cell reports article reports article global changes in the mammary epigenome are induced by hormonal...

26
Cell Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal, 1,3 Toula Bouras, 1,3,8 Wei Shi, 2,5,8 Franc ¸ ois Vaillant, 1,3 Julie M. Sheridan, 1,3 Naiyang Fu, 1,3 Kelsey Breslin, 1 Kun Jiang, 1 Matthew E. Ritchie, 2,3 Matthew Young, 2 Geoffrey J. Lindeman, 1,4,7,8 Gordon K. Smyth, 2,6,8 and Jane E. Visvader 1,3, * 1 ACRF Stem Cells and Cancer Division 2 Bioinformatics Division The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia 3 Department of Medical Biology 4 Department of Medicine 5 Department of Computing and Information Systems 6 Department of Mathematics and Statistics The University of Melbourne, Parkville, VIC 3050, Australia 7 Department of Medical Oncology, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia 8 These authors contributed equally to this work *Correspondence: [email protected] http://dx.doi.org/10.1016/j.celrep.2012.12.020 SUMMARY The mammary epithelium is a dynamic, highly hormone-responsive tissue. To explore chromatin modifications underlying its lineage specification and hormone responsiveness, we determined genome- wide histone methylation profiles of mammary epithe- lial subpopulations in different states. The marked differences in H3K27 trimethylation between subpop- ulations in the adult gland suggest that epithelial cell-fate decisions are orchestrated by polycomb- complex-mediated repression. Remarkably, the mam- mary epigenome underwent highly specific changes in different hormonal contexts, with a profound change being observed in the global H3K27me3 map of luminal cells during pregnancy. We therefore exam- ined the role of the key H3K27 methyltransferase Ezh2 in mammary physiology. Its expression and phosphorylation coincided with H3K27me3 modifica- tions and peaked during pregnancy, driven in part by progesterone. Targeted deletion of Ezh2 impaired alveologenesis during pregnancy, preventing lacta- tion, and drastically reduced stem/progenitor cell numbers. Taken together, these findings reveal that Ezh2 couples hormonal stimuli to epigenetic changes that underpin progenitor activity, lineage specificity, and alveolar expansion in the mammary gland. INTRODUCTION The mammary gland, which comprises a branching ductal epi- thelial network embedded in an adipose-rich stromal matrix, is remarkably adaptive to physiological requirements and undergoes dramatic morphological changes during puberty and pregnancy. At birth, it manifests as a rudimentary branched structure, but ductal elongation and branching commence with puberty and pregnancy provokes the rapid expansion of alveolar units that differentiate into milk-secretory cells prior to parturi- tion. The steroid hormones estrogen and progesterone exert pivotal roles during mammary development via their cognate receptors, the estrogen receptor (ER) and progesterone receptor (PR) (Brisken and O’Malley, 2010). ER is essential for ductal morphogenesis in puberty (Mallepell et al., 2006; Mueller et al., 2002), whereas PR governs ductal side-branching and alveolar development during pregnancy (Brisken et al., 1998; Lydon et al., 1995; Mulac-Jericevic et al., 2003). The mammary epithelium can be divided into two primary line- ages: the myoepithelial lineage constitutes the outer layer of cells that contact the basement membrane, whereas the luminal lineage comprises both ductal and alveolar cells. Adult stem cells prospectively isolated from the mouse mammary gland display the requisite stem cell properties of multilineage differen- tiation and self-renewal (Shackleton et al., 2006; Stingl et al., 2006). A recent study has added a new layer of complexity to the prevailing model of the mammary epithelial hierarchy through the identification of unipotent cells that contribute to homeo- stasis of the gland (Van Keymeulen et al., 2011). It is not yet clear whether these correspond to stem or progenitor cells and what their relationship is to the prospectively isolated epithelial sub- sets. In the mouse, mammary stem cells (MaSCs) have been shown to be highly responsive to steroid hormones (Asselin- Labat et al., 2010; Joshi et al., 2010), while progesterone augmented the number of human bipotent stem-like cells in cellular assays (Graham et al., 2009). MaSCs lie at the apex of the hierarchy and give rise to progenitors and mature cells through progressive restriction. Two distinct types of luminal progenitor cells have been isolated from the mouse mammary Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 411

Upload: ngodat

Post on 15-Jul-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Cell Reports

Article

Global Changes in the Mammary EpigenomeAre Induced by Hormonal Cuesand Coordinated by Ezh2Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8 Francois Vaillant,1,3 Julie M. Sheridan,1,3 Naiyang Fu,1,3 Kelsey Breslin,1

Kun Jiang,1 Matthew E. Ritchie,2,3 Matthew Young,2 Geoffrey J. Lindeman,1,4,7,8 Gordon K. Smyth,2,6,8

and Jane E. Visvader1,3,*1ACRF Stem Cells and Cancer Division2Bioinformatics Division

The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia3Department of Medical Biology4Department of Medicine5Department of Computing and Information Systems6Department of Mathematics and Statistics

The University of Melbourne, Parkville, VIC 3050, Australia7Department of Medical Oncology, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia8These authors contributed equally to this work*Correspondence: [email protected]

http://dx.doi.org/10.1016/j.celrep.2012.12.020

SUMMARY

The mammary epithelium is a dynamic, highlyhormone-responsive tissue. To explore chromatinmodifications underlying its lineage specification andhormone responsiveness, we determined genome-wide histonemethylation profiles ofmammary epithe-lial subpopulations in different states. The markeddifferences in H3K27 trimethylation between subpop-ulations in the adult gland suggest that epithelialcell-fate decisions are orchestrated by polycomb-complex-mediated repression.Remarkably, themam-maryepigenomeunderwenthighly specificchanges indifferent hormonal contexts, with a profound changebeing observed in the global H3K27me3 map ofluminal cells during pregnancy. We therefore exam-ined the role of the key H3K27 methyltransferaseEzh2 in mammary physiology. Its expression andphosphorylation coincided with H3K27me3 modifica-tions and peaked during pregnancy, driven in partby progesterone. Targeted deletion of Ezh2 impairedalveologenesis during pregnancy, preventing lacta-tion, and drastically reduced stem/progenitor cellnumbers. Taken together, these findings reveal thatEzh2 couples hormonal stimuli to epigenetic changesthat underpin progenitor activity, lineage specificity,and alveolar expansion in the mammary gland.

INTRODUCTION

The mammary gland, which comprises a branching ductal epi-

thelial network embedded in an adipose-rich stromal matrix,

C

is remarkably adaptive to physiological requirements and

undergoes dramatic morphological changes during puberty

and pregnancy. At birth, it manifests as a rudimentary branched

structure, but ductal elongation and branching commence with

puberty and pregnancy provokes the rapid expansion of alveolar

units that differentiate into milk-secretory cells prior to parturi-

tion. The steroid hormones estrogen and progesterone exert

pivotal roles during mammary development via their cognate

receptors, the estrogen receptor (ER) and progesterone receptor

(PR) (Brisken and O’Malley, 2010). ER is essential for ductal

morphogenesis in puberty (Mallepell et al., 2006; Mueller et al.,

2002), whereas PR governs ductal side-branching and alveolar

development during pregnancy (Brisken et al., 1998; Lydon

et al., 1995; Mulac-Jericevic et al., 2003).

The mammary epithelium can be divided into two primary line-

ages: themyoepithelial lineage constitutes the outer layer of cells

that contact the basement membrane, whereas the luminal

lineage comprises both ductal and alveolar cells. Adult stem

cells prospectively isolated from the mouse mammary gland

display the requisite stem cell properties of multilineage differen-

tiation and self-renewal (Shackleton et al., 2006; Stingl et al.,

2006). A recent study has added a new layer of complexity to

the prevailingmodel of themammary epithelial hierarchy through

the identification of unipotent cells that contribute to homeo-

stasis of the gland (Van Keymeulen et al., 2011). It is not yet clear

whether these correspond to stem or progenitor cells and what

their relationship is to the prospectively isolated epithelial sub-

sets. In the mouse, mammary stem cells (MaSCs) have been

shown to be highly responsive to steroid hormones (Asselin-

Labat et al., 2010; Joshi et al., 2010), while progesterone

augmented the number of human bipotent stem-like cells in

cellular assays (Graham et al., 2009). MaSCs lie at the apex of

the hierarchy and give rise to progenitors and mature cells

through progressive restriction. Two distinct types of luminal

progenitor cells have been isolated from the mouse mammary

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 411

Page 2: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

CA

B

50%

Score4 8 12

H3K4me3 H3K27me3H3K9me2

100%-5 TSS +5 -5 TSS +5 -5 TSS +5

MaSC/basal subset

H3K4me3 and H3K27me3H3K27me3 onlyH3K4me3 onlyNo H3K4me3/H3K27me3

MaS

C/b

asal

LP ML

1.0

0.8

0.6

0.4

0.2

0

LP vs MaSC/basal

DE genes

TSS Body

K4me3

K27me3

TrilJag2Tbx2Fjx1Wnt10aId4Il17bDlk2Bmp7Igfbp3Vwa2Dll1Elovl4Kirrel3Lama3Dpysl3Ntrk32810032GO3RikGnai1Arhgap24Rtn1ArcCol14a1Ccdc106Wif1Psd2Ism1Nrg1Dkk3Hs3st3a1Lama1Mtap91500009L16RikSema6dLhfpLgals7Apobec1Cacna1gBcat1Sept3Abcg5Fkbp10Sema5aCol23a1Ank2Ugt1a6aArt4TaglnVcanSema3cArsjCtgf1600014C10RikKirrelCd70Antxr1PappaCol17a1Diras2Kcnmb1Sostdc11500015O10RikAardC030048H21RikSh3gl2Moxd1Camk4Tnfrsf11b4732456N10RikCol17a1Krt5Adamts18Irx4PdpnWnt4Upk3aEsrrbCrymCapslLtkScinHomer2Capn6Smarca1Slc38a5Tceal5Ric3MgpUgt1a10Speer4aEgr4Crispld2Sox11MdkAdamts7Zcchc18Hs6st2Drp2Krt15Serpinb11

TSS Body

2

0

-2

1

-1Col

or K

ey

ML vs LP subset

DE genes

K4me3

Tnfsf11Bmp3Slc7a2Defb45Slc16a5Clca3Zbtb8aCapsiRundc3aAdra1aWtipFasIfit2Flrt3MmdVldlrE130203B14RikNtng1Mob3bCcdc129NdnStac2MkxBtbd11Pdlim3EgfrCst3Hey1PkibSnta1Rftn2HexbDock9Crispld2AmtnPtprz1EdnraCcrl2Bcl2St6galnac4Col9a1Gcnt1Nat8Chi3l12610528A11RikAlp1Srgap3Hsd11b1MgllGng7Cyp2d22Il4i1Slc13a2CckSfrp2AbpbCdhr1Cyp24a1Tspan8Cd177Anxa8Egln3Gprc5bPaqr6Cd44Celf2Ncam1Adam23Pi16Zic4Efhd1Mfap2Ramp1Cited2Rb1Cdc42ep3Maml2Slc4a4Nedd9Serpinf1Fam49aEndod1Ephx1Ppap2b1300014l06RikPmp22Chn2Arhgef6Plb1Cdr2Slitrk4PdgfraPaplnPgm5Stard8Fbn1Pisd-ps2Gsg1lH2-OaCol5a1

K27me3

Figure 1. Histone Methylation Profiles of Mammary Epithelial Subpopulations in the ‘‘Steady State’’ and Their Correlation with Gene

Expression Changes

(A) Genome-wide heat map showing the pattern of H3K4me3, H3K9me2, and H3K27me3marks in MaSC-enriched cells from 5 kb upstream to 5 kb downstream

of the TSS of each gene. Rows correspond to genes clustered by coverage pattern. All 26,310 genes in themm9 genome are shown. The first three columns show

(legend continued on next page)

412 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 3: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

gland, but committed basal progenitor cells remain elusive (As-

selin-Labat et al., 2007, 2011; Sleeman et al., 2007).

Although a number of transcription factors and pathways have

been implicated in controlling specific steps along the mammary

differentiation hierarchy (reviewed in Visvader, 2009), the role of

the epigenome in regulating cell-fate decisions and differentia-

tion within this epithelial compartment remains unclear. In other

systems, there is substantial evidence that histone methylation

governs lineage-specific developmental programs and that its

deregulation leads to oncogenesis (Bracken and Helin, 2009;

Sauvageau and Sauvageau, 2010). It is thought that histone

modifications establish discrete domains of active and inactive

chromatin to effect gene expression. Histone lysine methylation

can serve as either an active or repressive mark: trithorax-medi-

ated methylation of lysine 4 on histone H3 within nucleosomes is

associated with activated gene expression, while methylation of

lysine 27 by polycomb group (PcG) proteins is linked with gene

repression and chromatin condensation (Margueron and Rein-

berg, 2011). Ezh2, a member of the PcG family, is a histone

methyltransferase that forms the catalytic component of the

polycomb repressive complex PRC2. This complex silences

lineage specification genes to regulate the maintenance and

differentiation of embryonic and adult stem cells (reviewed in

Margueron and Reinberg, 2011). In embryonic stem cells, where

genome-wide histone methylation patterns have been exten-

sively studied, key developmental genes often exhibit both

repressive H3K27me3 marks and activating H3K4me3 marks

(Bernstein et al., 2006). This bivalent modification has been

proposed tomaintain these genes ‘‘poised’’ for subsequent acti-

vation or repression upon lineage specification. In vivo mapping

studies indicate that PcG-dependent H3K27me3 selectively

marks genes in the epidermal lineages and controls gene ex-

pression changes during the differentiation of skin stem cells

(Lien et al., 2011).

In this report, we examine the contribution of epigenetic mech-

anisms to regulation of the lineage hierarchy in the steady-state

mammary gland and in response to different hormonal milieu.

We determined genome-wide histone methylation profiles of

the MaSC-enriched, luminal progenitor, and mature luminal

subsets. Correlating the global H3K4me3 and H3K27me3 modi-

fication maps with gene expression signatures indicated that

the epigenome has an important role in directing cell-fate

changes from the basal to luminal cell lineage. Moreover, the

mammary epigenome was found to be highly sensitive to dif-

ferent hormonal environments. H3K27 trimethylation of chro-

matin emerged as a key mediator of gene expression changes

during pregnancy, concomitant with high levels of Ezh2, appar-

coverage depth on a linear color scale, with the x axis showing distance from the T

In this case, the x axis shows the scale from 4 (nonexpressed) to 12 (maximum

ordered by expression level: the fifth gene group shows little histone marking o

H3K4me3 and increasing H3K27me3 levels. Genes are sorted by expression wit

(B) Segmented bar graphs showing genome-wide percentage of genes with histon

marks in the TSS region are shown, but virtually identical data were obtained for

(C) Heatmaps of gene expression and histone modification changes as cells res

mature luminal (ML) cells. Columns give log2-fold changes for differential gene e

H3K27me3 marking across the broad gene, respectively, for the 200 most differ

See also Figure S1 and Table S1.

C

ently activated by phosphorylation. Targeted deletion of Ezh2 in

the mammary epithelium dramatically reduced both ductal and

alveolar morphogenesis. The expression of Ezh2 and its phos-

phorylation appear to be coordinated through progesterone,

a key pregnancy hormone. Thus, hormonally driven expansion

of the alveolar compartment is programmed, at least in part,

by Ezh2-mediated changes in chromatin modification. Given

the critical importance of both progesterone and EZH2 to breast

cancer, these data implicate progesterone-induced global

changes in chromatin structure in the genesis of this disease.

RESULTS

Histone Methylation Landscapes of Mammary EpithelialSubpopulations in the ‘‘Steady State’’To explore the relevance of histone modification to the regula-

tion of gene expression along the mammary differentiation hier-

archy, chromatin immunoprecipitation sequencing (ChIP-seq)

was performed on distinct epithelial populations that have

been prospectively isolated from the mouse mammary gland.

These correspond to MaSC-enriched (CD29hiCD24+), com-

mitted luminal progenitor (CD29loCD24+CD61+), and mature

luminal cells (CD29loCD24+CD61�) (Asselin-Labat et al., 2007;Shackleton et al., 2006), all readily isolated from FVB/N glands.

The MaSC-enriched population also contains mature myoepi-

thelial cells and likely basal progenitor cells and is referred to

as the MaSC/basal subset from here on. To avoid changes

that are known to occur in epithelial cells during cell culture,

freshly sorted subsets (approximately 250,000 cells) isolated

from young adult females were used for ChIP. No preamplifica-

tion step was incorporated prior to library preparation in order

to avoid potential bias. High-resolution genome-wide maps

were determined for H3K4me3, H3K27me3, and H3K9me2

modifications. Between 17 and 40 million DNA fragments were

sequenced for each ChIP or input sample using 35 bp paired-

end reads (Table S1).

The overall pattern of histone methylation marking was exam-

ined using heatmaps (Figure 1A and Figure S1A) and density

plots (Figure S1B) of fragment coverage. H3K4me3 occupancy

typically peaked sharply around the transcriptional start site

(TSS) of each gene, whereas the repressive H3K9me2 and

H3K27me3 marks were more evenly spread over the promoter

region and gene body, although with weaker peaks and troughs

still discernible around the TSS. In the MaSC/basal subset,

genes could be clustered into five broad groups by the level

of H3K4me3 marking, from very high to very low (Figure 1A).

Apart from the fifth cluster, lacking any of the marks, the other

SS. The far right column shows log2-normalized expression for the same genes.

expression). Genes are clustered into five groups by their histone pattern and

r expression, while the other four groups correspond roughly to decreasing

hin each cluster, so the right panel appears as an increasing curve.

e methylationmarks in each epithelial subset (FDR < 0.05). Percent H3K27me3

H3K27me3 marks across the gene body plus the TSS.

trict from the MaSC/basal to luminal progenitor (LP) cells and from the LP to

xpression (DE), H3K4me3 marking, H3K27me3 marking in the TSS region, and

entially expressed genes.

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 413

Page 4: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

C

A

ckit

Trp63Krt5Snai2

Elf5 Hey2

Fol

d ch

ange

vs In

put

MaSC

ML

LP

ML

LP

TSS TSS TSS

TSS TSS TSS

Fol

d ch

ange

vs In

put

H3K4me3 H3K27me3

1.8

1.0

0.2

4.0

2.0

1.2

0.8

0.4

1.2

0.8

0.4

1.8

1.2

0.6

6.0

4.0

2.0

MaSC LP ML

1.0

3.0

0

0.6

1.4

0 0

000

MaSC

MaSC LP ML MaSC LP ML

MaSC LP ML MaSC LP ML MaSC LP ML

ML vs LP subsetLP vs MaSC/basal subset

2

1

0

-1

-2

2

1

0

-1

-2-6 -4 0 2 4-2 -4 0 2-2

3

-3

2

1

0

-1

-2

-6 -4 0 2 4-2

3

-3

2

1

0

-1

-2

-4 0 2-2

Log

fold

cha

nge

Expression log fold change

H3K

4me3

H3K

27m

e3

Log

fold

cha

nge

Expression log fold change

H3K

4me3

H3K

27m

e3

B

(legend on next page)

414 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 5: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

four clusters showed an inverse pattern of H3K4me3 and

H3K27me3 marking. H3K9me2 occupancy was relatively inde-

pendent of H3K4me3 and H3K27me3 status. The luminal pro-

genitor and mature luminal cell populations exhibited similar

patterns (Figure S1A).

H3K4me3 and H3K27me3 but Not H3K9me2 Correlatewith Overall Gene ExpressionNext, we related the histone methylation patterns to our previ-

ously published microarray gene expression profiles for the

same cell subpopulations (Lim et al., 2010). Expression level is

shown in the right-most panel of each heatmap, with genes

sorted from high to low expression within each cluster (Figures

1A and S1A). While all the clusters contain genes with a range

of expression levels, average expression strongly increased

with H3K4me3 coverage and decreased with H3K27me3

coverage (Figure 1A). This held for all cell subpopulations (Fig-

ure S1A). While H3K27me3 marks were distributed across the

whole promoter region and gene body, gene expression was

especially sensitive to coverage just upstream and downstream

of the TSS, with low expressed genes showing a trough in

H3K27me3 coverage around the TSS relative to the gene body

and highly expressed genes showing a downstream peak (Fig-

ure S1B). Unlike the other two histone marks, H3K9me2 showed

no overall correlation with expression and was equally associ-

ated with both high and low expression.

To assess enrichment for each histone mark statistically,

we recorded the number of fragments mapping within 3 kb

upstream to 2 kb downstream of the TSS of each gene. For

H3K27me3, the number of fragments mapping to a broad region

comprising the TSS and the entire genomic span of the gene

were recorded. For each cell sample and each histone mark,

enrichment was assessed using a statistical model that treats

the input fragment count profile as representative of nonenriched

genes. Most genes were significantly marked by at least one of

the three modifications (Figure 1B). Interestingly, the number of

genes showing enriched H3K27me3 occupancy at the TSS

increased upon luminal lineage specification (21%, 32%, and

34% in the MaSC/basal, luminal progenitor, and mature luminal

subsets, respectively). Moreover, bivalent marking at the TSS

increased upon basal to luminal cell commitment (10% and

22% in the MaSC/basal and luminal progenitor subsets). As

the MaSC subset remains heterogeneous, it is not clear whether

bivalency plays a role in these adult stem cells.

Expression Changes Correlate with HistoneMethylationChanges during Lineage RestrictionGenes with increased expression in the luminal progenitor rela-

tive to the MaSC/basal subset tended to show increased

H3K4me3 and decreased H3K27me3 marking, whereas genes

Figure 2. Histone Methylation Profiles across Key MaSC-Enriched and

(A and B) Scatter plots show that expression changes for the 500 most DE gen

correlated with H3K27me3 (bottom panels) (p < 10�6).

(C) Read coverage graphs for H3K4me3 (red) and H3K27me3 (blue) in each epith

luminal progenitor-specific genes. Y-axes show fragments per million on the sca

samples; error bars show SEM.

See also Figure S1 and Table S1.

C

with decreased expression showed the opposite epigenetic

changes (Figures 1C and 2A). The same was true of genes differ-

entially expressed in the mature luminal versus the luminal

progenitor subset (Figures 1C and 2B). This shows that histone

methylation is dynamically associated with gene expression

during lineage restriction and is likely to be a key mediator of

expression changes that direct basal to luminal cell-fate switch-

ing and luminal maturation. Illustrative read coverage graphs of

H3K4me3 and H3K27me3 patterns across genes characteristic

of the MaSC/basal (snai2, cytokeratin 5, and Trp63) and luminal

progenitor subsets (c-kit, Elf-5, and Hey2) are shown in Fig-

ure 2C. Histone modifications in the TSS regions were verified

for a number of candidates using ChIP combined with quantita-

tive RT-PCR (Figure 2C).

Hormonal Changes Drive Global Mammary EpigeneticAlterationsSince hormone deprivation and pregnancy drastically alter

MaSC numbers and the gene expression profiles of different

epithelial subtypes (Asselin-Labat et al., 2010), we investigated

whether histone methylation is a regulator of these changes.

ChIP-seq profiles were generated of H3K4me3 and H3K27me3

marks in the MaSC/basal and luminal populations from the

glands of ovariectomized or midpregnant (12.5 days) mice as

well as from control virgin mice. The total luminal subset

(CD29loCD24+) was used, given that the CD61+ progenitor pop-

ulation declines precipitously during pregnancy (Asselin-Labat

et al., 2007). Expression changes in ovariectomized mice were

limited to a few dozen genes (15 differentially expressed genes

in the MaSC/basal subset and 82 genes in luminal cells at

a 5% false discovery rate [FDR]) and were only modestly

correlated with epigenetic changes (Figure 3A). Nevertheless,

changes in the expression of specific genes, such as Arf and

cyclin D2, in the MaSC/basal population correlated with

changes in H3K27me3 occupancy at their TSS (Figure S2A).

Cell cycle genes were significantly enriched among differentially

H3K27me3-marked genes in the luminal population upon ovari-

ectomy (p = 0.02), consistent with cell cycle regulators driving

proliferation in response to steroid hormones.

A more global function for H3K27 trimethylation was revealed

in the hormonal milieu of pregnancy that is largely governed by

progesterone and prolactin. Most noticeably, a strong inverse

relationship was apparent between H3K27me3 marking and

expression changes in the luminal cells of pregnant mice (Fig-

ure 3B). Key luminal genes required for differentiation and

concomitant milk production and upregulated in pregnancy

(Elf-5, Wap, and Csn2) showed strongly decreased H3K27me3

modifications that were confirmed by ChIP-quantitative RT-

PCR (qRT-PCR) analysis (Figure 3C). Conversely, luminal genes

expressed in the steady-state gland but downregulated during

Luminal Progenitor Genes

es are directly correlated with H3K4me3 changes (top panels) and inversely

elial subset 10 kb ± of the TSS region of three MaSC/basal-specific and three

le 0–10. Bar plots show ChIP qRT-PCR data for three independent biological

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 415

Page 6: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Log

fold

cha

nge

MaSC/ basal Lum

Expression log fold change

OVX vs control pregnant vs virgin

H3K

4me3

H3K

27m

e3

3

10

-2

0 4-6

-4 0 2 4-2 6

-4 0 2 4-2 6

MaSC/ basal Lum

32

0

-2-3

0 5-5

-5

Expression log fold change

1

-1

-4-8 -2 2

2

-1

-3-4-4

3

10

-2

2

-1

-3-4

0 4-6 -4-8 -2 2

32

0

-2-3

0 5-5

1

-1

-4

32

0

-2-3

1

-1

-4

32

0

-2-3

1

-1

-4

32

0

-2-3

1

-1

-4

0 5

0 5-5

32

0

-2-3

1

-1

-4

FoxA1

Wnt7b

Hey1

Fol

d ch

ange

vs

Inpu

t

1.51.0

0.50

3.0

2.5

2.0

1.5

1.0

0.5

0

3.02.5

2.0

0.4

0

1.8

1.2

0.8

virgin

12.5 dP

virgin

12.5 dP

Wap

Elf5

Csn2

Fol

d ch

ange

vs

Inpu

t F

old

chan

ge v

s In

put

Fol

d ch

ange

vs

Inpu

t

virgin 12.5 dP

virgin 12.5 dP

virgin 12.5 dP

virgin

12.5 dP

1.2

0.6

1.2

0.4

0.8

1.2

0.4

0.8

1.6

0

virgin

12.5

0

0

BA

DC

virgin

12.5

virgin

12.5

virgin

12.5

virgin

12.5

virgin

12.5

Fol

d ch

ange

vs

Inpu

tF

old

chan

ge v

s In

put

virgin 12.5 dP

virgin 12.5 dP

virgin 12.5 dP

Figure 3. The Mammary Epithelial Epigenome Is Influenced by Hormonal Status

(A and B) Scatter plots of expression versus epigenetic log2-fold changes in the mammary epithelial subsets of (A) ovariectomized mice and (B) pregnant mice.

Increased H3K27me3 marking strongly mediates decreased expression in luminal cells from 12.5 day pregnant glands (p < 10�6). Other correlations were also

significant (p < 0.05), except H3K27me3 modifications in the MaSC subset at midpregnancy.

(C) Derepression of milk genes and Elf-5 in the luminal subset of pregnant glands at 12.5 days. The left panel shows read coverage of H3K27me3 marks around

the TSS of each gene. The right panel shows ChIP-qRT-PCR confirmation (n = 3; error bars show SEM).

(D) Repression of luminal commitment genes (Wnt7b, Foxa1, and Hey1) in the luminal subset during pregnancy correlates with increased H3K27me3 marks. The

left panel shows read coverage for H3K4me3 (red) and H3K27me3 (blue) around the TSS of each gene. The right panel shows ChIP-qRT-PCR confirmation (n = 3;

error bars show SEM).

See also Figure S2, and Tables S1 and S2.

416 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 7: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

pregnancy, such as wnt7b, and the luminal commitment genes

Foxa1 (Bernardo et al., 2010) and Hey1 (Bouras et al., 2008)

had abundant H3K27me3 modifications at 12.5 days of

pregnancy, as validated by ChIP-qRT-PCR (Figure 3D). These

changes likely reflect a shift in gene expression within the

emerging alveolar cells toward the highly specialized function

of milk production. In general, genes repressed in pregnancy

with a concomitant increase in H3K27me3 marking were en-

riched for the mammary morphogenesis and developmental

gene categories. Genes upregulated during pregnancy and

with reduced H3K27me3 modifications showed enrichment for

lipid biosynthesis and lipid catabolism (Table S2), commensu-

rate with the changed mammary function during pregnancy.

Although H3K27me3 was not consistently correlated with ex-

pression changes in the MaSC/basal subset of pregnant mice, it

nevertheless appeared to play an important role for specific

genes. In particular, it was associated with derepression of a

number of genes that are normally expressed only in the luminal

lineage. This observation is of particular interest given the

dramatic expansion of theMaSC pool and its altered gene signa-

ture during pregnancy (Asselin-Labat et al., 2010). Quantitative

RT-PCR confirmed expression of the milk protein genes Wap

and Csn2 and the luminal progenitor transcription factor Elf-5,

all of which are normally restricted to luminal subpopulations

(Lim et al., 2010; Figure S2B). Compatible with their derepres-

sion, each of these genes showed diminished H3K27me3 marks

at their TSS during pregnancy, as confirmed by ChIP-qRT-PCR

analysis (Figure S2C). Thus, lineage-priming may occur in the

expanded stem cell population during pregnancy prior to com-

mitment along the alveolar lineage. Intriguingly, expression of

the basal-specific gene Lgr5 was extinguished in the MaSC

pool during pregnancy, accompanied by augmented H3K27 tri-

methylation (Figures S2B and S2C).

At a more global level, the total number of genes within the

luminal subset with significant (FDR< 0.05) H3K27me3modifica-

tions relative to input increased in pregnancy but decreased in

ovariectomized mice significantly (Figure S2D). In summary,

striking epigenetic changes occurred within a specific cellular

subset during pregnancy and were selectively observed for

H3K27me3 but not H3K4me3 or H3K9Ac modifications (data

not shown), which showed small changes.

Dynamic Expression of the Polycomb Group RepressorEzh2 in the Mammary GlandIn view of the marked hormone-induced changes in the global

H3K27me3 profile during pregnancy, we examined the ex-

pression of Ezh2 during mammary ontogeny. Ezh2 is the core

enzymatic subunit of PRC2 that catalyzes K27 trimethylation

on H3 (Margueron and Reinberg, 2011) and has emerged as

an important prognostic marker in breast cancer. Western blot

analysis showed that Ezh2 expression was low in virgin glands

and peaked during early to midpregnancy before declining in

late pregnancy (Figure 4A). Interestingly, the profile of total

H3K27me3-modified protein closely mirrored that of Ezh2 (Fig-

ure 4A). Immunohistochemical staining confirmed the ex-

pression of Ezh2 during mammary morphogenesis and further

revealed that it was abundant in the terminal end buds (TEBs)

of the developing pubertal gland, with lower levels visible in the

C

nuclei of myoepithelial and luminal cells of mature ducts (Fig-

ure 4B). Ezh2 staining was most intense in the ducts and alveoli

during pregnancy (Figure 4B) and declined to low levels in

lactating and involuting glands (Figure 4B).

Ezh2 Deficiency Delays Mammary Morphogenesisduring PubertyTo investigate the physiological role of Ezh2 in the mammary

gland, we conditionally targeted the Ezh2 locus using cre recom-

binase driven by the mouse mammary tumor virus (MMTV)

promoter. Fluorescence-activated cell sorting (FACS) analysis

of reporter mice demonstrated that the MMTV promoter is active

in both luminal and basal mammary epithelial cells (Figure S3A)

but not in the stroma (<0.05% cells). Immunohistochemistry

and western blot analysis confirmed efficient deletion of Ezh2

in MMTV-cre;Ezh2f/f glands (Figures 4A and 4C). Its deletion

markedly impaired elongation and branching of the mammary

epithelial tree during puberty (n = 6), resulting inmarkedly smaller

ductal trees in young adult mice (n = 14) relative to control age-

matched glands (n > 40) from either MMTV-cre;Ezh2f/+, Ezh2f/f,

or Ezh2f/+ littermates or MMTV-cre mice (Figures 4D and S3B).

The same phenotype was observed on a mixed C57Bl6/FVB/N

or pure FVB/N background. MMTV-cre transgenic females

behaved like wild-typemice and could support large-sized litters

through normal lactation, thus differing from the cre strains

recently described (Robinson and Hennighausen, 2011). The

delay in morphogenesis was also associated with decreased

filling of the fat pad in Ezh2-deficient mice (Figure 4E) and the

persistence of TEBs (Figure S3B). By 12 weeks of age, ductal

morphogenesis of most Ezh2-deficient mammary glands was

partially rescued, presumably through recurrent hormonal stim-

ulation (Figure S3B). Cell fate appeared unaffected by loss of

Ezh2, as assessed by immunostaining for luminal, myoepithelial,

and alveolar markers (K18, K14, p63, and Npt2b) (Figures S3C

and S3D; data not shown).

Ezh2Controls the Activity ofMammary Stem/ProgenitorCellsA potential role for Ezh2 in regulating MaSC function was next

addressed using the mammary fat pad reconstitution assay.

Limiting dilution assays of freshly sorted CD29hiCD24+ cells re-

vealed a 14-fold decrease in the frequency of mammary repopu-

lating cells in MMTV-cre;Ezh2f/f compared to control mammary

glands (Table 1). This observation indicates that Ezh2 has amajor

role in either stem or descendant progenitor cells. Further delin-

eation of the hierarchy will be required to distinguish between

these possibilities.

The effect of Ezh2 loss on epithelial proliferation was next

assessed by in vivo bromodeoxyuridine (BrdU) labeling. Prolifer-

ative activity was significantly impaired in the TEBs of MMTV-

cre;Ezh2f/f mammary glands compared to MMTV-cre;Ezh2f/+

and Ezh2f/+ control glands (Figures 4F and S3E). Furthermore,

both the MaSC/basal and luminal populations from Ezh2-defi-

cient glands had little clonogenic activity in vitro on fibroblast

feeders, with only small colonies visible (Figure 4G). These

data point to a critical role for Ezh2 in regulating the activity

of multiple progenitor cell types in the mammary gland. It

seems likely that the developmental defect in MMTV-cre;Ezh2f/f

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 417

Page 8: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

A

CB

4 w

eeks

8 w

eeks

6.5

days

12

.5 d

ays

16.5

day

s

pregnancy

H3K27me3

Tubulin

4 w

eeks

8 w

eeks

6.5

days

12

.5 d

ays

16.5

day

s

Tubulin

Ezh2

D

F

Ezh2

f/+

cKO

18.5

day

s

virgin pregnancy

18.5

day

s 2

dL

virgin

6 weeks virgin 6.5 dP

12.5 dP 1 dL 4 dI

8 weeks virgin

MM

TV

-cre

; Ezh

2f/f

Ezh2

Ezh

2f/+

CD29hi CD29lo

MM

TV

-cre

; E

zh2f/f

Ezh

2f/+

Col

onie

s pe

r 10

0 ce

lls

CD29lo CD29hi

10

0

35

30

20

25

15

5

* *

Ezh2f/+

MMTV-cre; Ezh2f/f

MMTV-cre; Ezh2f/fMMTV-cre; Ezh2f/+Ezh2f/+

G

MMTV-cre; Ezh2f/fEzh2f/+ MMTV-cre; Ezh2f/+

E

Ezh2f/+

MMTV-cre; Ezh2f/f

Fat

pad

fi lli

ng (

%)

6 7 8

weeks

0

100

60

80

40

20

(legend on next page)

418 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 9: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Table 1. Limiting Dilution Analysis of theMammary Repopulating

Frequency of CD29hiCD24+ Cells Isolated from YoungMMTV-cre;

Ezh2f/f or Control Glands

Number of CD29hiCD24+ Cells

Injected per Mammary Fat Pad

Number of Positive Outgrowthsa

Ezh2f/+ MMTV-cre; Ezh2f/f

100 5/8

200 2/5 0/6

400 11/16 0/10

800 4/7 1/17

1,600 5/6 1/5

3,200 9/12

6,000 2/7

Repopulating frequency 1/460 1/6,419

(95% confidence interval): (1/722–1/293) (1/11,188–1/3,683)

p value <0.00001

CD29hiCD24+ cells frommammary glands of 8- to 9-week-old mice were

injected into the cleared mammary fat pads of 3-week-old nonobese dia-

betic-severe combined immunodeficient female recipients. Data are

pooled from three independent experiments collected 8 weeks post-

transplantation. The repopulation frequency was calculated using limiting

dilution analysis as described (Hu and Smyth, 2009).aShown as number of outgrowths per number of injected cleared

mammary fat pads.

mammary glands manifests in puberty, because this stage

requires large numbers of progenitor cells to orchestrate ductal

growth (Asselin-Labat et al., 2007).

Loss of Ezh2 Profoundly Affects the Expression of CellCycle and Epidermal GenesTo identify potential downstream effectors of Ezh2, the genome-

wide transcriptional profiles of the MaSC/basal and luminal pop-

ulations in their steady state were determined following ex vivo

cre-mediated excision of Ezh2. Similar to freshly sorted cells

(Figure 4G), the clonogenic capacity of these Ezh2-deficient

MaSC/basal and luminal populations was dramatically reduced

compared to control cultures (Figure S4A). Gene ontology anal-

ysis of the top 500 differentially expressed genes revealed a sig-

nificant association with cell cycle, DNA replication, and DNA

Figure 4. Ezh2 Is Required for Normal Mammary Gland Development

(A) Western blot analysis of mammary gland lysates for expression of Ezh2 and H3

provided the controls.

(B) Immunohistochemical staining of mammary gland tissue sections from FVB

(6 weeks), mature ducts in 8-week-old mice; alveoli in early pregnancy (6.5 d

bars, 50 mm.

(C) Immunohistochemical staining of TEBs for Ezh2 expression in MMTV–cre; Ez

(D) Whole-mounts of mammary glands from 7-week-old virgin MMTV–cre; Ezh2f/f

lymph node in the inguinal gland is marked by a white arrow. Scale bars, 2.0 mm

(E) Extent of fat pad filling was estimated in virgin mice at 6, 7, and 8 weeks of a

(F) Immunohistochemical staining of terminal end buds for BrdU incorporation, s

glands compared to those from Ezh2f/+ and MMTV–cre; Ezh2f/+ mice. Scale bar

(G) Colony-forming capacity of sorted MaSC-enriched (CD29hiCD24+; labeled C

fibroblast feeders from 8-week-old MMTV–cre; Ezh2f/f mice compared to Ezh2f

CD61+ luminal progenitors that could be isolated from the smaller targeted gland

quantitation of the colony forming capacity of the CD29loCD24+ and CD29hiCD2

represent mean ± SD of three independent experiments, with eight replicates fo

See also Figure S3.

C

repair (Figure S4B). Of the top ranked genes, three potent cell

cycle inhibitors were derepressed: Cdkn1c (p57), Cdkn2a

(Ink4a/Arf), and Cdkn1a (p21). Consistent with Arf being an

important target of Ezh2, Arf transcript levels were considerably

lower in the luminal population from pregnant glands than in

other subsets and Arfwas derepressed in Ezh2-deficient luminal

cells at midpregnancy (Figure S4C). Gene expression changes in

Ezh2-deficient cells were inversely correlated with changes in

pregnancy (Asselin-Labat et al., 2010), with 67% (52 of 78) of

differentially expressed genes in the MaSC/basal subset and

91% (146 of 160) in the luminal subset showing changes in oppo-

site directions.

Interestingly, one of the top derepressed gene sets in the

MaSC/basal population, other than those related to cell cycle

regulation, was keratinocyte differentiation (Figure S4B). The

expression of genes within the epidermal differentiation complex

on mouse chromosome 3, previously shown to be a target of

Ezh2 repression in skin (Ezhkova et al., 2009), was activated in

the MaSC-enriched subset from Ezh2-deficient glands. This is

consistent with previous reports of misexpression of nonlineage

genes associated with Ezh2 deletion (see Discussion). To inves-

tigate a potential relationship between the gene expression sig-

natures of Ezh2-deficient cells and metaplastic breast cancers,

the molecular profiles of the different breast cancer subtypes

were interrogated with the Ezh2-deficient MaSC/basal cell

signature. Intriguingly, this signature was found to bemost highly

represented in the claudin-low subgroup based on signature ex-

pression scores (Figure S4D). The claudin-low subtype exhibits

metaplastic features, expresses lower levels of Ezh2 than the

other subtypes, and can even display epidermal traits (Keller

et al., 2012), suggesting that they have undergone metaplasia

as a result of aberrant differentiation.

Ezh2 Deficiency Leads to Reduced AlveolarDevelopment and Failure of LactationWe next examined the effect of Ezh2 deficiency on pregnancy

and lactation. Although alveoli formed in MMTV-cre;Ezh2f/f

mammary glands during pregnancy (Figure 5A), they were fewer

and more disorganized than those in Ezh2f/+, Ezh2f/f, or MMTV-

cre mammary glands (Figure 5B; data not shown). This pheno-

type appeared most obvious from midpregnancy (n = 9), where

K27me3 protein during development. Ezh2 cKO tissue (12.5 dP) and antitubulin

/N females for Ezh2 expression, representing TEBs that characterize puberty

P) and midpregnancy (12.5 dP); lactation (1 dL); and involution (4 dI). Scale

h2f/f mice compared to littermate Ezh2f/+ glands. Scale bars, 50 mm.

mice compared to glands from Ezh2f/+ and MMTV-cre; Ezh2f/+ littermates. The

.

ge using ImageJ software, with four to nine mice for each time point.

howing reduced numbers of proliferating cells in MMTV–cre; Ezh2f/f mammary

s, 50 mm. Isotype control antibody panels are shown in insets.

D29hi) and luminal cells (CD29loCD24+; labeled CD29lo) grown on irradiated/+ littermates. The same results were obtained for 6-week-old mice. The few

s also had reduced clonogenic activity (data not shown). Histogram showing

4+ subpopulations (200 and 300 cells were plated per well, respectively). Data

r each. *t test p < 0.0001 compared to Ezh2f/+ controls for both subsets.

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 419

Page 10: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

A

B

16.5

dP

12.5

dP

MMTV-cre; Ezh2f/f

16.5

dP

Ezh2f/+

E

MMTV-cre; Ezh2f/fMMTV-cre

C

D

Tubulin

Ezh2

H3K27me3

MMTV-cre;Ezh2f/f Ezh2f/+

Cdk

n1c

Fol

d ch

ange

vs

Inpu

t

0

1.5

1.0

0.5

2.0

MMTV-cre; Ezh2f/f

Ezh2f/+

Fol

d ch

ange

vs

Inpu

tC

amk2

n1

0

3.5

2.5

1.5

0.5

MMTV-cre; Ezh2f/f

Ezh2f/+ Fol

d ch

ange

vs

Inpu

tW

nt7b

0

2.5

1.5

1.0

0.5

2.0

MMTV-cre; Ezh2f/f

Ezh2f/+

Cdk

n2a

Fol

d ch

ange

vs

Inpu

t

MMTV-cre; Ezh2f/f

Ezh2f/+0

4

3

2

1

Ezh2f/+

12.5

dP

MMTV-cre; Ezh2f/fMMTV-cre; Ezh2f/+ Ezh2f/+

Figure 5. Ezh2 Deficiency Leads to Abnormal Alveolar Development

(A) Whole-mounts of mammary glands from MMTV–cre; Ezh2f/f mice (right panel) compared to those from Ezh2f/+ and MMTV–cre; Ezh2f/+ mice at day 12.5 of

pregnancy show retardation of ductal growth. Mice were mated at 7 weeks of age. No gross abnormalities in the alveolar units were evident in early pregnancy

(6.5 dP; data not shown). Scale bars, 4.0 mm.

(B) H&E sections of mammary glands from MMTV–cre;Ezh2f/f, Ezh2f/+ littermates, and MMTV-cre mice at days 12.5 and 16.5 of pregnancy. Scale bars, 50 mm.

(C) Western blot analysis of mammary glands from MMTV-cre; Ezh2f/f and Ezh2f/+ littermate control mice for expression levels of Ezh2, H3K27me3 protein, and

tubulin.

(D) Derepression of cell cycle genes in pregnant glands lacking Ezh2. ChIP-qRT-PCR for H3K27me3 marks across Cdkn2a/Arf, Cdkn1c (p57), Wnt7b, and

Camk2n1 in the expanding luminal population from day 12.5 pregnant mice. Histograms show the mean of two independent samples with at least two technical

replicates for each.

(E) Immunostaining for milk protein of MMTV-cre; Ezh2f/f tissue sections (right panel) compared to an Ezh2f/+ littermate control at 16.5 days of pregnancy (left

panel). Scale bars, 100 mm.

See also Figures S3 and S4.

ductal elongation of the mammary tree remained stunted in 50%

of Ezh2-deficient glands (Figure 5A). Heterozygotes seemed to

have an intermediate phenotype, with less dense but apparently

normal alveoli (Figures 5A and S3F). Notably, progenitor cell

activity was severely compromised in all three epithelial subsets

isolated frommidpregnant Ezh2-deficient mammary glands (Fig-

ure S5). Western blot analysis confirmed loss of Ezh2 in these

glands and showed a pronounced decrease in H3K27me3

protein in Ezh2-deficient glands in either the pregnant (Figure 5C)

or virgin state (Figure S5D). The residual level of H3K27me3

protein in targeted glands may reflect low levels of Ezh2 de-

tectable in MMTV-cre;Ezh2f/f mammary glands but also sug-

gests that other methylases contribute to H3K27 trimethylation.

Importantly, ChIP combined with qRT-PCR confirmed that the

420 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

cell cycle inhibitors Arf and p57 (Cdkn1c) were derepressed in

primary Ezh2-deficient epithelial cells at midpregnancy (Fig-

ure 5D), as well as other genes abundantly marked by

H3K27me3 at this time-point, including Wnt7b and Camk2n1.

Overall, these data indicate that Ezh2 plays an important role

in the methylation of H3K27 in mammary epithelial cells.

Milk production was readily detectable in MMTV-cre;Ezh2f/f

glands at 16.5 days of pregnancy, suggesting that Ezh2 does

not affect alveolar differentiation (Figure 5E). However, lactation

was severely compromised (n = 4), resulting in all pups dying

within 2 or 3 days of birth. Concordantly, Ezh2-deficient mam-

mary glands showed grossly abnormal morphology at day two

of lactation, relative to control glands that underwent normal

lactation (Figure S3G). This phenotype is likely to result from

Page 11: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

a decrease in alveologenesis rather than differentiation, reflect-

ing the critical role of Ezh2 in regulating progenitor cell activity.

Progesterone-Mediated Regulation of Ezh2 ExpressionWe explored whether posttranscriptional control mechanisms

contributed to augmented Ezh2 expression during pregnancy,

since only a modest increase in Ezh2 transcript levels was evi-

dent in epithelial subsets from midpregnant glands (Asselin-

Labat et al., 2010). Ezh2 has been shown to be phosphorylated

on multiple residues, including serine 21 and threonine residues

345 and 487 (reviewed in Caretti et al., 2011). Western blot

analysis using phosphospecific antibodies revealed a striking

increase in the level of phosphorylated Ezh2 on Thr487 in early

tomidpregnancy (Figure 6A). Little or no change in the phosphor-

ylation of Ser21 or Thr345 was detected (data not shown). Inter-

estingly, expression of the cell cycle-dependent kinases Cdk1

and Cdk2, which have been demonstrated to phosphorylate

Ezh2 on threonine residues, mimicked that of Ezh2 and phos-

pho-Ezh2 (Figure 6A). Hence, it is plausible that phosphorylation

of Thr487, perhaps byCdk1 or Cdk2, activates or stabilizes Ezh2.

To investigate a more direct role for pregnancy hormones in

regulating Ezh2 and its phosphorylation, we treated mice in vivo

with progesterone or the peptide hormone prolactin that con-

tribute to the formation and differentiation of alveoli during

pregnancy (Oakes et al., 2008). It is noteworthy that PR levels

follow a similar pattern to that of Ezh2 and phospho-Ezh2, in

part reflecting epithelial content (Figure 6B). Analysis of mam-

mary glands following hormonal treatment showed that phos-

phorylated Ezh2 (Thr487) increased with progesterone but not

prolactin (Figure 6C). At 72 hr after progesterone treatment,

phospho-Ezh2 and Cdk1 were both substantially elevated (Fig-

ure 6D). Moreover, knockdown of PR in T-47D breast cancer

cells was accompanied by a decrease in Ezh2 and phospho-

Ezh2 levels (Figure 6E), suggesting that both transcriptional

and posttranslational mechanisms contribute.

To further examine the effects of progesterone on Ezh2 ex-

pression in different cellular compartments, we isolated discrete

subtypes of luminal progenitor cells that differ in their hormone

receptor status through the use of anti-CD49b and Sca-1 anti-

bodies (Li et al., 2009; Figure S6). Quantitative RT-PCR analysis

showed that the hormone receptor (HR)+ and HR� progenitor

populations contained cells highly enriched for PR+ and PR�

cells (Figure 6F). Interestingly, progesterone strongly induced

Ezh2 in the PR� subset, indicating a paracrine mode of stimula-

tion (Figure 6G). Conversely, Ezh1 expression was not induced

by progesterone (data not shown). As Rankl is a known target

of PR, we evaluated the expression of Rankl and the Rank

receptor in the three luminal subsets. Rankl was most highly

induced by progesterone in mature PR-positive ductal cells

(CD24+CD29loCD49b�Sca-1+), whereas Rank was abundantly

expressed in PR� luminal progenitor cells, suggesting that the

Rankl/Rank axis is an important paracrine mediator of proges-

terone-induced Ezh2 expression.

DISCUSSION

The epigenome is presumed to play a critical role in adult stem

cells and their progressive commitment to differentiated cells.

C

Indeed, elucidation of the genome-wide histone methylation

profiles of distinct mammary epithelial subtypes revealed that

H3K27me3-mediated epigenetic silencing is a key determinant

of gene expression during lineage restriction in the steady-state

mammary gland. Moreover, we found that pregnancy hormones

triggered striking genome-wide changes in H3K27me3 chro-

matin modifications in the expanding alveolar cell population.

Our elucidation of the physiological role of Ezh2 in controlling

mammary progenitor activity and alveolar development provides

direct evidence that this histone methylase has a critical role in

coordinating changes in the epigenome with regulatory gene

networks in response to hormonal stimuli.

The H3K4 and H3K27 trimethylated ‘‘landscapes’’ of the

mammary epithelium are highly dynamic in the steady-state

gland. Chromatin analyses combined with expression profiling

showed that these histone modifications tightly correlated with

transcriptional activity. In particular, H3K27me3 modifications

increased profoundly upon restriction of the MaSC/basal popu-

lation to the luminal lineage, suggesting that it contributes to

gene expression changes during lineage restriction. Histone

and DNA methylation patterns have been addressed in human

breast epithelial cells (CD44+ and CD24+), but these are not

directly comparable to the three cellular subsets defined in the

mouse mammary gland (Maruyama et al., 2011). Pertinently,

in another ectodermal lineage, the transition from activated

skin stem cells to committed transit amplifying cells involves

K27me3-PcG-mediated repression of stem cell genes and dere-

pression of PcG-silenced regulators (Lien et al., 2011). Given that

the MaSC/basal subset is heterogeneous at the cellular level,

although remarkably similar at the gene expression level (Stingl

et al., 2006; unpublished data), it remains unclear whether the

observed epigenetic changes in this population reflect those

occurring in MaSCs. The proportion of genes exhibiting bivalent

marks at their TSS also increased with restriction along the hier-

archy, with lowest levels in theMaSC/basal cell subset. It is inter-

esting to note that hair follicle stem cells contain few bivalent

marks (Lien et al., 2011).

Steroid hormones profoundly influence the mammary epige-

nome. Modulation of hormone levels via ovariectomy, preg-

nancy, or treatment with antiestrogen inhibitors strongly affects

both MaSCs and the luminal cell compartment (Asselin-Labat

et al., 2010). Hormone deprivation only modestly changed

H3K4me3 and H3K27me3 marking in both the stem cell-en-

riched and luminal populations, implying that other epigenetic

mechanisms may contribute to changes in gene expression.

However, the changes apparent in cell cycle regulators may

suffice to drive MaSCs into a more quiescent state, as indicated

by molecular profiling and cellular analyses (Asselin-Labat et al.,

2010). On the other hand, a striking role for H3K27me3

modifications was revealed during pregnancy, specifically in

the expanding alveolar luminal subset. In that subset, there

was a dramatic increase in the number of genes marked by

H3K27me3. Moreover, Ezh2 and H3K27-trimethylated protein

levels mirrored each other during mammary gland develop-

ment, with highest levels in early to midpregnancy, returning

to low levels in the late stages of pregnancy. In parallel, a

decrease in H3K27me3 marks has been observed during the

differentiation of keratinocytes (Sen et al., 2008), neuronal cells

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 421

Page 12: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

4 w

eeks

8 w

eeks

6.5

dP12

.5 d

P16

.5 d

P

P-Ezh2T487

Cdk1

Cdk2

Tubulin

Ezh2

cKO 4 w

eeks

8 w

eeks

6.5

dP12

.5 d

P16

.5 d

P18

.5 d

P

PR

Tubulin

Ezh2

Ezh2

P-Ezh2T487

Tubulin

Pg (72 h)vehicle

D

Prlvehicle

Cdk1

P-Ezh2T487

Tubulin

P-Ezh2T487

Tubulin

Pgvehicle

CBA

E

Ezh2

siR

NA

PR s

iRN

A

RIS

C-fr

ee

Ezh2

Tubulin

PR

P-Ezh2T487

H

luminal cell expansion

breast cancer

gene repression

progesterone

Ezh2

P-Ezh2 (T487)

&

CDK1/2

PRC2

steady state

Ezh2

K27

me3 me m3 e3me m3 e3

GOil Pg

F

Ezh

2 (

rela

tive

to 1

8S r

RN

A)

0

3.5

0.5

2.5

1.5

4.5X10-2

ML LP(PR+)

LP(PR–)

Ran

k (

rela

tive

to 1

8S r

RN

A)

0

1.2X10-5

0.8

0.4

0.2

0.6

1.0

ML LP(PR+)

LP(PR–)

Ran

kL (

rela

tive

to 1

8S r

RN

A)

0

4

2

1

5

6

3

7

ML LP(PR+)

LP(PR–)

PR

(re

lativ

e to

18S

rR

NA

)

0

2.5X10-2

2.0

1.0

0.5

1.5

ML LP(HR+)

LP(HR–)

Ezh2

Ezh2

K27 K27 K27 K27P P

Figure 6. Pregnancy Hormones Induce Phosphorylation of Ezh2 and Expression of Cdk-1/2, with Progesterone as an Important Mediator

(A) Western blot analysis of whole gland cell lysates for total Ezh2, phospho-Ezh2(Thr 487), Cdk1, and Cdk2 during mammary gland development. dP, days

pregnancy; cKO corresponds to MMTV-cre;Ezh2f/f at 12.5 dP. Antitubulin provided the loading control.

(legend continued on next page)

422 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 13: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

(Mikkelsen et al., 2007), and skeletal muscle (Caretti et al.,

2004).

Our findings indicate that upregulation of Ezh2 in pregnancy is

a key event that coordinates changes in the chromatin land-

scape with modulation of gene expression. The critical role of

Ezh2 in governing the activity of progenitor cells appears to

underlie its requirement for driving expansion of the alveolar

cell compartment. Notably, Ezh2 deficiency in the mammary

gland led to a global decrease in H3K27me3 levels in pregnancy,

indicating that it is a key methyl-transferase for H3K27. Ezh1

and/or demethylases may also contribute to the overall level of

H3K27me3 protein in the mammary epithelia. It is noteworthy

that targeted deletion of Eed, which is required for the function

of both Ezh1 and Ezh2, yielded a similar phenotype to that

following Ezh2 ablation (S. Orkin, K.B., and J.E.V., unpublished

data).

Ezh2 is also essential for the activity of basal and luminal

progenitor cells in the mammary gland during puberty. In Ezh2-

deficient mice, mammary morphogenesis was severely compro-

mised but could be effectively rescued through recurrent estrus

cycling by 15 weeks of age. Notably, mammary repopulating

activity was reduced 14-fold upon loss of Ezh2. Whether this

reflects a defect in the stem cell itself or in bipotent or committed

progenitor cells is uncertain. Pertinently, Ezh2-deficient progen-

itor cells in the MaSC/basal and luminal subsets had almost no

clonogenic activity in vitro. The cell cycle GO groups were

substantially affected by Ezh2 loss, consistent with Ezh2 func-

tioning as a silencer of cell cycle inhibitors in progenitor cells

and thus preventing exit from the cell cycle. Indeed, targeted

disruption of Ezh2 has demonstrated that it is required for the

proliferation of myogenic and skin basal progenitors, in part

due to repression of the Ink4a/Arf locus in the skin (Ezhkova

et al., 2009) and Ink4a in muscle stem cells (Juan et al., 2011).

The Ink4a/Arf locus is also likely to be a key mediator of Ezh2-

dependent cell cycle regulation in mammary progenitor cells,

given the reciprocal expression of Ezh2 and Arf. As a regulator

of proliferation, Ezh2 also plays an essential role in lymphopoie-

sis (Su et al., 2003), adipogenesis (Wang et al., 2010), postnatal

cardiac homeostasis (Delgado-Olguın et al., 2012), and hair

follicle morphogenesis in adult skin (Ezhkova et al., 2011). In

the latter case, however, targeted deletion of both Ezh1 and

Ezh2 was required to perturb hair follicle homeostasis.

Cell-fate decisions and differentiation in the mammary epithe-

lium do not appear to require Ezh2. Its absence did not affect

(B) Western blot analysis of PR and Ezh2 expression in whole mammary gland ly

(C) Western blot analysis of phospho-Ezh2(Thr 487) expression in whole gland lys

for 16 hr. No induction by prolactin was observed 24 hr posttreatment (data not

(D) Western blot analysis of total Ezh2, phospho-Ezh2(Thr 487), and Cdk1 express

(E) Knockdown of PR in human T-47D cells using small interfering RNAs (siRNAs)

Western blot analysis of PR, Ezh2, phospho-Ezh2(T487), and tubulin expression

(F) Quantitative RT-PCR analysis of PR expression in steady-state populations

Sca-1+) and hormone receptor-negative luminal progenitor (CD24+CD29loCD49

CD29, CD24, Sca1, and CD49b, as in Figure S6. n = 3 independent experiments

(G) Quantitative RT-PCR analysis of Ezh2, Rank, and Rankl expression in mature l

vehicle for 48 hr (n = 3; error bars show SEM). Note the induction of Ezh2 in the

(H) Schematic model of the role of progesterone in coordinating changes in the e

during pregnancy. Continual exposure to progesterone is hypothesized to result

See also Figures S5 and S6.

C

expression of mammary lineage markers, including alveolar

markers, and those required for milk synthesis. Although Ezh2-

deficient glands could produce milk, lactation failed due to the

low density of alveolar units. Furthermore, Ezh2 levels decline

precipitously in late pregnancy and lactation when terminal

differentiation occurs. Indeed, downregulation of Ezh2 prior to

lactation (when progesterone levels fall) may serve as a switch

to mediate derepression of genes required for lactogenesis (Ru-

dolph et al., 2003), including lipid biosynthesis genes, as sug-

gested by our bioinformatic analysis. The role of Ezh2 is thus

distinct from that of the polycomb family member Bmi1, which

represses alveolar differentiation in the adult mammary gland

(Pietersen et al., 2008a). However, polycomb-mediated repres-

sion of broad lineage determination programs can prevent the

activation of ‘‘extraneous’’ differentiation networks. Expression

profiling of Ezh2-deficient epithelial cells revealed inappropriate

expression of genes within the epidermal differentiation complex

(EDC), which is normally expressed in keratinocytes. The EDC

comprises six late-differentiation genes, including involucrin.

Interestingly, increased numbers of involucrin-positive luminal

cells appeared in Ezh2-deficient glands compared to control

glands (data not shown). Thus, Ezh2 normally represses expres-

sion of EDC genes in the mammary epithelium. In parallel, non-

skin lineage genes are also silenced in hair stem cells by

H3K27me3 (Ezhkova et al., 2011; Lien et al., 2011), and unsched-

uled expression of nonmyogenic lineage genes has been ob-

served in Ezh2-null skeletal muscle (Juan et al., 2011).

Ezh2 phosphorylation may be an important mechanism

by which hormones influence mammary epithelial expansion

during pregnancy. Phosphorylation of Ezh2 on threonine resi-

due 487 and levels of the cell cycle kinases Cdk1 and Cdk2

increased markedly during pregnancy, mimicking the pattern

for H3K27me3 protein. Both Cdk1 and Cdk2 can phosphorylate

Ezh2, and Cdk1 is positively associated with cell proliferation

(Malumbres and Barbacid, 2009). Furthermore, Cdk-mediated

phosphorylation of Ezh2 during cell cycle progression has

emerged as an important mechanism regulating Ezh2 function.

Thr345-phosphorylated Ezh2 appears to orchestrate epigenetic

silencing by promoting the recruitment of PRC2 to Ezh2 targets

(Chen et al., 2010; Kaneko et al., 2010). However, the role of Ezh2

phosphorylation on Thr487 remains unclear, since Thr487 phos-

phorylation impaired Ezh2 methyltransferase activity in one

study (Wei et al., 2011) but did not correlate with Ezh2 loading

at target genes in two other studies (Kaneko et al., 2010; Wu

sates from mice at different developmental time points.

ates from mice treated in vivo with progesterone (Pg), prolactin (Prl), or vehicle

shown). Tubulin provided a loading control.

ion in whole gland lysates frommice treated in vivo with Pg or vehicle for 72 hr.

. Cells were transfected with siGenome Smartpools for RISC-free, PR, or Ezh2.

was performed 60 hr following transfection.

of ML, hormone receptor-positive luminal progenitor (CD24+CD29loCD49b+

b+Sca-1�). Lineage-negative cells were fractionated using a combination of

; error bars show SEM.

uminal and progenitor subsets isolated from mice treated with progesterone or

PR� LP subset.

pigenome with gene expression changes in the alveolar luminal compartment

in epigenetic changes that culminate in breast cancer.

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 423

Page 14: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

and Zhang, 2011). Our data suggest that Thr487 phosphorylation

is more likely to promote Ezh2 methyltransferase activity in the

mammary gland, since K27-trimethylated H3, total Ezh2, and

phospho-Ezh2 levels were tightly correlated in pregnancy.

Progesterone appears to regulate Ezh2 through different

mechanisms. Acute treatment of mice with progesterone but

not prolactin led to a striking elevation in total Ezh2 and phos-

phorylated protein, suggesting that progesterone influences

the epigenetic ‘‘landscape’’ in pregnancy through modulating

the level of Ezh2 and its activity. Notably, the upregulation of

Ezh2 transcript levels occurred predominantly in the PR-nega-

tive luminal progenitor subset, implying a paracrine regulatory

mechanism. The Rank-Rankl signaling axis emerged as a likely

paracrine mediator, as the progesterone-target Ranklwas highly

induced in mature luminal cells, while Rank was most abundant

in PR-negative progenitors, in which Ezh2 expression was pro-

foundly stimulated. The Rank pathway has also been implicated

in paracrine signaling toMaSCs (Asselin-Labat et al., 2010; Joshi

et al., 2010). The dynamic expression pattern of phosphorylated

(T487) Ezh2 protein during mammary ontogeny suggests that

posttranslational mechanisms also regulate Ezh2 activity. In-

deed, it is tempting to speculate that Cdk1 (or 2)-mediated phos-

phorylation of Ezh2 is an important mechanism for integrating

cues from progesterone with chromatin modifications in order

to regulate the balance between proliferation and differentiation

in the developing gland (Figure 6H). Progesterone may also

directly influence cell cycle progression by receptor-mediated

recruitment of the cyclin A/Cdk2 complex to progesterone-

responsive promoters (Narayanan et al., 2005). Interestingly,

phosphorylation-mediated repression by Ezh2 occurs in a cell

stage-specific manner during the differentiation of satellite cells

to myotubes in response to signals from regenerating muscle

(Palacios et al., 2010).

Overexpression of EZH2 is a marker of poor prognosis and

metastatic disease in many solid carcinomas, including breast

(Bachmann et al., 2006; Bracken et al., 2003; Kleer et al., 2003;

Pietersen et al., 2008b; Raaphorst et al., 2003) and prostate

(Varambally et al., 2008), both of which are hormone-associated

malignancies. Notably, BRCA1-associated basal-like breast

tumors are selectively dependent on high levels of EZH2, thus

rendering them sensitive to the small molecule inhibitor DZNep

(Puppe et al., 2009). Moreover, EZH2 promotes the expansion

of breast tumor-initiating cells, possibly through the repression

of DNA damage repair pathways (Chang et al., 2011; Zeidler

et al., 2005). EZH2 overexpression is most frequently observed

in basal-like breast cancers. In these cancers, aberrant EZH2

expression may occur early in the oncogenic process and

within luminal progenitor cells that are exposed to sustained

progesterone signaling before they transition to a hormone

receptor-independent state. Interestingly, in utero exposure to

diethylstilbestrol may also influence breast cancer risk through

Ezh2 (Doherty et al., 2010). Our findings that Ezh2 coordinates

signals from progesterone with global changes in the mammary

epigenome have important implications for cancer, given the

impact of ovarian hormones on breast tumorigenesis. Specifi-

cally, the data suggest that sustained hormone exposure may

initiate oncogenesis through dysregulated histone methylation

of chromatin.

424 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

EXPERIMENTAL PROCEDURES

Mouse Strains and Hormone Treatment

Floxed Ezh2 mice were a kind gift from Dr. A. Tarakovsky (Su et al., 2003), and

the MMTV–cre (line A) mice were a gift from K.-U. Wagner (Eppley Institute,

Omaha, NE). MMTV-cre mice were maintained as a pure strain on a FVB/N

background. Floxed or deleted Ezh2 mice were analyzed on either a FVB/N

or mixed FVB/N C57/Bl6 background. For in vivo hormonal treatment,

8-week-old FVB/N female mice were injected intrascapularly daily with 1 mg

progesterone in 200 ml peanut oil or vehicle alone for either 16 or 72 hr or

prolactin (12.5 mg/kg) intraperitoneally for 16 hr. Prolactin (gift from the

National Hormone and Peptide Program, NIDDK) was dissolved at 1 mg/ml

in 0.1% BSA in PBS. The inguinal mammary glands (minus lymph node)

were harvested and snap-frozen. All animal experiments conform to regulatory

standards and were approved by the Walter and Eliza Hall Institute Animal

Ethics Committee.

Mammary Cell Preparation, Cell Sorting, and Transplantation

Mammary epithelial cell suspensions from female mice and flow cytometry

were performed as described (Shackleton et al., 2006). Cells sorted by flow

cytometry were manually counted and transplanted at limiting dilution as

described. Whole-mounting of glands, BrdU-labeling, immunohistochemistry,

and western blotting are described in Extended Experimental Procedures.

Cell Culture and Retroviral-Mediated Infection

For primary cell culture, freshly sorted mammary cells were plated on irradi-

ated fibroblast feeder layers (3,000 Rads) on collagen-coated, six-well plates

and infected as described (Bouras et al., 2008).

ChIP Sample Preparation and Sequencing

Freshly sorted cells were crosslinked with 1% paraformaldehyde and the ChIP

assay performed according to themanufacturer’s protocol (Millipore #17-371).

Briefly, cells were lysed and the Diagenode Bioruptor used for chromatin

shearing to a size range of 200 to 400 bp. Sheared chromatin was diluted

and incubated at 4�C overnight with antibodies against Histone H3 trimethyl

Lys4 (Millipore #07-473), Histone H3 trimethyl Lys27 (Millipore #07-449),

Histone H3 dimethyl Lys9 (Abcam #ab1220), or mouse isotype control (Milli-

pore #12-371). Immune complexes were handled as per the manufacturer’s

protocol.

Libraries for paired-end sequencing were prepared by GeneWorks

(Adelaide, Australia) using 10 ng ChIP DNA according to the Illumina ChIP-

seq Sample Preparation protocol, revision A, with the following modifications.

Paired-end adapters were substituted for standard genomic adapters and

library amplification (18 cycles) performed using primers PE 1.0 and 2.0.

Size selection for 300 bp DNA was performed prior to amplification. Final

libraries were analyzed using the Agilent Bioanalyzer High Sensitivity DNA

Kit and sequenced on the Illumina Genome Analyzer IIx running SCS 2.8,

generating 35 base paired-end reads using the CASAVA 1.7 analysis pipeline.

The statistical analyses are described in the Extended Experimental Proce-

dures. Sequence data have been submitted to the Gene Expression Omnibus

under accession number GSE43212.

ACCESSION NUMBERS

The GEO database accession number for the ChIP-seq data is GSE43212.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Extended Experimental Procedures, two

tables, and six figures and can be found with this article online at http://dx.

doi.org/10.1016/j.celrep.2012.12.020.

LICENSING INFORMATION

This is an open-access article distributed under the terms of the Creative

Commons Attribution-NonCommercial-No Derivative Works License, which

Page 15: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

permits non-commercial use, distribution, and reproduction in any medium,

provided the original author and source are credited.

ACKNOWLEDGMENTS

We are very grateful to A. Tarakovsky for providing floxed Ezh2 mice and J.

Adams for critical review of the manuscript. We also thank GeneWorks for

library preparation and sequencing of ChIP samples; D. Wu for help with bio-

informatic analysis; E. Nolan, T. McLennan, B. Capaldo, and T.Ward for expert

help; C. Clarke, K. Simpson, D. Reinberg, G. McArthur, and B. Sarcevic for

reagents; and the animal, FACS, and histology facilities at WEHI. We thank

C. Clarke, J. Carroll, and S. Clark for discussions. This work was supported

by the Australian National Health and Medical Research Council (NHMRC)

#461224, #461221, #637307, #637308, #1016701, and Australia Fellowship

(to J.E.V.); NHMRC #490037 (to G.K.S.); the WEHI Genomics Fund; NHMRC

IRIISS; the Victorian State Government through VCA funding of the Victorian

Breast Cancer Research Consortium and Operational Infrastructure Support;

and the ACRF. B.P. and T.B. were supported by NHMRC Biomedical and

National Breast Cancer Foundation (Australia) Fellowships, respectively.

Received: May 29, 2012

Revised: December 20, 2012

Accepted: December 28, 2012

Published: January 31, 2013

REFERENCES

Asselin-Labat, M.L., Sutherland, K.D., Barker, H., Thomas, R., Shackleton, M.,

Forrest, N.C., Hartley, L., Robb, L., Grosveld, F.G., van der Wees, J., et al.

(2007). Gata-3 is an essential regulator of mammary-gland morphogenesis

and luminal-cell differentiation. Nat. Cell Biol. 9, 201–209.

Asselin-Labat, M.L., Vaillant, F., Sheridan, J.M., Pal, B., Wu, D., Simpson, E.R.,

Yasuda, H., Smyth, G.K., Martin, T.J., Lindeman, G.J., and Visvader, J.E.

(2010). Control of mammary stem cell function by steroid hormone signalling.

Nature 465, 798–802.

Asselin-Labat, M.L., Sutherland, K.D., Vaillant, F., Gyorki, D.E., Wu, D., Hol-

royd, S., Breslin, K., Ward, T., Shi, W., Bath, M.L., et al. (2011). Gata-3 nega-

tively regulates the tumor-initiating capacity of mammary luminal progenitor

cells and targets the putative tumor suppressor caspase-14. Mol. Cell. Biol.

31, 4609–4622.

Bachmann, I.M., Halvorsen, O.J., Collett, K., Stefansson, I.M., Straume, O.,

Haukaas, S.A., Salvesen, H.B., Otte, A.P., and Akslen, L.A. (2006). EZH2

expression is associated with high proliferation rate and aggressive tumor

subgroups in cutaneousmelanoma and cancers of the endometrium, prostate,

and breast. J. Clin. Oncol. 24, 268–273.

Bernardo, G.M., Lozada, K.L., Miedler, J.D., Harburg, G., Hewitt, S.C., Mosley,

J.D., Godwin, A.K., Korach, K.S., Visvader, J.E., Kaestner, K.H., et al. (2010).

FOXA1 is an essential determinant of ERalpha expression and mammary

ductal morphogenesis. Development 137, 2045–2054.

Bernstein, B.E., Mikkelsen, T.S., Xie, X., Kamal, M., Huebert, D.J., Cuff, J., Fry,

B., Meissner, A., Wernig, M., Plath, K., et al. (2006). A bivalent chromatin

structure marks key developmental genes in embryonic stem cells. Cell 125,

315–326.

Bouras, T., Pal, B., Vaillant, F., Harburg, G., Asselin-Labat, M.L., Oakes, S.R.,

Lindeman, G.J., and Visvader, J.E. (2008). Notch signaling regulatesmammary

stem cell function and luminal cell-fate commitment. Cell Stem Cell 3,

429–441.

Bracken, A.P., and Helin, K. (2009). Polycomb group proteins: navigators of

lineage pathways led astray in cancer. Nat. Rev. Cancer 9, 773–784.

Bracken, A.P., Pasini, D., Capra, M., Prosperini, E., Colli, E., and Helin, K.

(2003). EZH2 is downstream of the pRB-E2F pathway, essential for prolifera-

tion and amplified in cancer. EMBO J. 22, 5323–5335.

Brisken, C., and O’Malley, B. (2010). Hormone action in the mammary gland.

Cold Spring Harb. Perspect. Biol. 2, a003178.

C

Brisken, C., Park, S., Vass, T., Lydon, J.P., O’Malley, B.W., andWeinberg, R.A.

(1998). A paracrine role for the epithelial progesterone receptor in mammary

gland development. Proc. Natl. Acad. Sci. USA 95, 5076–5081.

Caretti, G., Di Padova, M., Micales, B., Lyons, G.E., and Sartorelli, V. (2004).

The Polycomb Ezh2 methyltransferase regulates muscle gene expression

and skeletal muscle differentiation. Genes Dev. 18, 2627–2638.

Caretti, G., Palacios, D., Sartorelli, V., and Puri, P.L. (2011). Phosphoryl-EZH-

ion. Cell Stem Cell 8, 262–265.

Chang, C.J., Yang, J.Y., Xia, W., Chen, C.T., Xie, X., Chao, C.H., Woodward,

W.A., Hsu, J.M., Hortobagyi, G.N., and Hung, M.C. (2011). EZH2 promotes

expansion of breast tumor initiating cells through activation of RAF1-b-catenin

signaling. Cancer Cell 19, 86–100.

Chen, S., Bohrer, L.R., Rai, A.N., Pan, Y., Gan, L., Zhou, X., Bagchi, A., Simon,

J.A., and Huang, H. (2010). Cyclin-dependent kinases regulate epigenetic

gene silencing through phosphorylation of EZH2. Nat. Cell Biol. 12, 1108–

1114.

Delgado-Olguın, P., Huang, Y., Li, X., Christodoulou, D., Seidman, C.E., Seid-

man, J.G., Tarakhovsky, A., and Bruneau, B.G. (2012). Epigenetic repression

of cardiac progenitor gene expression by Ezh2 is required for postnatal cardiac

homeostasis. Nat. Genet. 44, 343–347.

Doherty, L.F., Bromer, J.G., Zhou, Y., Aldad, T.S., and Taylor, H.S. (2010). In

utero exposure to diethylstilbestrol (DES) or bisphenol-A (BPA) increases

EZH2 expression in the mammary gland: an epigenetic mechanism linking

endocrine disruptors to breast cancer. Horm. Cancer 1, 146–155.

Ezhkova, E., Pasolli, H.A., Parker, J.S., Stokes, N., Su, I.H., Hannon, G., Tara-

khovsky, A., and Fuchs, E. (2009). Ezh2 orchestrates gene expression for the

stepwise differentiation of tissue-specific stem cells. Cell 136, 1122–1135.

Ezhkova, E., Lien, W.H., Stokes, N., Pasolli, H.A., Silva, J.M., and Fuchs, E.

(2011). EZH1 and EZH2 cogovern histone H3K27 trimethylation and are essen-

tial for hair follicle homeostasis and wound repair. Genes Dev. 25, 485–498.

Graham, J.D., Mote, P.A., Salagame, U., van Dijk, J.H., Balleine, R.L., Huscht-

scha, L.I., Reddel, R.R., and Clarke, C.L. (2009). DNA replication licensing and

progenitor numbers are increased by progesterone in normal human breast.

Endocrinology 150, 3318–3326.

Hu, Y., and Smyth, G.K. (2009). ELDA: extreme limiting dilution analysis for

comparing depleted and enriched populations in stem cell and other assays.

J. Immunol. Methods 347, 70–78.

Joshi, P.A., Jackson, H.W., Beristain, A.G., Di Grappa, M.A., Mote, P.A.,

Clarke, C.L., Stingl, J.,Waterhouse, P.D., and Khokha, R. (2010). Progesterone

induces adult mammary stem cell expansion. Nature 465, 803–807.

Juan, A.H., Derfoul, A., Feng, X., Ryall, J.G., Dell’Orso, S., Pasut, A., Zare, H.,

Simone, J.M., Rudnicki, M.A., and Sartorelli, V. (2011). Polycomb EZH2

controls self-renewal and safeguards the transcriptional identity of skeletal

muscle stem cells. Genes Dev. 25, 789–794.

Kaneko, S., Li, G., Son, J., Xu, C.F., Margueron, R., Neubert, T.A., and Rein-

berg, D. (2010). Phosphorylation of the PRC2 component Ezh2 is cell cycle-

regulated and up-regulates its binding to ncRNA. Genes Dev. 24, 2615–2620.

Keller, P.J., Arendt, L.M., Skibinski, A., Logvinenko, T., Klebba, I., Dong, S.,

Smith, A.E., Prat, A., Perou, C.M., Gilmore, H., et al. (2012). Defining the cellular

precursors to human breast cancer. Proc. Natl. Acad. Sci. USA 109, 2772–

2777.

Kleer, C.G., Cao, Q., Varambally, S., Shen, R., Ota, I., Tomlins, S.A., Ghosh, D.,

Sewalt, R.G., Otte, A.P., Hayes, D.F., et al. (2003). EZH2 is a marker of aggres-

sive breast cancer and promotes neoplastic transformation of breast epithelial

cells. Proc. Natl. Acad. Sci. USA 100, 11606–11611.

Li, W., Ferguson, B.J., Khaled, W.T., Tevendale, M., Stingl, J., Poli, V., Rich, T.,

Salomoni, P., and Watson, C.J. (2009). PML depletion disrupts normal

mammary gland development and skews the composition of the mammary

luminal cell progenitor pool. Proc. Natl. Acad. Sci. USA 106, 4725–4730.

Lien, W.H., Guo, X., Polak, L., Lawton, L.N., Young, R.A., Zheng, D., and

Fuchs, E. (2011). Genome-wide maps of histone modifications unwind in vivo

chromatin states of the hair follicle lineage. Cell Stem Cell 9, 219–232.

ell Reports 3, 411–426, February 21, 2013 ª2013 The Authors 425

Page 16: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Lim, E., Wu, D., Pal, B., Bouras, T., Asselin-Labat, M.L., Vaillant, F., Yagita, H.,

Lindeman, G.J., Smyth, G.K., and Visvader, J.E. (2010). Transcriptome anal-

yses of mouse and human mammary cell subpopulations reveal multiple

conserved genes and pathways. Breast Cancer Res. 12, R21.

Lydon, J.P., DeMayo, F.J., Funk, C.R., Mani, S.K., Hughes, A.R., Montgomery,

C.A., Jr., Shyamala, G., Conneely, O.M., and O’Malley, B.W. (1995). Mice lack-

ing progesterone receptor exhibit pleiotropic reproductive abnormalities.

Genes Dev. 9, 2266–2278.

Mallepell, S., Krust, A., Chambon, P., and Brisken, C. (2006). Paracrine

signaling through the epithelial estrogen receptor alpha is required for prolifer-

ation and morphogenesis in the mammary gland. Proc. Natl. Acad. Sci. USA

103, 2196–2201.

Malumbres, M., and Barbacid, M. (2009). Cell cycle, CDKs and cancer:

a changing paradigm. Nat. Rev. Cancer 9, 153–166.

Margueron, R., and Reinberg, D. (2011). The Polycomb complex PRC2 and its

mark in life. Nature 469, 343–349.

Maruyama, R., Choudhury, S., Kowalczyk, A., Bessarabova, M., Beresford-

Smith, B., Conway, T., Kaspi, A., Wu, Z., Nikolskaya, T., Merino, V.F., et al.

(2011). Epigenetic regulation of cell type-specific expression patterns in the

human mammary epithelium. PLoS Genet. 7, e1001369.

Mikkelsen, T.S., Ku, M., Jaffe, D.B., Issac, B., Lieberman, E., Giannoukos, G.,

Alvarez, P., Brockman, W., Kim, T.K., Koche, R.P., et al. (2007). Genome-wide

maps of chromatin state in pluripotent and lineage-committed cells. Nature

448, 553–560.

Mueller, S.O., Clark, J.A., Myers, P.H., and Korach, K.S. (2002). Mammary

gland development in adult mice requires epithelial and stromal estrogen

receptor alpha. Endocrinology 143, 2357–2365.

Mulac-Jericevic, B., Lydon, J.P., DeMayo, F.J., and Conneely, O.M. (2003).

Defective mammary gland morphogenesis in mice lacking the progesterone

receptor B isoform. Proc. Natl. Acad. Sci. USA 100, 9744–9749.

Narayanan, R., Adigun, A.A., Edwards, D.P., and Weigel, N.L. (2005). Cyclin-

dependent kinase activity is required for progesterone receptor function: novel

role for cyclin A/Cdk2 as a progesterone receptor coactivator. Mol. Cell. Biol.

25, 264–277.

Nicol, J.W., Helt, G.A., Blanchard, S.G., Jr., Raja, A., and Loraine, A.E. (2009).

The Integrated Genome Browser: free software for distribution and exploration

of genome-scale datasets. Bioinformatics 25, 2730–2731.

Oakes, S.R., Rogers, R.L., Naylor, M.J., and Ormandy, C.J. (2008). Prolactin

regulation of mammary gland development. J. Mammary Gland Biol.

Neoplasia 13, 13–28.

Palacios, D., Mozzetta, C., Consalvi, S., Caretti, G., Saccone, V., Proserpio, V.,

Marquez, V.E., Valente, S., Mai, A., Forcales, S.V., et al. (2010). TNF/p38a/pol-

ycomb signaling to Pax7 locus in satellite cells links inflammation to the epige-

netic control of muscle regeneration. Cell Stem Cell 7, 455–469.

Pietersen, A.M., Evers, B., Prasad, A.A., Tanger, E., Cornelissen-Steijger, P.,

Jonkers, J., and van Lohuizen, M. (2008a). Bmi1 regulates stem cells and

proliferation and differentiation of committed cells in mammary epithelium.

Curr. Biol. 18, 1094–1099.

Pietersen, A.M., Horlings, H.M., Hauptmann, M., Langerød, A., Ajouaou, A.,

Cornelissen-Steijger, P., Wessels, L.F., Jonkers, J., van de Vijver, M.J., and

van Lohuizen, M. (2008b). EZH2 and BMI1 inversely correlate with prognosis

and TP53 mutation in breast cancer. Breast Cancer Res. 10, R109.

Puppe, J., Drost, R., Liu, X., Joosse, S.A., Evers, B., Cornelissen-Steijger, P.,

Nederlof, P., Yu, Q., Jonkers, J., van Lohuizen, M., and Pietersen, A.M. (2009).

BRCA1-deficient mammary tumor cells are dependent on EZH2 expression

and sensitive to PolycombRepressive Complex 2-inhibitor 3-deazaneplanocin

A. Breast Cancer Res. 11, R63.

426 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Raaphorst, F.M., Meijer, C.J., Fieret, E., Blokzijl, T., Mommers, E., Buerger, H.,

Packeisen, J., Sewalt, R.A., Otte, A.P., and van Diest, P.J. (2003). Poorly differ-

entiated breast carcinoma is associated with increased expression of the

human polycomb group EZH2 gene. Neoplasia 5, 481–488.

Robinson, G.W., and Hennighausen, L. (2011). MMTV-Cre transgenes can

adversely affect lactation: considerations for conditional gene deletion in

mammary tissue. Anal. Biochem. 412, 92–95.

Rudolph, M.C., McManaman, J.L., Hunter, L., Phang, T., and Neville, M.C.

(2003). Functional development of the mammary gland: use of expression

profiling and trajectory clustering to reveal changes in gene expression during

pregnancy, lactation, and involution. J. Mammary Gland Biol. Neoplasia 8,

287–307.

Sauvageau, M., and Sauvageau, G. (2010). Polycomb group proteins: multi-

faceted regulators of somatic stem cells and cancer. Cell Stem Cell 7,

299–313.

Sen, G.L.,Webster, D.E., Barragan, D.I., Chang, H.Y., and Khavari, P.A. (2008).

Control of differentiation in a self-renewing mammalian tissue by the histone

demethylase JMJD3. Genes Dev. 22, 1865–1870.

Shackleton, M., Vaillant, F., Simpson, K.J., Stingl, J., Smyth, G.K., Asselin-

Labat, M.L., Wu, L., Lindeman, G.J., and Visvader, J.E. (2006). Generation of

a functional mammary gland from a single stem cell. Nature 439, 84–88.

Sleeman, K.E., Kendrick, H., Robertson, D., Isacke, C.M., Ashworth, A., and

Smalley, M.J. (2007). Dissociation of estrogen receptor expression and in vivo

stem cell activity in the mammary gland. J. Cell Biol. 176, 19–26.

Statham, A.L., Strbenac, D., Coolen, M.W., Stirzaker, C., Clark, S.J., and Rob-

inson, M.D. (2010). Repitools: an R package for the analysis of enrichment-

based epigenomic data. Bioinformatics 26, 1662–1663.

Stingl, J., Eirew, P., Ricketson, I., Shackleton, M., Vaillant, F., Choi, D., Li, H.I.,

and Eaves, C.J. (2006). Purification and unique properties of mammary epithe-

lial stem cells. Nature 439, 993–997.

Su, I.H., Basavaraj, A., Krutchinsky, A.N., Hobert, O., Ullrich, A., Chait, B.T.,

and Tarakhovsky, A. (2003). Ezh2 controls B cell development through histone

H3 methylation and Igh rearrangement. Nat. Immunol. 4, 124–131.

Van Keymeulen, A., Rocha, A.S., Ousset, M., Beck, B., Bouvencourt, G., Rock,

J., Sharma, N., Dekoninck, S., and Blanpain, C. (2011). Distinct stem cells

contribute to mammary gland development and maintenance. Nature 479,

189–193.

Varambally, S., Cao, Q., Mani, R.S., Shankar, S., Wang, X., Ateeq, B., Laxman,

B., Cao, X., Jing, X., Ramnarayanan, K., et al. (2008). Genomic loss of micro-

RNA-101 leads to overexpression of histone methyltransferase EZH2 in

cancer. Science 322, 1695–1699.

Visvader, J.E. (2009). Keeping abreast of themammary epithelial hierarchy and

breast tumorigenesis. Genes Dev. 23, 2563–2577.

Wang, L., Jin, Q., Lee, J.E., Su, I.H., and Ge, K. (2010). Histone H3K27 meth-

yltransferase Ezh2 represses Wnt genes to facilitate adipogenesis. Proc. Natl.

Acad. Sci. USA 107, 7317–7322.

Wei, Y., Chen, Y.H., Li, L.Y., Lang, J., Yeh, S.P., Shi, B., Yang, C.C., Yang, J.Y.,

Lin, C.Y., Lai, C.C., and Hung, M.C. (2011). CDK1-dependent phosphorylation

of EZH2 suppresses methylation of H3K27 and promotes osteogenic differen-

tiation of human mesenchymal stem cells. Nat. Cell Biol. 13, 87–94.

Wu, S.C., and Zhang, Y. (2011). Cyclin-dependent kinase 1 (CDK1)-mediated

phosphorylation of enhancer of zeste 2 (Ezh2) regulates its stability. J. Biol.

Chem. 286, 28511–28519.

Zeidler, M., Varambally, S., Cao, Q., Chinnaiyan, A.M., Ferguson, D.O., Mer-

ajver, S.D., and Kleer, C.G. (2005). The Polycomb group protein EZH2 impairs

DNA repair in breast epithelial cells. Neoplasia 7, 1011–1019.

Page 17: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Supplemental Information

EXTENDED EXPERIMENTAL PROCEDURES

Mouse StrainsAll animal experiments were conducted using mice bred at and maintained in our animal facility according to institutional and the

Melbourne Health Research Directorate Animal Ethics Committee guidelines. Ezh2 mice were on a pure FVB/N or mixed C57Bl/6

and FVB/N background; the same results were obtained for each. Adult female mice were subjected to timed pregnancies, scored

by the presence of vaginal plugs and confirmed by examination of embryos on collection of mammary glands. Mice were genotyped

using the primers listed below.

Mammary Cell Preparation and Cell SortingAntibodies against mouse antigens were purchased from BD PharMingen (San Diego, CA) unless otherwise specified, and included

CD24-PE, biotinylated CD31, CD45 and Ter119, CD29–FITC (Chemicon, Temecula, CA), CD61-APC and streptavidin–APC–Cy7,

CD14-biotin (eBioscience, San Diego, CA). For the luminal progenitor fractionation experiments, CD49b-FITC, Sca1-APC, CD45-

PECy-7 and CD31-PECy-7 were from eBioscience, and CD29-APC-Cy7 from Biolegend (San Diego, CA). Single cell suspensions

were sorted on a FACSAria or FACSDiva (BD PharMingen).

Histology and Whole MountingFor histological examination of mouse mammary glands, tissues were fixed in 4% paraformaldehyde overnight and embedded in

paraffin. Sections (5 mm) were prepared and stained with hematoxylin and eosin (H&E). For whole-mount analysis, mammary glands

were harvested and fixed in Carnoy’s solution (six parts 100% ethanol, three parts chloroform, one part glacial acetic acid) and

stained with hematoxylin.

Bromodeoxyuridine ImmunodetectionMicewere injected with BrdUCell Labeling Reagent (0.5mg/10 g body weight, AmershamBiosciences) 1 hr prior to tissue collection.

Tissueswere fixed in 4%paraformaldehyde and embedded in paraffin. For immunohistochemical detection of BrdU-labeled cells, rat

anti-BrdU (Becton Dickinson) and biotinylated rabbit anti-rat IgG antibody (Dako) were used, followed by HRP-conjugated strepta-

vidin (Dako, LSAB2).

Cell Culture and Retroviral-Mediated InfectionFor primary cell culture, freshly sorted mammary epithelial cells were plated on irradiated fibroblast feeders on collagen-coated

6-well plates and infected as described (Bouras et al., 2008). Colony assays in 2D have been described (Shackleton et al., 2006).

Primary cells following transduction with cre-MIG (MSCV-cre-IRES-GFP) or empty retrovirus were manually counted after sorting

for GFP and replated at 200 or 300 cells per well in a 24-well plate containing a feeder layer of irradiated fibroblasts. After 7 days,

cultures were fixed with ice-cold acetone/methanol and stained with Giemsa for enumeration.

For siRNA-mediated knockdown of PR in T-47D cells, the following siGENOME SMARTpools (Dharmacon) were used: Ezh2 (hu)

M-004218-03-0005, RISC-free D-001220-01 and progesterone receptor (hu) M-003433-01-0005. Transfections were performed as

described by the manufacturer (Dharmacon) using Dharmafect-1 and cells harvested for analysis after 60 hr.

ImmunohistochemistryParaffin-embedded sections (5 mm) were dewaxed in xylene and rehydrated through an alcohol series, blocked with 3% hydrogen

peroxide, and subjected to antigen retrieval by boiling in 10 mM citrate buffer pH 6.0 for 30 s at pressure using a DAKO pressure

cooker. The mouse-on-mouse (MOM) kit (Vector) was used for mousemonoclonal antibodies. Immunostaining with other antibodies

was performed using the streptavidin-biotin peroxidase detection system as per the manufacturer’s instructions (ABC reagent,

Vector Laboratories). 3,3-diaminobenzidine was used as substrate (DAKO). In all cases, an isotype-matched control IgG was

used as a negative control. The following antibodies were used: anti-SMA (Sigma), anti-milk (Accurate Chemical and Scientific),

anti-p63 (BD PharMingen), anti-keratin 18 (Progen Biotechnik), anti-keratin 14 (Covance), anti-BrdU (Becton Dickinson), anti-cas-

pase-3, anti-Ezh2 (Lake Placid Biologicals, Active Motif). Secondary antibodies were biotin-conjugated anti-rabbit IgG and anti-

mouse IgG (Vector Laboratories). The Npt2b antibody was a kind gift of Dr Juerg Biber from the University of Zurich, Switzerland.

Western Blot AnalysisWhole mammary gland lysates were prepared in RIPA buffer (50 mM Tris pH7.4, 150 mM NaCl, 0.25% Na-deoxycholate, 1% NP40,

1mMPMSF, 1X Roche complete mini protease inhibitor cocktail, 1X Roche PhosSTOP phosphatase inhibitor cocktail). The following

antibodies were used for western blot analysis: anti-H3K27me3 (Millipore #07-449); anti-Cdk1 (Cell Signaling #9112); anti-Cdk2: (Cell

Signaling #2546); anti-Ezh2 (BD Biosciences #612666); anti-phospho-Ezh2-Thr487 (Abcam #ab109398); anti-phospho-Ezh2-Ser21

(Abcam #ab84989); anti-tubulin (Sigma #T9026); anti-progesterone receptor Ab-7 was kindly provided by C. Clarke.

ChIP-seq Read MappingReads were mapped to the mouse reference genome mm9 using the Subread aligner (http://sourceforge.net/projects/subread).

A fragment was judged to be successfully mapped if the paired ends mapped to locations between 50 and 500 bp apart.

Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors S1

Page 18: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Genome-wide heatmaps of methylation mark coverage were drawn using Repitools (Statham et al., 2010). Density plots were

drawn using in-house scripts written in R (http://www.r-project.org). Coverage graphs were generated for genes of interest using

the Integrated Genome Browser (Nicol et al., 2009).

ChIP-seq Statistical AnalysisFragment counts were formed for each gene. Fragments were counted if the center of the fragment was contained in the TSS or

broad region for that gene. Statistical analysis of the count data was performed using edgeR package (Robinson et al., 2010) of

the Bioconductor software project (Gentleman et al., 2004). The Benjamini-Hochberg method was used to control the FDR. Genes

were called as significantly enriched for each histone mark using the normalizeChIPtoInput function, which normalizes the ChIP

counts to input and evaluates enrichment using a negative binomial statistical model. Log2-fold-changes in histone mark coverage

between cell populations or between conditions were computed using the predFC function. This function adjusted the ChIP counts

for input by fitting a negative binomial log-linear generalized linear model (McCarthy et al., 2012), using a prior count of 0.5 per sample

to avoid unreliably large log-fold-changes that might otherwise arise from zero or small counts. The negative binomial dispersion was

set to 0.01 for all calculations, allowing for a degree of biological variation typical of mouse experiments (McCarthy et al., 2012). Gene

Ontology analysis used the DAVID tool (Huang da et al., 2009). Gene set enrichment used the mean-rank gene set enrichment test

(Michaud et al., 2008).

RNA Extraction and Quantitative RT-PCR AnalysisTotal RNA was isolated from primary mammary epithelial subpopulations with the RNeasy Micro kit (QIAGEN). Reverse transcription

by using oligo(dT) primer and Moloney murine leukemia virus reverse transcriptase (Invitrogen) was according to the manufacturer’s

protocol. Quantitative RT-PCR was carried out by using a Rotorgene RG-6000 (Corbett Research) and SensiMix (dT) DNA kit

(Quantace) under the following conditions: 10 min at 95�C followed by 35 cycles consisting of 15 s at 95�C, 20 s at 62�C, and20 s at 72�C. Gene expression was determined with the Rotor-Gene software (version 1.7). The primer sequences are listed below.

Microarray AnalysisMicroarray expression data for steady-state epithelial cell subsets, pregnant and ovariectomized mice were analyzed as described

previously (Asselin-Labat et al., 2010; Lim et al., 2010). For Ezh2-deficient profiles, cell subsets were sorted from three independent

mouse pools, and up to 250 ng of RNA per sample was labeled, amplified and hybridized to Illumina Mouse-WG6 V2 Expression

BeadChips according to Illumina standard protocols at the Australian Genome Research Facility (Melbourne, Australia). Summary

probe profiles were exported from GenomeStudio and was analyzed using the limma software package (Smyth, 2004). Expression

values were normexp background adjusted and quantile normalized using control probes (Shi et al., 2010). The data have been

uploaded into GSE38203. Probes were filtered if not expressed (detection p-value > 0.05 across all arrays) or poorly annotated

(Barbosa-Morais et al., 2010). Differential expression between between Ezh2-deficient and control cells was assessed using linear

models and empirical Bayes moderated t-statistics (Smyth, 2004). Reliability was improved using array quality weights (Ritchie et al.,

2006), and each mouse pool was treated as a random effect to allow for dependence between samples from the same pool

(Smyth et al., 2005). Gene ontology enrichment for biological process (BP) terms was carried out on the top 500 genes from each

contrast using the GOstats package (Falcon and Gentleman, 2007). Focused gene set testing using Ezh2 signatures obtained

from Kamminga et al. (2006) (34 genes matched by gene symbols) and Ezhkova et al. (2009) (65 genes matched by gene symbols)

were performed using the roast method (Wu et al., 2010). Microarray probes werematched to ChIP-seq profiles by gene symbol. The

probe with highest average expression data was chosen to represent each gene.

Genes with FDR < 0.05 and at least 50% fold-change were selected as Ezh2-deficient signature genes and compared with the

human breast cancer data set from Herschkowitz et al. (2007). Where multiple probes were present for the same gene on a given

platform, the probe with the highest average expression level was kept for further analysis. Genes were matched between platforms

usingGene symbols and a signature score calculated for each sample as in Lim et al. (2010). TheEzh2-deficient signature geneswere

also compared across tumor subtypes using roast gene set tests (Wu et al., 2010) with Ezh2–deficient log-fold changes as gene

weights.

Oligonucleotides Used in the StudyGenotyping oligonucleotides (50 to 30)

Ezh2

Fwd 1: TTATTCATAGAGCCACCTGG

Fwd 2: ACGAAACAGCTCCAGATTCAGGG

Rev: CTGCTCTGAATGGCAACTCC

ChIP-q-RT-PCR oligonucleotides

c-Kit

Fwd: TCAGGGGTGCCACGATCCGT

Rev: TAGTCGGGATTGCCGGGCGA

S2 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 19: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Lgr5

Fwd: TATCAAAGCCTCAAAGAAATC

Rev: GTGTTCTTCCAAGGTGTCTGA

Snai2

Fwd: TGTCATCAGCCGGTGGACTTCCT

Rev: TCCAAGGACCCAGGGGTTGTG

Hey2

Fwd: TTGGCTTGCCCAGAAGCACCT

Rev: TGCGCAGCTCAGCCTGTTTAG

Krt5

Fwd: GGCCTGTGACCTGTGAGGGACA

Rev: TCTCCTCCAGAGGTTGCCCCA

DNp63

Fwd: GAGTCCCGCCCCTCATGCCT

Rev: CTGAGAGCCTTGCGCTGCGA

Elf5

Fwd: GAGCCCCGACACCCCCTTTCA

Rev: TACAGTCCGCTGGTGCTGGGA

Wap

Fwd: AGCCACACCCGGTAGTAAGGTG

Rev: CTGGAGGTGGCCCTCGCTCA

Csn2

Fwd: ACAAGGCAGCAATTCAGAAGCTGGT

Rev: CCCCGGTCCTCTCACTTGGC

FoxA1

Fwd: CTAGCGCCACCCAGCGGTC

Rev: GTCGGTGCTCGCTTACCGGG

Wnt7b

Fwd: AGGCTGGGCTAACAGAGACCCC

Rev: GGAAGGACCTGGGTGCCCGA

Hey1

Fwd: CACAGCTCGCTTCGCTCCTGT

Rev: TGGCGTCAAGGGAGGCAGGT

Cdkn2a

Fwd: CAGCCGGTAAGAAGGGTTCACCT

Rev: GCTACCCGATAGCAAGCACTAGGA

Cdkn1c

Fwd: GCTGGCCCTAAGACCCTCTA

Rev: ATGGGCCCAACTTGTGTCTC

Camk2n1

Fwd: ACCTACGGGTAGAGACCCAG

Rev: GGGCTTTACCTTCAGTTGCC

Ccnd2

Fwd: GCCTCGGCCACGCAGGAAAA

Rev: ACGCTCCGCGCAGACACCTA

Quantitative RT-PCR oligonucleotides

Elf5

Fwd: CCCTGAATACTGGACCAAGC

Rev: GCTGCCTCAATGAACTCCTC

Wap

Fwd: TGC CTC ATC AGC CTC GTT CTT G

Rev: CTG GAG CAT TCT ATC TTC ATT GGG

Csn2

Fwd: AAA GGA CTT GAC AGC CAT GAA

Rev: TAG CCT GGA GCA CAT CCT CT

Lgr5

Fwd: CCA ATG GAA TAA AGA CGA CGG CAA CA

Rev: GGG CCT TCA GGT CTT CCT CAA AGT CA

Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors S3

Page 20: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Arf

Fwd: CGCAGGTTCTTGGTCACTGTGAGG

Rev: TGCCCATCATCATCACCTGGTCC

Rankl

Fwd: TGTACTTTCGAGCGCAGATG

Rev: CCACAATGTGTTGCAGTTCC

Rank

Fwd: ACACCCTGCCTCCTGGGCTT

Rev: AAGCCTGGGCCTCCTTGGGT

PR

Fwd: GCTTGCATGATCTTGTGAAACAGC

Rev: GGAAATTCCACAGCCAGTGTCC

Ezh2

Fwd: GCAATTTAGAAAACGGAAATGC

Rev: GTACAAAACACTTTGCAGCTGG

18S rRNA

Fwd: GTAACCCGTTGAACCCCATT

Rev: CCATCCAATCGGTAGTAGCG

SUPPLEMENTAL REFERENCES

Barbosa-Morais, N.L., Dunning, M.J., Samarajiwa, S.A., Darot, J.F., Ritchie, M.E., Lynch, A.G., and Tavare, S. (2010). A re-annotation pipeline for Illumina

BeadArrays: improving the interpretation of gene expression data. Nucleic Acids Res. 38, e17.

Falcon, S., and Gentleman, R. (2007). Using GOstats to test gene lists for GO term association. Bioinformatics 23, 257–258.

Gentleman, R.C., Carey, V.J., Bates, D.M., Bolstad, B., Dettling, M., Dudoit, S., Ellis, B., Gautier, L., Ge, Y., Gentry, J., et al. (2004). Bioconductor: open software

development for computational biology and bioinformatics. Genome Biol. 5, R80.

Herschkowitz, J.I., Simin, K., Weigman, V.J., Mikaelian, I., Usary, J., Hu, Z., Rasmussen, K.E., Jones, L.P., Assefnia, S., Chandrasekharan, S., et al. (2007).

Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol. 8, R76.

Huang da, W., Sherman, B.T., and Lempicki, R.A. (2009). Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat.

Protoc. 4, 44–57.

Kamminga, L.M., Bystrykh, L.V., de Boer, A., Houwer, S., Douma, J., Weersing, E., Dontje, B., and de Haan, G. (2006). The Polycomb group gene Ezh2 prevents

hematopoietic stem cell exhaustion. Blood 107, 2170–2179.

McCarthy, D.J., Chen, Y., and Smyth, G.K. (2012). Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation.

Nucleic Acids Res. 40, 4288–4297.

Michaud, J., Simpson, K.M., Escher, R., Buchet-Poyau, K., Beissbarth, T., Carmichael, C., Ritchie, M.E., Schutz, F., Cannon, P., Liu, M., et al. (2008). Integrative

analysis of RUNX1 downstream pathways and target genes. BMC Genomics 9, 363.

Ritchie, M.E., Diyagama, D., Neilson, J., van Laar, R., Dobrovic, A., Holloway, A., and Smyth, G.K. (2006). Empirical array quality weights in the analysis of

microarray data. BMC Bioinformatics 7, 261.

Robinson, M.D., McCarthy, D.J., and Smyth, G.K. (2010). edgeR: a Bioconductor package for differential expression analysis of digital gene expression data.

Bioinformatics 26, 139–140.

Shi, W., Oshlack, A., and Smyth, G.K. (2010). Optimizing the noise versus bias trade-off for Illumina whole genome expression BeadChips. Nucleic Acids Res. 38,

e204.

Smyth, G.K. (2004). Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3,

Article 3.

Smyth, G.K., Michaud, J., and Scott, H.S. (2005). Use of within-array replicate spots for assessing differential expression in microarray experiments.

Bioinformatics 21, 2067–2075.

Vikstrom, I., Carotta, S., Luthje, K., Peperzak, V., Jost, P.J., Glaser, S., Busslinger, M., Bouillet, P., Strasser, A., Nutt, S.L., and Tarlinton, D.M. (2010). Mcl-1 is

essential for germinal center formation and B cell memory. Science 330, 1095–1099.

Wu, D., Lim, E., Vaillant, F., Asselin-Labat, M.L., Visvader, J.E., and Smyth, G.K. (2010). ROAST: rotation gene set tests for complex microarray experiments.

Bioinformatics 26, 2176–2182.

S4 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 21: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

H3K4me3 H3K27me3H3K9me2

100%

50%

Expression

-5 TSS +5

Score

Luminal Progenitor (LP) cells Mature Luminal (ML) cells

4 8 12 4 8 12H3K4me3 H3K27me3H3K9me2

-5 TSS +5 -5 TSS +5 -5 TSS +5 -5 TSS +5 -5 TSS +5

1.0

0.5

0

1.5

TSS 5kb 10kb-5kb-10kb TSS 5kb 10kb-5kb-10kb TSS 5kb 10kb-5kb-10kb

0.10

0.05

0

0.150.10

0.05

0

0.150.20

High expression

MaSC/basal

Luminal progenitor

TSS 5kb 10kb-5kb-10kb TSS 5kb 10kb-5kb-10kb TSS 5kb 10kb-5kb-10kb

Mature luminal

Low expression

TSS 5kb 10kb-5kb-10kb TSS 5kb 10kb-5kb-10kb TSS 5kb 10kb-5kb-10kb

0.04

0.02

0.06

0.10

0.08

0.12

0

0.04

0.02

0.06

0.10

0.08

0.12

0

1.0

0.5

0

1.5

0.04

0.02

0.06

0.10

0.08

0.12

0

1.0

0.5

0

1.5

0.04

0.02

0.06

0.10

0.08

0.12

0

0.14

Ave

rage

cov

erag

eA

vera

ge c

over

age

Ave

rage

cov

erag

e

H3K4me3 H3K9me2 H3K27me3

H3K4me3 H3K9me2 H3K27me3

H3K4me3 H3K9me2 H3K27me3

100%

50%

Score

Expression

A

B

Figure S1. Genome-Wide Heatmaps of Methylation Profiles for Luminal Progenitor and Mature Luminal Subpopulations in the ‘‘Steady

State’’, Related to Figures 1 and 2

(A) Heat maps for luminal progenitor (left) and mature luminal cells (right) show distribution of H3K4me3, H3K9me2, and H3K27me3 marks ± 5 kb of the TSS of

each gene. Genes are clustered into groups according to their histonemethylation profiles and ordered within groups by expression level. Far right column shows

log2-expression. Other columns show percentage coverage.

(B) Density plots of average histone methylation coverage stratified by gene expression. The x-axes show distance from TSS in base pairs. The y-axis shows

average number of covering reads at each base pair. For each cell subset, genes are stratified into four equal-size groups by expression level, and the average

coverage is shown for each group. Lines are red to green from high to low expression. Plots are shown for each epigenetic mark in each epithelial cell subset.

H3K4me3 marks peak sharply around the TSS and directly correlate with gene expression, whereas H3K27me3 shows a broad pattern and reverse association

with expression.

Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors S5

Page 22: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

Wap

Elf5 Csn2

Lgr5

Fol

d ch

ange

vs

Inpu

t

virgin 12.5 dP

virgin 12.5dp

virgin 12.5 dP

virgin 12.5 dP

virgin

12.5 dP

1.0

0.5

0.75

1.2

0.4

0.8

1.6

2.0

0.5

1.0

2.5

1.5virgin

12.5 dP

virgin

12.5 dP

virgin

12.5 dP

Fol

d ch

ange

vs

Inpu

t F

old

chan

ge v

s In

put

0.25

Fol

d ch

ange

vs

Inpu

t

1.0

0.5

0.75

0

0.25

0

0

0

Csn2Elf5

MaSC Luminal MaSC Luminal

Wap

MaSC Luminal

Lgr5

MaSC Luminal

0.35

0.05

0.15

0.45

0.25

0.014

0.002

0.006

0.018

0.010

0.35X101.0X102

0.05

0.25

0.2

0.4

0.6

0.8

0.15

00 0 0

A

B

8 wk virgin 12.5 dP

Exp

ress

ion

rela

tive

to 1

8S r

RN

A

control

Ovx

Ovx

Ccnd2

Cdkn2a

Ovx

control Ovx

4

2

3

1

0

1.6

0.8

1.2

0.4

0

control

control

TSS

TSS

Fol

d ch

ange

vs

Inpu

t F

old

chan

ge v

s In

put

C

OVXvirgin 12.5 dP1.0

0.8

0.6

0.4

0.2

0

MaSC/basal Luminal

1.0

0.8

0.6

0.4

0.2

0

OVXvirgin 12.5 dP

H3K4me3 and H3K27me3H3K27me3 onlyH3K4me3 onlyNo H3K4me3/H3K27me3

D

Figure S2. Genome-Wide Histone Modification Changes in Mammary Epithelial Cells from Pregnant and Ovariectomized Mice, Related to

Figure 3

(A) Read coverage maps and corresponding ChIP-qRT-PCR for H3K27me3 marks on Ccnd2 and Cdkn2a (Ink4a/arf) in the MaSC/basal population. n = 3

independent biological samples for each, error bars represent SEM.

(B) Pregnancy induces derepression of luminal genes in the MaSC-enriched subset invoking ‘‘lineage-priming’’. Quantitative RT-PCR was performed to

determine the levels of b-casein (Csn2), Wap, Elf-5, and Lgr5 mRNA in the MaSC/basal and luminal subsets isolated from virgin (8 weeks old) and 12.5 day

pregnant (12.5 dP) glands of FVB/N mice (n = 3, error bars represent SEM).

(C) Read coverage maps and corresponding ChIP-qRT-PCR for H3K27me3 marks on three luminal genes (Elf5, Csn2, Wap) and the MaSC/basal-specific

gene Lgr5 in the MaSC/basal population. Data are shown for virgin and 12.5 day pregnant (12.5 dP) mice. Coverage graphs show fragments per million on the

scale 0–10. ChIP-qRT-PCR was performed on three independent biological samples; error bars represent SEM.

(D) Genome-wide proportions of genes with histone methylation marks in pregnant or ovariectomized mice. Segmented bar graphs show the percentages of all

genes significantly marked (FDR < 0.05) with H3K4me3 and/or H3K27me3 for MaSC/basal or luminal subsets from virgin, ovariectomized, or pregnant mice.

S6 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 23: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

A

E

C

K18

Ezh2f/+ MMTV-cre; Ezh2f/f

p63

D

MM

TV

-cre

; Ezh

2f/f

6 wk 8 wk

8 wk

Ezh

2f/+

lactation (8 d)

F

Brd

U p

ositi

ve c

ells

per

TE

B

4

0

14

12

8

10

6

2

16Ezh2f/+

MMTV-cre; Ezh2f/f

GMMTV-cre; Ezh2f/f

2 dL

MMTV-cre

Ezh

2f/+

12 wk 6 wk

MM

TV

-cre

; Ezh

2f/f

B

MM

TV

-cre

; Ezh

2f/+

16.5 dP12.5 dP

hCD

4

FSC-A FSC-A

FSC-A FSC-A

Lum MaSC/basal

MM

TV

-cre

; Mcl

-1/h

CD

4 (f

/+)

Mcl

-1/h

CD

4 (f

/+) 1.74% 0.20%

83.5% 99.0%

hCD

496.9% 99.3%

12.7% 0.91%

Figure S3. Delayed Mammary Morphogenesis in Ezh2-Deficient Mice, Related to Figures 4 and 5

(A) Efficient MMTV-cre-mediated deletion in luminal and basal mammary epithelial cells based on hCD4 reporter mice. FloxedMcl-1-hCD4 reporter mice in which

human CD4 surface expression is activated upon cre-mediated excision of Mcl-1, thus serving as a reporter of Mcl-1 deletion (Vikstrom et al., 2010). Mcl-1 is

expressed throughout luminal and myoepithelial cells in the mammary gland (unpublished data). MMTV-cre induced effective deletion of the floxed Mcl-1-hCD4

reporter in the luminal (83%) andMaSC/basal (99%) subsets, respectively. MMTV-cre mediated deletion in GTRosa26 reporter mice confirmed activity of MMTV

in mammary epithelial cells but not stroma (data not shown).

(B) Whole-mounted mammary glands from virgin MMTV–cre; Ezh2f/f mice compared to Ezh2f/+ littermate controls. Stunted development was evident in

Ezh2-deficient glands at 6 weeks but generally not at 12 weeks of age. Scale bars, 2.0 mm.

(C) Cell fate appears unchanged in Ezh2-deficient mammary glands. Immunostaining for lineage markers in mammary gland tissue sections from virgin 8-week

old MMTV–cre; Ezh2f/f mice compared to Ezh2f/+ controls. Scale bars, 50 mm.

(D) Npt2b immunostaining of sections fromMMTV–cre; Ezh2f/f glands at 6 and 8weeks of age did not reveal premature alveolar differentiation in the ducts or TEBs

(top panels). A small minority of cells (<2%) were positive for Npt2B staining (middle right panel). Immunostaining of mammary gland sections from Ezh2f/+ mice

for Npt2b at 8 days of lactation (bottom left panel) serves as a positive control. Red = Npt2b, Green = E-cadherin, Blue = DAPI. Scale bars, 50 mm.

(E) Histogram showing the number of BrdU-positive cells per TEB in pubertal mammary glands from MMTV–cre; Ezh2f/f mice compared to Ezh2f/+ mice (n = 3

mice; error bars represent SEM).

(F) H&E sections of mammary glands from MMTV–cre; Ezh2f/+ mice at days 12.5 and 16.5 of pregnancy. Scale bars: 50 mm.

(G) H&E sections of mammary glands at day two of lactation from MMTV–cre; Ezh2f/f and MMTV–cre mice. Scale bars: 100 mm.

Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors S7

Page 24: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

A

B

CD

29hi

CD

24+

crecontrolCD29hi CD29lo controls

WT cKOcre - -+ +

WTΔ/Δ

C

GOBPID

GO:0007049GO:0022402GO:0000278GO:0022403GO:0051301GO:0000087GO:0000280GO:0007067GO:0048285GO:0000279GO:0016043GO:0019751GO:0050896GO:0006974GO:0051716GO:0006261GO:0006260GO:0006259GO:0006270GO:0014902GO:0031424GO:0033554GO:0006996GO:0006020

P-value

2.1E-111.3E-101.6E-097.4E-098.0E-098.0E-098.0E-098.0E-091.3E-087.7E-083.3E-055.4E-051.0E-041.7E-042.3E-042.4E-042.6E-042.9E-044.2E-044.2E-044.2E-047.4E-047.5E-048.7E-04

GO Term

cell cyclecell cycle processmitotic cell cyclecell cycle phasecell divisionM phase of mitotic cell cyclenuclear divisionmitosisorganelle fi ssionM phasecellular component organizationpolyol metabolic processresponse to stimulusresponse to DNA damage stimuluscellular response to stimulusDNA-dependent DNA replicationDNA replicationDNA metabolic processDNA replication initiationmyotube differentiationkeratinizationcellular response to stressorganelle organizationinositol metabolic process

GOBPID

GO:0022403GO:0022402GO:0007049GO:0000278GO:0000279GO:0000087GO:0000280GO:0007067GO:0048285GO:0051301GO:0006996GO:0006259GO:0006260GO:0016043GO:0071103GO:0006974GO:0006281GO:0051276GO:0006323GO:0007017GO:0006261GO:0033554GO:0006270GO:0030261

P-value

3.50E-341.49E-331.81E-317.35E-318.94E-309.35E-289.35E-289.35E-282.97E-274.25E-241.41E-203.81E-198.78E-161.33E-125.55E-114.00E-101.00E-093.45E-094.58E-096.55E-081.45E-072.27E-077.52E-077.52E-07

GO Term

cell cycle phasecell cycle processcell cyclemitotic cell cycleM phaseM phase of mitotic cell cyclenuclear divisionmitosisorganelle fi ssioncell divisionorganelle organizationDNA metabolic processDNA replicationcellular component organizationDNA conformation changeresponse to DNA damage stimulusDNA repairchromosome organizationDNA packagingmicrotubule-based processDNA-dependent DNA replicationcellular response to stressDNA replication initiationchromosomal condensation

MaSC/basal Luminal

MMTV-cre;Ezh2f/f

Ezh2f/+

Luminal (12.5 dP)

0

1.2X103

1.0

0.8

0.6

0.2

0.4

0

4.5X103

3.5

2.5

1.5

0.5

MaSC/basalLuminal

virgin 12.5 dP

(re

lativ

e to

18S

rR

NA

)

Arf

virgin 12.5 dP

Ezh

2-de

fi cie

nt s

igna

ture

(M

aSC

/bas

al)

Lum

A

clau

din-

low

HER

2

Lum

B

Basa

l

D

Figure S4. Derepression of Cell Cycle Genes in Ezh2-Deficient Mammary Glands, Related to Figure 5

(A) MaSC/basal (CD29hiCD24+, denoted CD29hi) and luminal (CD29loCD24+, denoted CD29lo) populations fromEzh2f/f micewere transducedwith cre-expressing

or empty control retrovirus in 2D cultures and harvested after 72 hr. PCR analysis of genomic DNA confirmed excision. The top and bottom bands represent the

wild-type and floxed alleles respectively. Ex vivo cre-mediated excision of Ezh2 in the MaSC/basal (and luminal subsets) severely impaired clonogenic capacity.

Shown here are data for the MaSC/basal (CD29hi) population.

(B) Microarray analysis of the MaSC/basal (CD29hi) and luminal (CD29lo) subsets from control (undeleted) and Ezh2-deficient cells following ex vivo excision

revealed significant enrichment of gene signatures related to cell cycle regulation. Comparative analyses of the Ezh2 signature genes fromKamminga et al. (2006)

and Ezhkova et al. (2009) with our gene expression profiles showed that the signatures were significantly differentially expressed in the basal and luminal subsets:

1) for upregulated genes in the MaSC/basal and luminal subsets, p = 0.0009 and 0.0003 respectively (Kamminga et al., 2006) and 2) for up or downregulated

genes in the MaSC/basal and luminal subsets, p = 0.0009 and 0.0099 respectively.

(C) Quantitative RT-PCR analysis of Arf expression in cellular subsets from virgin versus pregnant glands (left panel). qRT-PCR showed that Arf is derepressed in

Ezh2-deficient luminal cells isolated from 12.5 day pregnant mice (right panel) (n = 3 independent experiments; error bars represent SEM).

(D) Ezh2 transcriptional signature by breast cancer tumor subtype. Box plots show the aggregate gene expression score in each tumor for genes associated

with Ezh2-deficiency in the MaSC/basal subset. The Ezh2-deficient expression score is highest in the claudin-low subtype and lowest in the basal and luminal

B subtypes. Gene set testing (Wu et al., 2010) confirms that these comparisons are statistically significant (p = 0.0002 for claudin-low versus basal subtypes and

p = 0.0027 for claudin-low versus luminal B).

S8 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors

Page 25: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

A

MM

TV

-cre

; Ezh

2f/f

CD14+ CD14–CD29hi

Col

onie

s pe

r 10

0 ce

lls

10

0

40

30

20

CD14+

CD29lo

CD24+

CD29hi

CD24+

Ezh2f/+

MMTV-cre; Ezh2f/f

B

C

MMTV-cre; Ezh2f/f

CD

24

CD29

CD

24

CD14

CD

24

CD29

CD

24

MMTV-cre; Ezh2f/f

CD14

CD

24

CD29

CD

24

CD14

CD

24

CD29

CD

24

CD14

virg

inpr

egna

nt

CD14_

CD29lo

CD24+

Ezh

2f/+

Ezh2f/+

Ezh2f/+

H3K27Me3

Tubulin

4 w

k

8 w

k6.

5 d

12.5

d16

.5 d

6 w

k

Ezh2

f/+

Virgin Pregnancy

cKO

12.5 dP

D

8 w

k

6 w

k

Ezh2f/+

8 w

k8

wk

H3K27me3

Tubulin

Virgin

MMTV-cre;Ezh2f/f

Figure S5. Ezh2 Deficiency Dramatically Reduces Progenitor Activity among the Epithelial Subsets during Pregnancy, Related to Figure 6

(A) CD24/CD29 and CD24/CD14 flow cytometric plots of lineage-negative mammary epithelial cells (CD45�CD31�Ter119�) from MMTV–cre; Ezh2f/f mice and

Ezh2+/f littermate controls in either the virgin or pregnant (day 12.5) states. CD14 was used to subdivide the luminal population since CD61 is rapidly down-

regulated during pregnancy. In virgin glands, CD14 expression enriches for luminal progenitor activity whereas the CD14� subset is enriched for mature luminal

cells (Asselin-Labat et al., 2011). In pregnancy, the spectrum of CD14 expression is broader, with progenitor activity detectable in all subsets, thus suggesting

a continuum of alveolar and ductal progenitor cells. Ezh2-deficiency leads to diminution of all progenitor activity.

(B) Colony forming capacity of freshly sorted CD29hiCD24+MaSC/basal (denoted CD29hi), CD29loCD24+CD14+ (denoted CD14+), and CD29loCD24+CD14�

(denoted CD14�) luminal subfractions from mid-pregnant MMTV–cre; Ezh2f/f mice or Ezh2f/+ mice, grown on irradiated fibroblast feeders.

(C) Histogram showing the colony forming capacity of each subpopulation. Data represent the mean of two independent biological experiments with eight

replicates for each.

(D) Global diminution of H3K27me3 protein in Ezh2-deficient mammary epithelial cells: Western blot analyses of total H3K27me3 protein and tubulin in lysates

from Ezh2-deficient (MMTV-cre; Ezh2f/f) versus littermate control mammary glands (Ezh2f/+) (upper panel). Loss of trimethylated H3K27 protein in Ezh2-deficient

glands at 12.5 days pregnancy (lower panel, right lane).

Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors S9

Page 26: Cell Reports Article Reports Article Global Changes in the Mammary Epigenome Are Induced by Hormonal Cues and Coordinated by Ezh2 Bhupinder Pal,1,3 Toula Bouras,1,3,8 Wei Shi,2,5,8

FSC-H

FS

C-W

SSC-HS

SC

-WFSC-A

PI

FSC-A

Line

age

Sca

1-A

PC

CD49b-FITC

CD

24-P

E

CD29-APC-Cy7

Sca

1-A

PC

CD49b-FITCC

D24

-PE

CD29-APC-Cy7

Oil Pg

Mat Lum Lum Prog (PR+)

Lum Prog (PR–)

Figure S6. Isolation of Hormone Receptor-Positive and Negative Luminal Progenitor Populations from Young Adult Mammary Glands,

Related to Figure 6

CD24/CD29 and CD49b/Sca-1 flow cytometric plots of lineage-negative mammary epithelial cells (CD45�CD31�) from 8 week-old mice. CD49b/Sca-1 flow

cytometric plots are shown for lineage-negative CD29loCD24+ cells derived from mice treated with progesterone or vehicle (oil) for 48 hr. The three luminal

subsets used for qRT-PCR analysis are depicted: mature luminal, PR+, and PR� luminal progenitor cells.

S10 Cell Reports 3, 411–426, February 21, 2013 ª2013 The Authors