2 expression levels wereindistinguishable between −3.9+/+ and −3.9−/− mice (Fig. 2D).In Ter119+ cells, Gata2 expression was not greater than in thecontrol lacking reverse transcriptase. Sca-1 and c-Kit double-pos-itive cells were sorted from the Lin− population (Lin−Sca+Kit+)of −3.9+/+ and −3.9−/− bone marrow to enrich for HSPCs (Fig.2E). Gata2 expression in these cells was not influenced signifi-cantly by the −3.9 mutation (Fig. 2F).Despite certain shared molecular attributes of the −3.9 and

+9.5 sites, the conserved GATA motifs of the −3.9 site weredispensable for Gata2 expression in the embryo and adult, steady-state hematopoiesis, and embryogenesis. To elucidate the uniquemolecular underpinnings of the critical +9.5 site activity, wemechanistically compared the four GATA switch sites (−3.9, −2.8,−1.8, and +9.5) that have been functionally analyzed in vivo.

Requirements for Assembly of an Intronic Enhancer ComplexContaining Master Regulators of Hematopoiesis. The −3.9 and +9.5GATA switch sites are DNaseI-hypersensitive in murine eryth-roleukemia cells (Fig. 1B) and E14.5 fetal liver (48) (Fig. 3A),indicative of an open chromatin configuration. As an alternativeapproach to evaluate chromatin accessibility at these sites, weconducted formaldehyde-assisted isolation of regulatory elements(FAIRE) analysis (49). Genome-wide analyses indicated thatFAIRE peaks overlap with multiple open chromatin parameters,and FAIRE can be conducted with fewer cells than conventionalChIP or DNase I hypersensitivity and/or sensitivity assays (50, 51).FAIRE analysis of E12.5 fetal liver cells demonstrated en-

hanced accessibility at the −3.9 and +9.5 sites (Fig. 3B), with theopen chromatin restricted to the GATA switch sites (Fig. 3C).Allele-specific FAIRE analysis was conducted on −3.9+/− and

+9.5+/− E12.5 fetal livers using primers specific for the WT ormutant −3.9 and +9.5 alleles. Chromatin accessibility was con-siderably reduced at the mutant alleles (Fig. 3D). Despite thereduced accessibility resulting from the −3.9 mutation, Gata2expression in −3.9−/− fetal livers was indistinguishable from thatof −3.9+/+ controls (Fig. 2B). Thus, the −3.9 site confers ac-cessibility at this site but is dispensable for Gata2 expression.Because +9.5−/− mice are deficient in fetal liver HSPCs (5), wetook advantage of the intronic location of the +9.5 site to con-duct allele-specific measurements of Gata2 primary transcriptsgenerated from WT and mutant alleles in primary cells from+9.5+/− mice. In E12.5 fetal liver and adult bone marrow,transcription of the mutant allele was substantially reduced (P <0.001) (Fig. 3E), whereas both alleles were expressed equiva-lently in E12.5 fetal brain (Fig. 3E), consistent with our priorfindings (5). In summary, although mutations of the −3.9 and +9.5sites abrogate local chromatin accessibility, this altered molecularattribute is only linked to loss ofGata2 transcription with the +9.5site mutation.Of the Gata2 GATA switch sites analyzed in vivo, only the

+9.5 site contains a conserved E-box in proximity to the GATAmotif. GATA-2, TAL1, and LDB1 ChIP-sequencing analysis inLin− bone marrow (37) revealed the +9.5 site as the sole regionof the Gata2 locus occupied by all of these factors (Fig. 4A).Quantitative ChIP analysis in E12.5 fetal liver revealed TAL1and LDB1 occupancy at the +9.5 site but not at other GATAswitch sites (Fig. 4B). The +9.5 site deletion abrogated TAL1 andLDB1 occupancy (P < 0.001) at the mutant, but not WT, allelesin +9.5+/− fetal liver cells (Fig. 4C). The analyses with primaryfetal liver cells from +9.5+/− mice described above revealed +9.5site-dependent chromatin accessibility and regulatory complexassembly at the Gata2 locus.

Molecular Attributes That Distinguish the +9.5 Site from Other GATASwitch Sites. Because targeted deletion of the −3.9, −2.8, or −1.8site individually did not evoke major biological phenotypes, the

D E

A

CB

-1.2 -0.6 0 +0.6 +1.2

Distance from -3.9 (kb)

+/+

FA

IRE

Sig

nal

(Rel

ativ

e U

nit

s)

0

0.2

0.4

0.6

0.8-3.9

-1.2 -0.6 0 +0.6 +1.20

0.2

0.4

0.6

0.8

Distance from +9.5 (kb)

+/++9.5

cificep

S- elellA

2at aG

stpir cs

nar T) sti

nU evit al e

R(

Fetal Liver

100

1

200+9.5-3.9

-2.8-1.8

Seq

uen

ce R

ead

s

+9.5-2.8 -1.8-3.9

1.5

1.0

0.5

0

RPII215

Krt5

FAIR

E S

ign

al

(Rel

ativ

e U

nit

s)

+/+

Alle

le-S

pec

ific

FA

IRE

Sig

nal

(R

elat

ive

Un

its)

MtWT MtWT0

0.2

0.4

0.6

***

***

Fetal Liver

*

WT WTMt Mt MtWT0

0.4

0.6

0.8

0.2

***

+/+ +/-+9.5

-RT1 Transcripts

0

0.15

0.30

0.45

Bone Marrow

**

****

**

**

Fetal Brain

WT WTMt Mt MtWT-RT

+/+ +/-+9.5

WT WTMt Mt MtWT0

0.4

0.6

0.8

0.2

+/+ +/-+9.5

-RT

+/-+9.5+/--3.9

DNase HS

o 1 Transcriptso 1 Transcriptso

2 kb1S

1G

Primers:Primers:

Fig. 3. GATA switch site mutations abrogate chromatin accessibility at −3.9 and +9.5 sites. (A) DNaseI hypersensitivity at the Gata2 locus in fetal liver mined frommouse Encyclopedia of DNA Elements data (48). (B) Quantitative FAIRE analysis of GATA switch sites in WT fetal liver (n = 4, mean ± SEM). The promoters of theactively transcribed RNA Polymerase II (RPII215) and inactive Keratin 5 (Krt5) genes were used as positive and negative controls, respectively. (C) Quantitative FAIREanalysis of chromatin accessibility at and surrounding the −3.9 and +9.5 sites (n = 4, mean ± SEM). The dashed line illustrates the average FAIRE signal at the Krt5promoter. (D) Allele-specific FAIRE analysis of WT and mutated (Mt) alleles in fetal liver cells from −3.9+/− (n = 4) and +9.5+/− (n = 5) E13.5 embryos (mean ± SEM).Primers used for the allele-specific FAIRE analysis are indicated in Table S3. (E) Allele-specific analysis of Gata2 primary transcripts fromWT and Mt alleles in +9.5+/−

E13.5 fetal liver and brain (n = 8) and adult bone marrow (n = 3) samples (mean ± SEM). *P < 0.05; **P < 0.01; ***P < 0.001 (two-tailed unpaired Student t test).

E1094 | www.pnas.org/cgi/doi/10.1073/pnas.1400065111 Sanalkumar et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 5: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

+9.5 element uniquely endows hemogenic endothelium of theAGM with the capacity to generate long-term repopulatingHSCs (LT-HSCs) that populate the fetal liver (5, 25). In addi-tion, the small number of HSCs generated from +9.5−/− AGMundergo apoptosis and lack LT activity, indicating that the +9.5element also confers LT-HSC survival (25); a similar conclusionemerged from analysis of a conditional Gata2 KO mouse (52).The +9.5 site confers maximal Gata2 expression in the AGMand definitive hematopoietic precursors in the fetal liver (5, 25).A critical question is what molecular attributes underlie theuniquely important +9.5 site activity.To identify molecular attributes that distinguish the +9.5

site from the other GATA switch sites, we developed a Gata2repression/reactivation system using GATA-1–null G1E pro-erythroblast-like cells (53). G1E cells express GATA-2 (53), andectopic expression of GATA-1 represses GATA-2 and overcomesan erythroid maturation blockade (10, 54). Using a conditionallyactive GATA-1 allele, in which GATA-1 is fused to the estrogenreceptor ligand binding domain (ER–GATA-1), β-estradiol treat-ment of G1E–ER–GATA-1 cells rapidly induces Gata2 repression(10). We reasoned that removing β-estradiol from the culturemedia subsequent to Gata2 repression would induce time-dependent loss of ER–GATA-1 activity, ER–GATA-1 dissocia-tion from chromatin, and reversion of ER–GATA-1 influenceson gene expression (Fig. 5A). β-Estradiol treatment of G1E–ER–

GATA-1 cells for 24 h strongly reduces Gata2 mRNA andprotein (10). β-Estradiol washout after 24 h induced reactivation

of Gata2 primary transcripts (Fig. 5B) and GATA-2 protein(Fig. 5C), concomitant with reduced ER–GATA-1 levels (Fig. 5C).Because we are unaware of a system that allows one to study theconversion of the repressed Gata2 locus to an active locus, thissystem has unique utility for elucidating GATA switch sitefunction.GATA-1–mediated Gata2 repression is associated with GATA-1

replacement of GATA-2 at GATA switch sites (10). To inves-tigate the impact of the Gata2 reactivation on the GATA switch,ChIP analysis was used to quantitate changes in ER–GATA-1and GATA-2 occupancy during Gata2 repression and reacti-vation (Fig. 5D). As expected (24), 24 h of β-estradiol treatmentinduced replacement of GATA-2 by ER–GATA-1 at all sitestested, although ER–GATA-1 occupancy of the −1.8 site was low,consistent with our prior findings. Surprisingly, 24 h after washingout β-estradiol, when GATA-2 is readily detected by Westernblotting, GATA-2 occupancy was only restored at the +9.5 and−3.9 sites (54% and 57% reoccupancy, respectively), despite lossof GATA-1 from all sites (Fig. 5D).Given that GATA-2 occupies all of the Gata2 GATA switch

sites before repression but only reoccupies +9.5 and −3.9 sitesfollowing Gata2 reactivation, we tested whether ER–GATA-1–induced repression creates inaccessible chromatin at the −1.8and −2.8 sites that persists after ER–GATA-1 dissociation fromchromatin and prevents subsequent GATA-2 occupancy. We usedquantitative FAIRE to analyze chromatin accessibility in theGATA-2 repression/reactivation system. In uninduced proliferating

0.10

0.15

0.20

0

0.05

Alle

le-S

pec

ific

TA

L1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

C***

A

B

0.02

0.04

0.06

0

TA

L1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

2 kb 1S1G

5.9+8.1-8.2-9.3-

LDB1

TAL1

GATA-220

430

318

3S

equ

ence

Rea

ds

PI TAL1

0.004

0.008

0.012

0

LD

B1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

-3.9 -2.8 -1.8 +9.5

Necdin

PI LDB1

PI TAL1

0.01

0.02

0.03

Alle

le-S

pec

ific

LD

B1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

WT Mt0

***

PI LDB1

***

***

Fig. 4. Unique propensity of TAL1 and LDB1 to occupy the +9.5 site. (A) ChIP-sequencing profiles of factor occupancy at the Gata2 locus in lineage-negativebone marrow cells mined from existing datasets (37). (B) Quantitative ChIP analysis of TAL1 and LDB1 occupancy at GATA switch sites of the Gata2 locus inE13.5 fetal liver (n = 3, mean ± SEM). ***P < 0.001. The Necdin promoter was used as a negative control. (C) Allele-specific ChIP analysis of TAL1 and LDB1occupancy at WT and Mt alleles in fetal liver from E13.5 +9.5+/− embryos (n = 4, mean ± SEM). ***P < 0.001 (two-tailed unpaired Student t test). PI,preimmune.

Sanalkumar et al. PNAS | Published online March 10, 2014 | E1095

GEN

ETICS

PNASPL

US

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 6: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

G1E-ER-GATA-1 cells expressing Gata2, chromatin accessibil-ity of the GATA switch sites was significantly greater than in theinactive Necdin promoter (Fig. 6A). ER–GATA-1–mediatedGATA-2 displacement reduced FAIRE signals at GATA switchsites 2.8- to 14-fold (Fig. 6B, 24 h). Upon Gata2 reactivation,chromatin accessibility was restored (108% of 0 h) only at the+9.5 site (Fig. 6B, 48 h; P < 0.001); accessibility of −1.8, −2.8,and −3.9 sites remained low (Fig. 6B, 48 h). The unique tripartitechromatin signature of the +9.5 site, in which accessibility is lostupon repression and restored upon reactivation, reflects +9.5site activity to mediate dynamic chromatin transitions duringGata2 activation. Because this tripartite chromatin signaturedistinguishes the +9.5 site from other switch sites, we evaluatedmechanisms underlying this unique behavior.In principle, certain factors might be selectively retained at the

+9.5 site, thus explaining its unique capacity to reestablish ac-cessible chromatin. Based on the +9.5 site-restricted TAL1 andLDB1 occupancy in fetal liver (Fig. 4B), we asked whether TAL1and LDB1 occupancy uniquely characterizes the +9.5 site. Usingthe repression/reactivation system, we quantitated TAL1 andLDB1 occupancy at the active (0 h, uninduced), repressed (24 h,estradiol-induced), and reactivated (48 h, washout) Gata2 locus.Twenty-four hours after β-estradiol treatment, when GATA-2

protein is undetectable, TAL1 and LDB1 levels were unchangedor increased slightly (Fig. 6C). Both factors occupied the −2.8,−1.8, and +9.5 sites of the active Gata2 locus (Fig. 6D). UponGata2 repression, TAL1 and LDB1 occupancy decreased at the−2.8 and −1.8 sites but was partially retained at the +9.5 site.Brahma related gene 1 (BRG1) occupied the active and re-pressed loci (Fig. 6E). The TAL1 and LDB1 levels retained atthe +9.5 site were comparable to the highly active βmajor pro-moter and were considerably higher (P < 0.001) than those at the−1.8 and −2.8 sites (Fig. 6D); ER–GATA-1 is known to increaseTAL1 recruitment to the βmajor promoter (40). It is attractive topropose that TAL1/LDB1/BRG1 retention at the +9.5 site ofthe repressed Gata2 locus reflects a priming mechanism thatcreates epigenetic memory to ensure a rapid increase in thechromatin accessibility and factor occupancy required for sub-sequent locus reactivation.

Establishing and Maintaining Physiological GATA-2 Levels: DualRequirement for a Chromatin Looping Factor and a ChromatinRemodeler. Of the −3.9, −2.8, −1.8, and +9.5 GATA switchsites analyzed in mutant mouse strains, TAL1/LDB1 chromatinoccupancy is unique to the +9.5 site. We conducted loss-of-function analyses to establish the importance of the +9.5 site-occupied components. Regarding the possibility of priming the+9.5 site to ensure rapid chromatin remodeling as a requisitestep in reactivation, because TAL1 and GATA-1 can localize tochromatin sites with and interact functionally with the ATPasecomponent (BRG1) of the SWI/SNF chromatin remodelingcomplex (55–57), BRG1 might mediate chromatin remodeling atthe +9.5 site. To assess the contributions of LDB1 and BRG1 tomaintenance of Gata2 expression, uninduced G1E–ER–GATA-1cells were treated twice with siRNA and harvested 24 h after thesecond treatment (Fig. 7A). When knocked down individually,loss of LDB1 or BRG1 did not influence GATA-2 levels (Fig.7B), whereas simultaneous loss of LDB1 and BRG1 substantiallyreduced GATA-2. LDB1 and BRG1 activity to establish Gata2expression was assessed with the reactivation system (Fig. 7C).Although knocking down LDB1 or BRG1 individually did notprevent Gata2 reactivation, GATA-2 levels were partially re-duced (Fig. 7D, Left and Fig. S1). Knocking down LDB1 didnot affect GATA-2 chromatin occupancy (Fig. S2). Resemblingthe results with uninduced cells, knocking down LDB1 andBRG1 simultaneously greatly reduced reactivation (Fig. 7D,Right). Thus, LDB1 and BRG1 establish and maintain GATA-2expression.In uninduced cells and reactivation contexts, the LDB1 knock-

down decreased TAL1 levels without a concomitant change inGATA-2 levels (Fig. 7B). This result is consistent with our priorTAL1 knockdown in G1E–ER–GATA-1 cells, which did notalter GATA-2 levels (58). LDB1 occupies the TAL1 locus (37)(Fig. S3), and therefore might directly regulate TAL1 expression.Although individually knocking down LDB1 prevented the res-toration of TAL1 occupancy at the +9.5 site to a level com-mensurate with the active Gata2 locus (Fig. 7E), under theseconditions, the washout still restored chromatin accessibility(Fig. 7F). TAL1 occupancy (Fig. 7E) and chromatin accessibility(Fig. 7F) decreased at the −1.8 site upon repression and were notrestored upon Gata2 reactivation. Unlike the individual factorknockdowns, the LDB1/BRG1 double knockdown, which blockedGata2 reactivation, prevented the washout-induced restoration ofopen chromatin at the +9.5 site (Fig. 7E). Because the washoutdecreased GATA-1 occupancy at all Gata2 GATA switch sites(Fig. 7G), the inability to reactivateGata2 when LDB1 and BRG1levels are limiting cannot be explained by GATA-1 retention. Theseresults suggest that the failure to establish an active enhancercomplex at the +9.5 site underlies the reactivation defect.

D

GATA-2

ER-GATA-1

-actin

0

Time (h)

A

B

CG

ata2

Pri

mar

yT

ran

scri

pts

0

0.8

0.4

1.2

12 24 36 48

Time (h)0 24 26 30 36 48

Mr x

10

-3

52

38

76

102

38

GA

TA-2

/ER

-GA

TA-1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

)h(emiT)h(emiT

0.04

0.08

0.12

0

0.12

0.24

0.36

0

0.06

0.12

0.18 0.18

0.06

0.12

0 12 24 36 48 0 12 24 36 480

0

GATA-2 ER-GATA-1

0 24 26 30 36 48

Wash+Estradiol Wash out

+Estradiol Wash out

+Estradiol Wash out +Estradiol Wash out

-3.9 -2.8

-1.8 +9.5

Fig. 5. Gata2 repression/reactivation assay. Evidence for distinct functionalproperties of the GATA switch sites is illustrated. (A) Schematic representa-tion of the experimental strategy for Gata2 repression and reactivation inG1E–ER–GATA-1 cells. Treatment of G1E–ER–GATA-1 cells with β-estradiolactivates ER–GATA-1, leading to loss of Gata2 transcripts and protein by 24 h(10). Washout of β-estradiol reverses Gata2 repression, leading to restorationof GATA-2 by 48-h treatment. (B) Quantitative real-time PCR was used tomeasure Gata2 primary transcripts during locus reactivation. After 24 h,β-estradiol treatment (+estradiol) cells were washed in PBS and cultured inmedia without β-estradiol (washout) for an additional 24 h. RNA was iso-lated and analyzed before β-estradiol treatment (0 h); after 24 h of β-es-tradiol treatment; and 2, 6, 12, and 24 h following washout (n = 4, mean ±SEM). (C) Representative Western blots of GATA-2 and ER–GATA-1 fromsamples isolated at the same times as the corresponding RNA samples. (D)Relative chromatin occupancy of ER-GATA-1 (○) and GATA-2 (●) duringGata2 reactivation using quantitative ChIP (n = 3, mean ± SEM).

E1096 | www.pnas.org/cgi/doi/10.1073/pnas.1400065111 Sanalkumar et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 7: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

DiscussionHow to distill large chromatin occupancy datasets into functionallycritical genomic sites, especially sites distal to genes, represents aformidable problem with far-reaching implications. Our mousestrains lacking GATA switch sites differ grossly in phenotypesand offer a unique opportunity to elucidate mechanisms thatendow GATA factor-bound chromatin sites with nonredundantactivity in vivo. Despite certain shared molecular attributes ofthe −3.9 and +9.5 sites, the conserved GATA motifs of the −3.9site were dispensable for Gata2 expression in the embryo or adult,in steady-state hematopoiesis, and in embryogenesis.Because the coupling of the E-box and GATA motif and

intronic location distinguish the +9.5 site from the −3.9, −2.8,and −1.8 sites, it is attractive to propose that these attributes areimportant determinants of +9.5 site activity in vivo. In Gata2-expressing fetal liver cells, the +9.5 site resided in open chro-matin and assembled a complex containing GATA-2, TAL1, andLDB1 (Fig. 7I). Simultaneous interrogation of the chromatinaccessibility of WT and mutant alleles in +9.5+/− fetal liver cellsand analyses with the repression/reactivation system revealedthat the +9.5 site mediates dynamic chromatin structure tran-sitions. Although ER–GATA-1 converted accessible chromatinat the +9.5 site and other Gata2 GATA switch sites into in-accessible chromatin, concomitant with repression, the washout-induced loss of GATA-1 activity selectively converted +9.5 site-inaccessible chromatin into accessible chromatin. Intriguingly,TAL1 and LDB1 were partially retained at the +9.5 site but notat other Gata2 GATA switch sites, and upon reactivation, chro-matin accessibility was only restored at the +9.5 site. These resultssuggest that TAL1/LDB1 retention creates an epigenetic memorythat ensures reassembly of the functional +9.5 site enhancer and

Gata2 transcriptional activation. This mechanism may be con-ceptually similar to the findings that target gene occupancy bycertain transcription factors is retained in mitotic chromatin andis associated with more rapid transcriptional activation upon entryinto G1 (59, 60).GATA-1 and GATA-2 occupy the TAL1 locus and loci-encoding

components of the TAL1 complex (e.g., the corepressor ETO2)(61, 62). Herein, we demonstrate that TAL1 protein expressionis sensitive to LDB1 protein levels. The relationship betweenLDB1, TAL1, and GATA-2 can be modeled as a coherent type Ifeed-forward loop (Fig. 7H), which is predicted to require per-sistently elevated input signals (e.g., those controlling the LDB1level/activity), although filtering out transiently elevated inputsignals, to achieve a robust output (e.g., enhanced HSC genera-tion) (63). LDB1 and BRG1 synergistically confer +9.5 site-accessible chromatin, enhancer activity, and Gata2 expression.Although BRG1 was reported to interact functionally with TAL1(32), we are unaware of examples of a mechanism requiring bothBRG1 and LDB1. The BRG1 activity may have broad implica-tions in diverse contexts, because BRG1 is required for Pax6-dependent control of neural fate (38), for Olig2 to establish anoligodendrocyte-specific transcriptional program (64), and forcardiovascular development (65, 66).In summary, we describe a mechanism that endows a stem

cell-generating enhancer element with its unique activity anddifferentiates it from other GATA factor-bound chromosomalsites that we have rigorously analyzed in vivo. Intrinsic to thismechanism is synergism between a chromatin looping factor anda chromatin remodeler to generate physiological levels of themaster hematopoietic regulator GATA-2 (Fig. 7I). In the contextof hematopoiesis, it will be particularly instructive to consider

la

ngi

SE

RIA

F)

stin

Ue

vital

eR(

1.0

0.5

0

1.5

0 12 24 36 480 12 24 36 48

Time (h)

1.0

0.5

0

1.5

2.0

-3.9

-2.8

-1.8+9

.5

Necdin

A B +Estradiol Wash out

-3.9 -2.8 +9.5+Estradiol Wash out

-1.8+Estradiol Wash out +Estradiol Wash out

0 12 24 36 480 12 24 36 48

D

TAL

1 O

ccu

pan

cy(R

elat

ive

Un

its)

LD

B1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

24 WO0

GATA-2

TAL1

LDB1

-actin

C

50

52

52

38

Mr x

10

-3

0 24 WO

*

-estradiol (h)24 WO0 24 WO0 24 WO0 24 WO0

***

***

***

***

la

ngi

SE

RIA

F)

stin

Ue

vital

eR(

0.015

0.010

0.005

0

***

** **

0.20

0.10

0

0.30-3.9 -2.8 +9.5-1.8 major

-3.9 -2.8 +9.5-1.8 major

TAL1PI

***

**

******

***

LDB1IgG

0 24

0.15

0.10

0.05

0BR

G1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

major +9.5 -1.8 Necdin

***

0 24 0 24 0 24

E BRG1PI

Fig. 6. Molecular attributes of the +9.5 site revealed by the repression/reactivation assay. (A) Quantitative FAIRE analysis of chromatin accessibility at GATAswitch sites in untreated G1E-ER-GATA-1 cells (0 h). The inactive Necdin locus was used as a negative control (n = 4, mean ± SEM). (B) Quantitative FAIREanalysis of chromatin accessibility at GATA switch sites upon Gata2 repression and reactivation. FAIRE signals for the 0-h times were normalized to 1.0 (n = 4,mean ± SEM). (C) Representative Western blots of GATA-2, TAL1, and LDB1 protein levels at the same times analyzed by ChIP. The asterisk representsa nonspecific band. (D) Quantitative ChIP analysis of TAL1 and LDB1 chromatin occupancy at the active (0 h), repressed (+24 h), and reactivated (WO) Gata2locus. The βmajor promoter was used as a positive control (n = 4, mean ± SEM). **P < 0.01; ***P < 0.001. (E) Quantitative ChIP analysis of BRG1 chromatinoccupancy at the active (0 h) and repressed (+24 h) Gata2 locus and control sites (βmajor and Necdin) (n = 3, mean ± SEM). ***P < 0.001. WO, washout.

Sanalkumar et al. PNAS | Published online March 10, 2014 | E1097

GEN

ETICS

PNASPL

US

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 8: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

whether this mechanism is quite selective for controlling GATA-2and the GATA-2–dependent genetic network or if it has animpact, more broadly, on the HSPC transcriptome.

Materials and MethodsGeneration of Gata2 Δ-3.9 Mutant Mice. The Gata2 −3.9 site sequence AGA-TAGGAAAATGGCCGCGCGCTATCT containing the inverted WGATAR motifswas replaced with a LoxP-phosphoglycerate kinase neomycin (neo)-LoxPcassette via homologous recombination. Targeting was confirmed bySouthern blotting. Chimeric mice were generated by blastocyst injection,and first filial generation pups were screened for germ-line transmission byPCR. NeoR excision was achieved by mating to CMV-cre strain B6.C-Tg(CMV-

cre)1Cgn/J mice (Jackson Laboratory). Cre-mediated excision of NeoR in theprogeny was confirmed by PCR using primers flanking the targeted se-quence. Primer sequences are provided in Table S3.

Analysis of Mouse Embryos and Tissues. Staged embryos were obtained fromtimed matings of Gata2 −3.9 heterozygotes. Embryo viability was scored bythe presence of a beating heart. E13.5 fetal livers and brains were harvestedinto TRIzol (Invitrogen) for RNA extraction. For CBC analysis, blood samplesfrom 33 anesthetized mice (four litters: 10 −3.9+/+, 14 −3.9+/−, and 9 −3.9−/−

mice) were collected by retroorbital bleeding at 2 and 6 mo of age. CBC mea-surements were collected using a Hemavet CBC Analyzer (Drew Scientific, Inc.).Bone marrow was isolated from femurs and tibias into PBS containing

FA

IRE

Sig

nal

(Rel

ativ

e U

nit

s)

TA

L1

Occ

up

ancy

(Rel

ativ

e U

nit

s)

E F ***

0.6

0.4

0.2

0- ++ + +- - -

-+

-- -

wo wo

+

G

GA

TA

-1 O

ccu

pan

cy(R

elat

ive

Un

its)

- ++ + + -- - - +

--

-- - -

wo wo wo - ++ + + -- - - +

-

+-

-- - -

wo wo wo

+9.5 -1.8 +9.5 -1.8

+

+-

-

++

-

wo wo+-

-

++

-

wo wo

-3.9 -2.8 -1.8*** ***

*** ***

2.4

1.6

0

PI GATA-1

0

0.05

0.10

0.15

0.20

**

***

+9.5

*** **

0.8

***

**

**

Estradiol - ++ + + -- - - +

--

-- - -

wo wo wo - ++ + + -- - - +

-

+-

-- - -

wo wo wo

+9.5 -1.8 +9.5 -1.8+

+-

-

++

-

wo wo+-

-

++

-

wo wo

Estradiol - ++ + + -- - - +

-

+-

-- - -

wo wo wo

50

52

52

38

260

C

wo wo

GATA-2

TAL1

LDB1

BRG1

-actin

*

+-

-+

- +

D

siRNA Wash & siRNA tsevraHloidartsE

h84h42h6h0

A

--

---

-

GATA-2

-actin

siRNAsiRNA Harvest

h84h42h0

*

BTAL1

LDB1

BRG1

Control siRNALdb1 siRNABrg1 siRNA

+

+

+

--

-+

++

50

52

52

38

260Mr x

10

-3

- ++ + +- - -

-+

-- -

wo wo

+

- ++ + +- - -

-+

-- -

wo wo

+

- ++ + +- - -

-+

-- -

wo wo

+

I

+9.5

Gata2

Wild type

-3.9

+9.5

+9.5 Mutant

-3.9 +9.5

Gata2

-3.9 Mutant

-3.9

LDB1

TAL1

GATA-2

Type 1 Coherent FeedForward Loop

H

Mr x

10

-3

Control siRNALdb1 siRNABrg1 siRNA

Control siRNALdb1 siRNABrg1 siRNA

EstradiolControl siRNA

Ldb1 siRNABrg1 siRNA

EstradiolControl siRNA

Ldb1 siRNABrg1 siRNA

Fig. 7. Mechanism underlying +9.5 site function. (A) Knockdown strategy. Cells were transfected with siRNA twice with a 24-h interval and harvested 48 hafter the first transfection. (B) Representative Western blots of GATA-2, TAL1, LDB1, and BRG1 following knockdown of Ldb1 and Brg1 mRNAs individually orin combination. The asterisk represents a nonspecific band. Schematic representation (C) and Western blots (D) of siRNA-mediated factor knockdown strategyduring Gata2 repression/reactivation. G1E–ER–GATA-1 cells were induced with β-estradiol (0 h) and transfected twice with factor-specific or control siRNAs at6 and 24 h postinduction. β-estradiol was washed out at the time of the second transfection. Protein and RNA samples were collected at 24 and 48 h. −,β-estradiol uninduced at 0 h; +, β-estradiol induced at 24 h; WO, β-estradiol washout. All conditions received specific siRNA or nontargeting control siRNA atsame molar concentration. (E) ChIP analysis of TAL1 occupancy at the +9.5 and −1.8 sites quantitated under individual knockdown or LDB1 and BRG1combined knockdown conditions (n = 4, mean ± SEM). **P < 0.01; ***P < 0.001. (F) FAIRE analysis of chromatin accessibility of +9.5 and −1.8 sites followingLDB1 and BRG1 individual or LDB1/BRG1 combined knockdown (n = 3, mean ± SEM). ***P < 0.001. (G) ChIP analysis of GATA-1 occupancy at GATA switch sitesfollowing LDB1/BRG1 combined knockdown during GATA-2 reactivation (n = 4, mean ± SEM). **P < 0.01; ***P < 0.001. (H) Type I coherent feed-forward loopnetwork motif that controls Gata2 expression. (I) Model depicts GATA switch-site chromatin architecture and its relationship to Gata2 expression. The red Xindicates that the motif has been deleted.

E1098 | www.pnas.org/cgi/doi/10.1073/pnas.1400065111 Sanalkumar et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 9: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

2% (vol/vol) FBS and 2 mM EDTA (wash buffer). Dissociated cells werepelleted at 300 × g for 10 min and resuspended in wash buffer at 1 × 108

cells per milliliter. Ter119+ cells were isolated by magnetic bead isolationusing rat anti-mouse Ter-119 Biotin (eBioscience). The Ter-119–depletedpopulation was depleted for additional lineage markers using the EasySepMouse Hematopoietic Progenitor Cell Enrichment Kit (Stemcell Technol-ogies). For isolation of Lin−Sca+Kit+ cells, Sca-1 and c-Kit double-positivecells were sorted from Lin− cells on a FACSAria II cell sorter (BD Biosciences)using rat anti-mouse CD117 allophycocyanin and anti-mouse Ly-6A/E perdi-dine chlorophyll protein-Cyaninine5.5 (eBioscience). Data were analyzed usingFlowJo v9.0.2 software (TreeStar).

Whole-Embryo Confocal Microscopy. Embryos were fixed, stained, and ana-lyzed as described (25, 67). Briefly, E10.5 embryos were stained for c-Kit usingrat anti-mouse c-Kit (BD Biosciences) and Alexa Fluor 647 goat anti-rat IgG(Invitrogen) and then for PECAM1 using biotinylated rat anti-mouse CD31(BD Biosciences) and Cy3-conjugated streptavidin (Jackson ImmunoResearch).Samples were cleared in a 1:2 mix of benzyl alcohol and benzyl benzoate toincrease transparency before imaging with a Nikon A1R Confocal Microscope.Three-dimensional reconstructions were generated from Z-stacks (50–150 op-tical sections) using Fiji software.

Cell Culture. G1E-ER-GATA-1 cells (10, 54) were maintained in Iscove’s mod-ified Dulbecco’s medium (GIBCO) supplemented with 15% FBS (Gemini), 1%penicillin/streptomycin (Gemini), 2 U/mL erythropoietin, 120 nM mono-thioglycerol (Sigma), 0.6% conditioned medium from a Kit ligand-producingCHO cell line, and 1 μg/mL puromycin (Sigma). To induce ER–GATA-1, cellswere treated with 1 μM β-estradiol. For Gata2 reactivation studies, cells wereinduced with β-estradiol for 24 h and then washed with 1× Dulbecco’sphosphate buffered saline to remove β-estradiol. Cells were grown in mediawithout β-estradiol for an additional 24 h, and samples were collected 2, 6,12, and 24 h after washout. Cell cultures were maintained in a 37 °C incubatorwith 5% CO2. siRNA-mediated genetic perturbation was used to knock downfactors in G1E-ER-GATA-1 cells. Specific SMART Pool siRNAs or nontargetingsiRNA pools (240 pmol each; Dharmacon) were transfected into cells usingAmaxa nucleofection kit R. For knockdown analyses in uninduced cells, cellswere transfected twice with a 24-h interval. For analyses in the reactivationparadigm, two transfections were conducted 6 and 24 h after β-estradiolinduction. Samples were harvested at 24 h and/or 48 h.

Quantitative Real-Time PCR Analysis. Total RNA was purified with TRIzol.cDNA was synthesized from 1.5 μg of purified total RNA by Moloney MLV

reverse transcriptase (M-MLV RT). Real-time PCR analysis was conducted withSYBR Green Master Mix (Applied Biosystems). Control reactions lacking M-MLV RT yielded little to no signal. Relative expression was determined froma standard curve of serial dilutions of cDNA samples, and values were nor-malized to 18S RNA expression. Primer sequences are provided in Table S3.

Quantitative FAIRE Assay. FAIRE analysis was conducted as described (50) withminor modifications. Cells were fixed with 1% formaldehyde for 5 min atroom temperature and sonicated to shear the DNA to an average size of200–700 bp. Ten percent of the sonicated chromatin was used as the inputcontrol. Following phenol/chloroform extraction, ethanol-precipitated DNApellets were resuspended in 50 μL of nuclease-free water. For FAIRE analysisof fetal livers, −3.9+/− and +9.5+/− embryos from timed matings were col-lected at E13.5 into ice-cold PBS. Livers were removed and dissociated bypipetting before formaldehyde fixation. An allele-specific quantitative PCRassay was conducted with allele-specific primers that distinguish WT andmutant alleles. Serial dilutions of input samples were used for generatinga standard curve, and relative FAIRE signals were calculated for specific sites.

Western Blot Analysis. Whole-cell lysates were prepared by boiling 1 × 107

cells per milliliter in SDS sample buffer [25 mM Tris (pH 6.8), 2% β-mercap-toethanol, 3% SDS, 0.1% bromophenol blue, 5% glycerol] for 10 min.Samples (10 μL) were resolved by SDS/PAGE and analyzed with specificantibodies. Rabbit anti–GATA-2 (68) and rabbit anti-TAL1 (11) were de-scribed previously. Rat anti–GATA-1 (N-6, sc-265), rabbit anti-BRG1 (H-88,sc10768), and goat anti-LDB1 (N18, sc11198) were from Santa Cruz Bio-technology, and mouse anti–β-actin (3700S) was from Cell Signaling.

Quantitative ChIP Assay.A quantitative ChIP assay was conducted as describedpreviously (8). Cells cross-linked with 1% formaldehyde were sonicated toyield DNA with an average size of 200–700 bp and immunoprecipitated withspecific antibodies [rabbit anti–GATA-1 (68), rabbit anti–GATA-2 (68), andrabbit anti-TAL1 (11)], LDB1 (N18, sc11198), and BRG1 (ab110641). For BRG1ChIP, cells were cross-linked with 1% formaldehyde for 20 min. ChIP sampleswere quantitated relative to the input DNA using real-time PCR analysis.Rabbit preimmune serum or normal IgG was used as a negative control.

ACKNOWLEDGMENTS. This work was supported by Grant R01 DK68634from the National Institutes of Health (NIH) (to E.H.B.), a University ofWisconsin–Madison Stem Cell and Regenerative Medicine Center postdoc-toral fellowship (to R.S.), and an NIH T32 Hematology Training Grant Award(to K.J.H.).

1. Bender MA, Bulger M, Close J, Groudine M (2000) Beta-globin gene switching andDNase I sensitivity of the endogenous beta-globin locus in mice do not require thelocus control region. Mol Cell 5(2):387–393.

2. Krivega I, Dean A (2012) Enhancer and promoter interactions-long distance calls. CurrOpin Genet Dev 22(2):79–85.

3. Harmston N, Lenhard B (2013) Chromatin and epigenetic features of long-range generegulation. Nucleic Acids Res 41(15):7185–7199.

4. Lee HY, Johnson KD, Boyer ME, Bresnick EH (2011) Relocalizing genetic loci intospecific subnuclear neighborhoods. J Biol Chem 286(21):18834–18844.

5. Johnson KD, et al. (2012) Cis-element mutated in GATA2-dependent immunodefi-ciency governs hematopoiesis and vascular integrity. J Clin Invest 122(10):3692–3704.

6. Snow JW, et al. (2010) A single cis element maintains repression of the key de-velopmental regulator Gata2. PLoS Genet 6(9):e1001103.

7. Snow JW, et al. (2011) Context-dependent function of “GATA switch” sites in vivo.Blood 117(18):4769–4772.

8. Grass JA, et al. (2006) Distinct functions of dispersed GATA factor complexes at anendogenous gene locus. Mol Cell Biol 26(19):7056–7067.

9. Wozniak RJ, Boyer ME, Grass JA, Lee Y, Bresnick EH (2007) Context-dependent GATAfactor function: Combinatorial requirements for transcriptional control in hemato-poietic and endothelial cells. J Biol Chem 282(19):14665–14674.

10. Grass JA, et al. (2003) GATA-1-dependent transcriptional repression of GATA-2 viadisruption of positive autoregulation and domain-wide chromatin remodeling. ProcNatl Acad Sci USA 100(15):8811–8816.

11. Wozniak RJ, et al. (2008) Molecular hallmarks of endogenous chromatin complexescontaining master regulators of hematopoiesis. Mol Cell Biol 28(21):6681–6694.

12. Martowicz ML, Grass JA, Boyer ME, Guend H, Bresnick EH (2005) Dynamic GATAfactor interplay at a multicomponent regulatory region of the GATA-2 locus. J BiolChem 280(3):1724–1732.

13. Tsai FY, et al. (1994) An early haematopoietic defect in mice lacking the transcriptionfactor GATA-2. Nature 371(6494):221–226.

14. Minegishi N, et al. (1999) The mouse GATA-2 gene is expressed in the para-aorticsplanchnopleura and aorta-gonads and mesonephros region. Blood 93(12):4196–4207.

15. Tsai FY, Orkin SH (1997) Transcription factor GATA-2 is required for proliferation/survival of early hematopoietic cells and mast cell formation, but not for erythroidand myeloid terminal differentiation. Blood 89(10):3636–3643.

16. Dorfman DM, Wilson DB, Bruns GA, Orkin SH (1992) Human transcription factorGATA-2. Evidence for regulation of preproendothelin-1 gene expression in endo-thelial cells. J Biol Chem 267(2):1279–1285.

17. Nardelli J, Thiesson D, Fujiwara Y, Tsai FY, Orkin SH (1999) Expression and geneticinteraction of transcription factors GATA-2 and GATA-3 during development of themouse central nervous system. Dev Biol 210(2):305–321.

18. Dasen JS, et al. (1999) Reciprocal interactions of Pit1 and GATA2 mediate signalinggradient-induced determination of pituitary cell types. Cell 97(5):587–598.

19. Rodrigues NP, et al. (2005) Haploinsufficiency of GATA-2 perturbs adult hematopoi-etic stem-cell homeostasis. Blood 106(2):477–484.

20. Ling KW, et al. (2004) GATA-2 plays two functionally distinct roles during the on-togeny of hematopoietic stem cells. J Exp Med 200(7):871–882.

21. Hsu AP, et al. (2011) Mutations in GATA2 are associated with the autosomal dominantand sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome.Blood 118(10):2653–2655.

22. Mansour S, et al.; Lymphoedema Research Consortium (2010) Emberger syndrome-primary lymphedema with myelodysplasia: Report of seven new cases. Am J MedGenet A 152A(9):2287–2296.

23. Hahn CN, et al. (2011) Heritable GATA2 mutations associated with familial myelo-dysplastic syndrome and acute myeloid leukemia. Nat Genet 43(10):1012–1017.

24. Bresnick EH, Lee HY, Fujiwara T, Johnson KD, Keles S (2010) GATA switches as de-velopmental drivers. J Biol Chem 285(41):31087–31093.

25. Gao X, et al. (2013) Gata2 cis-element is required for hematopoietic stem cell gen-eration in the mammalian embryo. J Exp Med 210(13):2833–2842.

26. Lim KC, et al. (2012) Conditional Gata2 inactivation results in HSC loss and lymphaticmispatterning. J Clin Invest 122(10):3705–3717.

27. Tripic T, et al. (2009) SCL and associated proteins distinguish active from repressiveGATA transcription factor complexes. Blood 113(10):2191–2201.

28. Porcher C, et al. (1996) The T cell leukemia oncoprotein SCL/tal-1 is essential for de-velopment of all hematopoietic lineages. Cell 86(1):47–57.

29. Wadman IA, et al. (1997) The LIM-only protein Lmo2 is a bridging molecule assem-bling an erythroid, DNA-binding complex which includes the TAL1, E47, GATA-1 andLdb1/NLI proteins. EMBO J 16(11):3145–3157.

30. Lécuyer E, et al. (2002) The SCL complex regulates c-kit expression in hematopoieticcells through functional interaction with Sp1. Blood 100(7):2430–2440.

Sanalkumar et al. PNAS | Published online March 10, 2014 | E1099

GEN

ETICS

PNASPL

US

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0

Page 10: Mechanism governing a stem cell-generating cis-regulatory ... · Mechanism governing a stem cell-generating cis-regulatory element Rajendran Sanalkumara,b, Kirby D. Johnsona,b, Xin

31. Lahlil R, Lécuyer E, Herblot S, Hoang T (2004) SCL assembles a multifactorial complexthat determines glycophorin A expression. Mol Cell Biol 24(4):1439–1452.

32. Xu Z, Huang S, Chang LS, Agulnick AD, Brandt SJ (2003) Identification of a TAL1target gene reveals a positive role for the LIM domain-binding protein Ldb1 in ery-throid gene expression and differentiation. Mol Cell Biol 23(21):7585–7599.

33. Palii CG, et al. (2011) Differential genomic targeting of the transcription factor TAL1in alternate haematopoietic lineages. EMBO J 30(3):494–509.

34. Shivdasani RA, Mayer EL, Orkin SH (1995) Absence of blood formation in mice lackingthe T-cell leukaemia oncoprotein tal-1/SCL. Nature 373(6513):432–434.

35. Warren AJ, et al. (1994) The oncogenic cysteine-rich LIM domain protein rbtn2 isessential for erythroid development. Cell 78(1):45–57.

36. Semerad CL, Mercer EM, Inlay MA, Weissman IL, Murre C (2009) E2A proteins main-tain the hematopoietic stem cell pool and promote the maturation of myelolymphoidand myeloerythroid progenitors. Proc Natl Acad Sci USA 106(6):1930–1935.

37. Li L, et al. (2011) Nuclear adaptor Ldb1 regulates a transcriptional program essentialfor the maintenance of hematopoietic stem cells. Nat Immunol 12(2):129–136.

38. Ninkovic J, et al. (2013) The BAF complex interacts with Pax6 in adult neural pro-genitors to establish a neurogenic cross-regulatory transcriptional network. Cell StemCell 13(4):403–418.

39. DengW, et al. (2012) Controlling long-range genomic interactions at a native locus bytargeted tethering of a looping factor. Cell 149(6):1233–1244.

40. Song SH, Hou C, Dean A (2007) A positive role for NLI/Ldb1 in long-range beta-globinlocus control region function. Mol Cell 28(5):810–822.

41. Mylona A, et al. (2013) Genome-wide analysis shows that Ldb1 controls essentialhematopoietic genes/pathways in mouse early development and reveals novel playersin hematopoiesis. Blood 121(15):2902–2913.

42. Li L, et al. (2013) Ldb1-nucleated transcription complexes function as primary medi-ators of global erythroid gene activation. Blood 121(22):4575–4585.

43. Soler E, et al. (2010) The genome-wide dynamics of the binding of Ldb1 complexesduring erythroid differentiation. Genes Dev 24(3):277–289.

44. Love PE, Warzecha C, Li L (2014) Ldb1 complexes: The new master regulators oferythroid gene transcription. Trends Genet 30(1):1–9.

45. Hsu AP, et al. (2013) GATA2 haploinsufficiency caused by mutations in a conservedintronic element leads to MonoMAC syndrome. Blood 121(19):3830–3837, S1–S7.

46. Barski A, et al. (2007) High-resolution profiling of histone methylations in the humangenome. Cell 129(4):823–837.

47. Visel A, et al. (2009) ChIP-seq accurately predicts tissue-specific activity of enhancers.Nature 457(7231):854–858.

48. Bernstein BE, et al.; ENCODE Project Consortium (2012) An integrated encyclopedia ofDNA elements in the human genome. Nature 489(7414):57–74.

49. Giresi PG, Kim J, McDaniell RM, Iyer VR, Lieb JD (2007) FAIRE (Formaldehyde-AssistedIsolation of Regulatory Elements) isolates active regulatory elements from humanchromatin. Genome Res 17(6):877–885.

50. Simon JM, Giresi PG, Davis IJ, Lieb JD (2012) Using formaldehyde-assisted isolation ofregulatory elements (FAIRE) to isolate active regulatory DNA. Nat Protoc 7(2):256–267.

51. Song L, et al. (2011) Open chromatin defined by DNaseI and FAIRE identifies regu-latory elements that shape cell-type identity. Genome Res 21(10):1757–1767.

52. de Pater E, et al. (2013) Gata2 is required for HSC generation and survival. J Exp Med210(13):2843–2850.

53. Weiss MJ, Yu C, Orkin SH (1997) Erythroid-cell-specific properties of transcriptionfactor GATA-1 revealed by phenotypic rescue of a gene-targeted cell line. Mol CellBiol 17(3):1642–1651.

54. Gregory T, et al. (1999) GATA-1 and erythropoietin cooperate to promote erythroidcell survival by regulating bcl-xL expression. Blood 94(1):87–96.

55. Xu Z, Meng X, Cai Y, Koury MJ, Brandt SJ (2006) Recruitment of the SWI/SNF proteinBrg1 by a multiprotein complex effects transcriptional repression in murine erythroidprogenitors. Biochem J 399(2):297–304.

56. Kim SI, Bultman SJ, Kiefer CM, Dean A, Bresnick EH (2009) BRG1 requirement for long-range interaction of a locus control region with a downstream promoter. Proc NatlAcad Sci USA 106(7):2259–2264.

57. Hu G, et al. (2011) Regulation of nucleosome landscape and transcription factortargeting at tissue-specific enhancers by BRG1. Genome Res 21(10):1650–1658.

58. Fujiwara T, Lee HY, Sanalkumar R, Bresnick EH (2010) Building multifunctionality intoa complex containing master regulators of hematopoiesis. Proc Natl Acad Sci USA107(47):20429–20434.

59. Blobel GA, et al. (2009) A reconfigured pattern of MLL occupancy within mitoticchromatin promotes rapid transcriptional reactivation following mitotic exit.Mol Cell36(6):970–983.

60. John S, Workman JL (1998) Bookmarking genes for activation in condensed mitoticchromosomes. Bioessays 20(4):275–279.

61. Fujiwara T, et al. (2009) Discovering hematopoietic mechanisms through genome-wide analysis of GATA factor chromatin occupancy. Mol Cell 36(4):667–681.

62. Wilson NK, et al. (2010) Combinatorial transcriptional control in blood stem/pro-genitor cells: Genome-wide analysis of ten major transcriptional regulators. Cell StemCell 7(4):532–544.

63. Shoval O, Alon U (2010) SnapShot: Network motifs. Cell 143(2):326–e1.64. Yu Y, et al. (2013) Olig2 targets chromatin remodelers to enhancers to initiate oli-

godendrocyte differentiation. Cell 152(1-2):248–261.65. Takeuchi JK, et al. (2011) Chromatin remodelling complex dosage modulates tran-

scription factor function in heart development. Nature Commun 2:187.66. Willis MS, et al. (2012) Functional redundancy of SWI/SNF catalytic subunits in

maintaining vascular endothelial cells in the adult heart. Circ Res 111(5):e111–e122.67. Yokomizo T, et al. (2012) Whole-mount three-dimensional imaging of internally lo-

calized immunostained cells within mouse embryos. Nat Protoc 7(3):421–431.68. Im H, et al. (2005) Chromatin domain activation via GATA-1 utilization of a small

subset of dispersed GATA motifs within a broad chromosomal region. Proc Natl AcadSci USA 102(47):17065–17070.

69. Shen Y, et al. (2012) A map of the cis-regulatory sequences in the mouse genome.Nature 488(7409):116–120.

70. Neph S, et al. (2012) An expansive human regulatory lexicon encoded in transcriptionfactor footprints. Nature 489(7414):83–90.

71. Trompouki E, et al. (2011) Lineage regulators direct BMP and Wnt pathways to cell-specific programs during differentiation and regeneration. Cell 147(3):577–589.

72. Wu W, et al. (2011) Dynamics of the epigenetic landscape during erythroid differ-entiation after GATA1 restoration. Genome Res 21(10):1659–1671.

E1100 | www.pnas.org/cgi/doi/10.1073/pnas.1400065111 Sanalkumar et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 24

, 202

0