estrogen-dependent gene transcription in human breast ... · assessing the molecular mechanisms...

15
Molecular and Cellular Pathobiology Estrogen-Dependent Gene Transcription in Human Breast Cancer Cells Relies upon Proteasome-Dependent Monoubiquitination of Histone H2B Tanja Prenzel 1 , Yvonne Begus-Nahrmann 1 , Frank Kramer 2 , Magali Hennion 1 , Chieh Hsu 4 , Theresa Gorsler 1 , Corinna Hintermair 5 , Dirk Eick 5 , Elisabeth Kremmer 6 , Mikael Simons 3,4 , Tim Beissbarth 2 , and Steven A. Johnsen 1 Abstract The estrogen receptor-a (ERa) determines the phenotype of breast cancers where it serves as a positive prognostic indicator. ERa is a well-established target for breast cancer therapy, but strategies to target its function remain of interest to address therapeutic resistance and further improve treatment. Recent findings indicate that proteasome inhibition can regulate estrogen-induced transcription, but how ERa function might be regulated was uncertain. In this study, we investigated the transcriptome-wide effects of the proteasome inhibitor bortezomib on estrogen-regulated transcription in MCF7 human breast cancer cells and showed that bortezomib caused a specific global decrease in estrogen-induced gene expression. This effect was specific because gene expression induced by the glucocorticoid receptor was unaffected by bortezomib. Surprisingly, we observed no changes in ERa recruitment or assembly of its transcriptional activation complex on ERa target genes. Instead, we found that proteasome inhibition caused a global decrease in histone H2B monoubiquitina- tion (H2Bub1), leading to transcriptional elongation defects on estrogen target genes and to decreased chromatin dynamics overall. In confirming the functional significance of this link, we showed that RNA interferencemediated knockdown of the H2B ubiquitin ligase RNF40 decreased ERa-induced gene transcrip- tion. Surprisingly, RNF40 knockdown also supported estrogen-independent cell proliferation and activation of cell survival signaling pathways. Most importantly, we found that H2Bub1 levels decrease during tumor progression. H2Bub1 was abundant in normal mammary epithelium and benign breast tumors but absent in most malignant and metastatic breast cancers. Taken together, our findings show how ERa activity is blunted by bortezomib treatment as a result of reducing the downstream ubiquitin-dependent function of H2Bub1. In supporting a tumor suppressor role for H2Bub1 in breast cancer, our findings offer a rational basis to pursue H2Bub1-based therapies for future management of breast cancer. Cancer Res; 71(17); 573953. Ó2011 AACR. Introduction The estrogen receptor-a (ERa) is a ligand-activated tran- scription factor which plays an essential role in a number of physiologic processes, including development, fertility, cardiovascular function, and bone metabolism (1), by inducing rapid and dramatic changes in gene expression. Importantly, over two thirds of breast tumors express ERa and depend upon its activity for their growth (1, 2). As such, antiestrogen therapies such as tamoxifen, fulvestrant (Faslo- dex), and aromatase inhibitors have become standard treat- ments for ERa-positive breast cancer. Nevertheless, about one third of ERa-positive breast cancers become resistant to antiestrogen therapy and develop into more aggressive hormone-independent tumors (3). Thus, more effective therapies which act on additional aspects of ERa function may help to eliminate ERa-positive tumors at an earlier stage and increase overall patient survival (4). One potential target of breast cancer therapy may be the ubiquitinproteasome system (UPS; ref. 5). The conjugation of multiple ubiquitin moieties (polyubiquitination) linked to one another through lysine residue 48 (K48) of ubiquitin generally lead to the degradation of the target protein by the 26S proteasome (6). Ubiquitination is carried out by ubiquitin ligases which frequently contain either RING or HECT Authors' Affiliations: 1 Department of Molecular Oncology, Gottingen Cen- ter for Molecular Biosciences, Departments of 2 Medical Statistics and 3 Neurology, University Medical Center Gottingen; 4 Max-Planck-Institute for Experimental Medicine, Gottingen; 5 Department of Molecular Epige- netics, Helmholtz Center for Environmental Health and Center of Integrated Protein Science Munich (CIPSM); and 6 Institute of Molecular Immunology, Helmholtz Center for Environmental Health, Munich, Germany Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Y. Begus-Nahrmann and F. Kramer contributed equally to this work. Current address for C. Hsu: Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland. Corresponding Author: Steven A. Johnsen, Department of Molecular Oncology, Göttingen Center for Molecular Biosciences, University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077 Gottingen, Germany. Phone: 49-551-39-10373; Fax: 49-551-39-13713; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-11-1896 Ó2011 American Association for Cancer Research. Cancer Research www.aacrjournals.org 5739 Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association for https://bloodcancerdiscov.aacrjournals.org Downloaded from

Upload: others

Post on 13-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

Molecular and Cellular Pathobiology

Estrogen-Dependent Gene Transcription in Human BreastCancer Cells Relies upon Proteasome-DependentMonoubiquitination of Histone H2B

Tanja Prenzel1, Yvonne Begus-Nahrmann1, Frank Kramer2, Magali Hennion1, Chieh Hsu4,Theresa Gorsler1, Corinna Hintermair5, Dirk Eick5, Elisabeth Kremmer6, Mikael Simons3,4,Tim Beissbarth2, and Steven A. Johnsen1

AbstractThe estrogen receptor-a (ERa) determines the phenotype of breast cancers where it serves as a positive

prognostic indicator. ERa is a well-established target for breast cancer therapy, but strategies to target itsfunction remain of interest to address therapeutic resistance and further improve treatment. Recent findingsindicate that proteasome inhibition can regulate estrogen-induced transcription, but how ERa function mightbe regulated was uncertain. In this study, we investigated the transcriptome-wide effects of the proteasomeinhibitor bortezomib on estrogen-regulated transcription in MCF7 human breast cancer cells and showed thatbortezomib caused a specific global decrease in estrogen-induced gene expression. This effect was specificbecause gene expression induced by the glucocorticoid receptor was unaffected by bortezomib. Surprisingly, weobserved no changes in ERa recruitment or assembly of its transcriptional activation complex on ERa targetgenes. Instead, we found that proteasome inhibition caused a global decrease in histone H2B monoubiquitina-tion (H2Bub1), leading to transcriptional elongation defects on estrogen target genes and to decreasedchromatin dynamics overall. In confirming the functional significance of this link, we showed that RNAinterference–mediated knockdown of the H2B ubiquitin ligase RNF40 decreased ERa-induced gene transcrip-tion. Surprisingly, RNF40 knockdown also supported estrogen-independent cell proliferation and activation ofcell survival signaling pathways. Most importantly, we found that H2Bub1 levels decrease during tumorprogression. H2Bub1 was abundant in normal mammary epithelium and benign breast tumors but absentin most malignant and metastatic breast cancers. Taken together, our findings show how ERa activity is bluntedby bortezomib treatment as a result of reducing the downstream ubiquitin-dependent function of H2Bub1. Insupporting a tumor suppressor role for H2Bub1 in breast cancer, our findings offer a rational basis to pursueH2Bub1-based therapies for future management of breast cancer. Cancer Res; 71(17); 5739–53. �2011 AACR.

Introduction

The estrogen receptor-a (ERa) is a ligand-activated tran-scription factor which plays an essential role in a number

of physiologic processes, including development, fertility,cardiovascular function, and bone metabolism (1), byinducing rapid and dramatic changes in gene expression.Importantly, over two thirds of breast tumors express ERaand depend upon its activity for their growth (1, 2). As such,antiestrogen therapies such as tamoxifen, fulvestrant (Faslo-dex), and aromatase inhibitors have become standard treat-ments for ERa-positive breast cancer. Nevertheless, aboutone third of ERa-positive breast cancers become resistantto antiestrogen therapy and develop into more aggressivehormone-independent tumors (3). Thus, more effectivetherapies which act on additional aspects of ERa functionmay help to eliminate ERa-positive tumors at an earlierstage and increase overall patient survival (4).

One potential target of breast cancer therapy may be theubiquitin–proteasome system (UPS; ref. 5). The conjugation ofmultiple ubiquitin moieties (polyubiquitination) linked to oneanother through lysine residue 48 (K48) of ubiquitin generallylead to the degradation of the target protein by the 26Sproteasome (6). Ubiquitination is carried out by ubiquitinligases which frequently contain either RING or HECT

Authors' Affiliations: 1Department of Molecular Oncology, G€ottingen Cen-ter for Molecular Biosciences, Departments of 2Medical Statistics and3Neurology, University Medical Center G€ottingen; 4Max-Planck-Institutefor Experimental Medicine, G€ottingen; 5Department of Molecular Epige-netics, Helmholtz Center for Environmental Health and Center of IntegratedProtein Science Munich (CIPSM); and 6Institute of Molecular Immunology,Helmholtz Center for Environmental Health, Munich, Germany

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

Y. Begus-Nahrmann and F. Kramer contributed equally to this work.

Current address for C. Hsu: Institute of Molecular Life Sciences, Universityof Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.

Corresponding Author: Steven A. Johnsen, Department of MolecularOncology, Göttingen Center for Molecular Biosciences, UniversityMedical Center Göttingen, Justus-von-Liebig-Weg 11, 37077 G€ottingen,Germany. Phone: 49-551-39-10373; Fax: 49-551-39-13713; E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-11-1896

�2011 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 5739

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 2: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

domains. Several ubiquitin ligases, such as RNF12 (7), havebeen shown to bind directly to ERa and regulate its activity.Furthermore, a number of studies have suggested that protea-some inhibition alters ERa-dependent gene transcription,although different effects and mechanisms have been sug-gested (8–12). Proposed mechanisms of action for proteasomeinhibition included decreased ERa mobility within thenucleus (10, 11) and decreased ERa binding and recruitmentof cofactors (10). In addition, changes in RNA polymerase(RNAP) II phosphorylation have been observed followingproteasome inhibition (13). However, most of these studiesutilized the laboratory proteasome inhibitor MG132, which isnot suitable for clinical use, and were mostly based on theinvestigation of a limited number of target genes.

Recently, the proteasome inhibitor bortezomib (Velcade)was shown to be safe for clinical use (14) and was approved bythe Food and Drug Administration for the treatment of multi-ple myeloma andmantle cell lymphoma (15). Although clinicaltrials for the treatment of metastatic breast cancer wereinitially disappointing when used as a single agent (14), anew study combining the pure antiestrogen fulvestrant withbortezomib suggests that the combination of antiestrogentherapies with proteasome inhibition may indeed increasetreatment efficacy (16). In line with these results, clinical trialsutilizing the combination of bortezomib and fulvestrant arecurrently underway for the treatment of ER-positive meta-static tumors in postmenopausal patients (NCT01142401).Therefore, a more complete understanding of the effectsof proteasome inhibition on ERa activity are essential forassessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;ERa-positive vs. ERa-negative) may respond better to protea-some inhibitors such as bortezomib.

In this study, we sought to determine the effects of theclinically safe proteasome inhibitor bortezomib on the rapidinduction of estrogen-regulated gene transcription in MCF7breast cancer cells. We provide evidence that bortezomibblocks estrogen-induced gene transcription by a unique post-transcriptional initiation mechanism involving a loss of his-tone H2B monoubiquitination (H2Bub1) and changes inchromatin dynamics. Moreover, we observe a dramatic lossof H2Bub1 during tumor progression which may lead toestrogen-independent cell proliferation and increased meta-static properties.

Methods

Cell culture, transfections, and siRNAsMCF7 cells were obtained from K. Pantel (University Hos-

pital Hamburg-Eppendorf, Hamburg, Germany) and A549 cellswere obtained from M. Dobbelstein (University of G€ottingen).Both cell lines were grown in phenol red–free high-glucoseDulbecco's Modified Eagle's Media (DMEM; Invitrogen) sup-plemented with 10% bovine growth serum (Thermo Scienti-fic). The identity of both the cell lines was routinely verifiedmicroscopically on the basis of cellular morphology and theexpression of ERa and estrogen responsiveness (for MCF7cells). Prior to hormone treatments, MCF7 and A549 cells were

grown in DMEM containing 5% charcoal-dextran–treated FBS(CSS; HyClone) 1 to 2 days prior to treatment with 10 nmol/L17b-estradiol (Sigma-Aldrich) or 100 nmol/L dexamethasone(Sigma-Aldrich), respectively, as indicated. For blocking pro-teasome activity, cells were pretreated for 15 minutes with 1 ofthe 3 following chemical proteasome inhibitors: 50 nmol/Lbortezomib (LC Laboratories), 20 mmol/L MG132 (Biomol), or1 mmol/L epoxomicin (Biomol). MCF10A cells were obtainedfromM. Oren (Weizmann Institute of Science, Rehovot, Israel)and grown in DMEM/F-12 medium supplemented with 5%horse serum (Sigma), 20 ng/mL epidermal growth factor (EGF;Sigma), 0.5 mg/mL hydrocortisone (Sigma), 0.1 mg/mL choleratoxin (Sigma), and 10 mg/mL insulin (Sigma). Cellular identitywas regularly verified microscopically on the basis of cellmorphology. Transfections were done using LipofectamineRNAiMAX (Invitrogen) for siRNAs and Lipofectamine 2000(Invitrogen) for plasmid DNA according to the manufacturer'sinstructions. siRNAs are listed in Supplementary Table S1.

Chromatin fractionation, Western blot, and tissuemicroarray analysis

Chromatin fractionation was done as described (17) andanalyzed by Western blotting with antibodies and dilutionslisted in Supplementary Table S6. Tissue microarray analyseswere carried out essentially as described previously (7, 18)with the modifications listed in Supplementary Methods usingthe newly generated anti-H2Bub1 mouse monoclonal anti-body 7B4 and the tissue microarrays BR8010 and BR951(Biomax) containing normal mammary epithelial tissue sam-ples, as well as benign, malignant, and metastatic samples.

Cell migration, colony formation, and apoptosis assayCell migration was assayed by seeding 100,000 MCF10A

cells 48 hours after transfection with the respective siRNAsinto 8.0-mm PET track-etched membrane cell culture inserts(BD Bioscience). Cells were grown for another 48 hours beforefixation with methanol for 10 minutes. Migrated cells werevisualized by crystal violet staining [0.1% (w/v) crystal violet,10% (v/v) formaldehyde] for 10 minutes. For determiningproliferative capacity, 20,000MCF7 cells were plated in normalgrowth medium which was replaced 10 hours later by hor-mone-deprived medium. Cells were treated with 17b-estradiolfor 7 days. Subsequently, colonies were fixed with 70% metha-nol for 30 minutes on ice before staining with crystal violetsolution for 2 hours. Apoptosis was analyzed using the GuavaNexin Assay (Guava Technologies, Millipore) according to themanufacturer's instructions.

Fluorescence recovery after photobleachingFor fluorescence recovery after photobleaching (FRAP)

analyses, MCF7 cells were transiently transfected with aplasmid in which the open reading frame of ERa was clonedin frame into the pEGFP-C2 vector (Clontech Laboratories)and grown for 48 hours in estrogen-free medium beforeconducting analyses. Eight-bit time lapse images of cells wereacquired at a frame rate of 3 frames per second with a confocalmicroscope setup and its software (TCS SP2 AOBS; Leica) witha 40� oil immersion objective (Type HCX PL Apo CS, NA 1.25;

Prenzel et al.

Cancer Res; 71(17) September 1, 2011 Cancer Research5740

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 3: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

Leica). The pinhole was set at 81.40 mm and the zoom factorwas adjusted to obtain 62.5� 62.5 mm2 images with voxel sizeof 244.26 � 244.26 nm2. After recording 10 images, the greenfluorescent protein (GFP) signal in a 13.2 � 2.9 mm2 regionwithin a nucleus was bleached with a 488-nm laser beam and120 postbleaching images were taken. To correct the intensityfrom imaging bleaching and background signal in thebleached region of the postbleaching series, the formula

IcorrðtÞ ¼ IoriðtÞ � Ibg� �� IrefðtÞ � Ibg

Irefð0Þ � Ibg

was used, where Icorr (t) represents the corrected intensity at acertain time point; Iori represents the original intensity; Ibg isthe average background signal in the series; and Iref is the GFPsignal from another nucleus in the same image field used formeasuring the GFP bleaching from image acquisition. Thefluorescence recovery of the bleached region was then fittedwith a simple exponential function using the SigmaPlot (Systat

Software) with 3 variables, Itheor ðtÞ ¼ y0 � a� e�tb , where y0

is the theoretical maximum intensity and b is the timeconstant. The Welchs two-sample t test was carried out usingthe free statistical software R (19) for statistical computingand graphics to evaluate the significance of the data.

Gene expression, chromatin immunoprecipitation, andquantitative real-time PCRRNA was isolated from cells with the TRIzol reagent (Invi-

trogen) according to the manufacturer's instructions. Onemicrogram of total RNA was reverse transcribed using randomnonamerprimers (Metabion). Chromatin immunoprecipitation(ChIP) and subsequent real-time PCR analyses were conductedessentially as previously described (7, 20) with modificationsdescribed in Supplementary Methods and the antibodies listedin Supplementary Table S6. ChIP and input samples werequantified using a standard curve made from ChIP inputDNA. ChIP samples were normalized to their correspondinginput samples and expressed as "percent input." cDNA sampleswere quantified using a standard curve made from all cDNAsamples. Prior to statistical analysis, all quantitative real-timePCR (qRT-PCR) samples were normalized to an internal refer-ence gene (28S ribosomal RNA or hnRNPK). The expressionlevels were determined relative to the vehicle-treated controlsample and expressed as "relative mRNA expression."

Microarray analysesTotal RNA for microarray experiments was isolated and

transcriptome-wide gene expression analysis was conductedon samples treated for 24 hours using GeneChip Human Gene1.0 ST Array (Affymetrix) carried out at the TranscriptomeAnalysis Laboratory, University of G€ottingen. FormRNAexpres-sion profiling after 6-hour treatments, RNA samples were pre-pared in the same way and Illumina whole-genome geneexpression analysis using a human HT-12 v4 beadchip wasconducted by the Vancouver Prostate Centre Laboratory forAdvanced Genome Analysis, Vancouver, Canada. Gene expres-sion data were analyzed using log2 transformation and quantilenormalizationof expression levels (21). Todetermine significantdifferences of expression levels between the different groups, a

moderated Student's t test was computed on a gene-by-genebasis using the empirical Bayes statistics in the "LIMMA"package (22). To avoid a high number of false positives andto stay below a false discovery rate of 5%, P values were adjustedfor multiple testing using the Benjamini–Hochberg method(23). All analyses were conducted using the free statisticalsoftware R (version 2.12.2; ref. 19). All gene expression datahave been deposited into the Geo Expression Omnibus repo-sitory under the accession numbers GSE30931 and GSE31118.

Chromosome conformation captureChromosome conformation capture (3C) analysis was done

as described (24) with the modifications described in theSupplementary Methods section. For comparison of interac-tions, DNA derived from bacterial artificial chromosomeclones (BAC information in Supplementary Table S5) wasisolated, digested, and ligated similarly to 3C samples andused for serial dilutions and quantitative analyses essentiallyas described (25). qRT-PCR analysis is described in Supple-mentary Methods. 3C values were normalized with valuesfrom an amplicon located between restriction sites to controlfor equal amounts of input DNA. The normalized levels weregraphed relative to the nontreated control sample (set to 1)and represented as "normalized relative interaction."

Results

Bortezomib blocks proteasome activity in MCF7 breastcancer cells

To investigate the effects of a clinically utilized proteasomeinhibitor on ERa activity, we validated the ability of borte-zomib to block proteasome activity and determined theoptimal concentration of bortezomib for blocking proteasomeactivity in ERa-positive MCF7 breast cancer cells. As shown inSupplementary Figure S1A, bortezomib effectively increasedpolyubiquitination of cellular proteins already at a concentra-tion of 10 nmol/L and exhibited a similar efficiency to that ofthe laboratory proteasome inhibitors MG132 and epoxomicin(Supplementary Fig. S1B). Knockdown of the proteasomesubunits PSMB3 or PSMB5 induced a similar accumulationof polyubiquitinated proteins compared with bortezomibtreatment (Supplementary Fig. S1C). Surprisingly, in contrastto previous reports (16, 26), whereas bortezomib effectivelydecreased estrogen-induced proteasomal degradation of ERa(Supplementary Fig. S1D) in a manner similar to that reportedfor MG132 (9, 27), we observed little or no effect of bortezomibon fulvestrant-induced ERa downregulation (SupplementaryFig. S1E).

Bortezomib does not affect RNAPII carboxy-terminaldomain phosphorylation and only mildly impairs ERanuclear mobility

A previous study suggested that proteasome inhibitionaffected glucocorticoid receptor (GR)-regulated transcriptionby increasing the global phosphorylation of the RNAPIIcarboxy-terminal domain (CTD; ref. 13). Therefore, we testedwhether bortezomib treatment also affects RNAPII phosphor-ylation in MCF7 cells. However, as shown in Figure 1A, no

Bortezomib Reduces ERa Activity by Decreasing H2Bub1

www.aacrjournals.org Cancer Res; 71(17) September 1, 2011 5741

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 4: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

major changes in the phosphorylation of any of the commonlyphosphorylated residues (Ser2, Ser5, or Ser7) of the RNAPIICTD were observed following bortezomib treatment.

Decreased nuclear mobility of ERa following proteasomeinhibition was also proposed as a mechanism by whichMG132 treatment impairs ERa binding to target genesand estrogen-regulated transcription (10, 11). We substan-tiated these data using FRAP where we observed a dramaticdecrease in ERa mobility following MG132 treatment withessentially no recovery observed up to 30 seconds afterphotobleaching (Fig. 1B). However, in comparison toMG132, bortezomib only mildly decreased ERa mobility(Fig. 1C), with effects more similar to those of estrogen.

In stark contrast to MG132, a nearly complete recovery ofGFP-ERa signal was observed 30 seconds after photobleach-ing following bortezomib treatment.

Neither short-term bortezomib treatment norproteasome subunit knockdown induces apoptosis inMCF7 cells

To test for secondary effects caused by the induction ofapoptosis, we investigated the effects of 24- or 48-hour borte-zomib treatment and transfection of siRNAs against theproteasome subunits PSMB3 or PSMB5. As shown in Supple-mentary Figure S2, neither 24-hour bortezomib treatment nortransfection of PSMB3 or PSMB5 siRNAs significantly induced

A B

C

Cont Bort

Total RNAPII

Co

nt

MG

132

Prebleach Postbleach0 s 30 s

RNAPII p-Ser2

RNAPII p-Ser5

RNAPII p-Ser7

HSC70

1.0

0.8

0.6

0.4

0.2

0

Norm

aliz

ed inte

nsity

Tim

e c

onsta

nt

(s)

Cont

Estrogen

Bort

0 5 10 15

Postbleach time (s)

Postbleach timePrebleach

Cont

E2

Bort

0 s 2 s 5 s 30 s

20 25 30 35 40

*****

n.s.

Con

t

Estro

gen

Bort

15

10

5

0

Figure 1. Bortezomib has noeffect on the phosphorylationstatus of RNAPII and in contrast toMG132, only slightly affectsnuclear ERa mobility. A, MCF7cells were treated with eithervehicle (ethanol, Cont) or 50nmol/L bortezomib (Bort) for 20hours and protein extracts wereanalyzed by Western blotting withthe indicated antibodies. HSC70 isshown as loading control. B,pEGFP-hERa–transfected MCF7cells were treated with eitherdimethyl sulfoxide (Cont) or 20mmol/L MG132 for 12 hours beforeFRAP analysis. C, MCF7 cellsexpressing pEGFP-hERa weresubjected to either vehicle(ethanol, Cont), 10 nmol/L17b-estradiol (E2), or 50 nmol/Lbortezomib (Bort) for 6 hoursbefore conducting FRAP. Thefluorescence signal in eachtreatment was normalized with theinitial intensity after bleaching(t ¼ 0) set to 0 and the theoreticalmaximum value set to 1. Averagedvalues in each treatment areshown. The bar graph shows thetime constants of each treatment,error bars representing SEM;n ¼ 36 to 41 cells from 3independent experiments; n.s.,not significant; **, 0.001 < P < 0.01;***, P < 0.001. Images in (B) and (C)show single representative nucleiobtained before bleaching(prebleach) and after the indicatedtime points (postbleach). Thebleached area is indicated as awhite rectangle in the firstpictures; bar ¼ 10 mm.

Prenzel et al.

Cancer Res; 71(17) September 1, 2011 Cancer Research5742

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 5: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

apoptosis in MCF7 cells. Only after prolonged bortezomibtreatment (48 hours) were apoptotic effects observed. There-fore, all subsequent experiments were carried out at timepoints of 24 hours or less.

Bortezomib blocks estrogen-induced gene expressionTo determine the transcriptome-wide effects of bortezomib

treatment on estrogen-regulated gene expression, we carriedout microarray analyses using MCF7 cells treated with estro-gen alone or a combination of bortezomib and estrogen foreither 24 hours (Fig. 2A and B) or 6 hours (SupplementaryFig. S4).We also compared these effects with those of knockingdown the proteasome subunits PSMB3 andPSMB5 (Fig. 2A–C).Ourmicroarray results for both 24 and 6 hours of estrogen or

combined estrogen and bortezomib treatment show thatproteasome inhibition dramatically decreases estrogen-induced gene expression (Fig. 2A and B; SupplementaryFig. S4). These effects were specific because an overwhelmingmajority of estrogen-induced genes (209 of 302)were decreasedin their induction (Fig. 2B). In contrast, although bortezomibalso had a significant effect on the expression of non–estrogen-regulated or estrogen-repressed genes, these conditionsshowed a similar number of genes whose expression wasdecreased or increased by proteasome inhibition. The effectsof bortezomib on estrogen-induced gene transcription arespecifically due to proteasome inhibition because siRNA-mediated knockdown of PSMB3 or PSMB5 resulted in similareffects on estrogen-induced gene expression (Fig. 2A and B)with a significant overlap in the changes observed (Fig. 2C).To verify the results of these microarray studies, we also

conducted qRT-PCR studies on several representative genesfollowing 2, 6, or 24 hours of estrogen, bortezomib, or combinedtreatments. As shown in Figure 2D, the estrogen-inducedexpression of all genes tested (CXCL12, GREB1, TFF1, PGR,PKIB, and WISP2) was dramatically decreased by bortezomibtreatment at each time point, except for TFF1 which showeddecreased induction at both 2 and 6 hours but an increasedinduction at 24 hours (Fig. 2D). The effects observed on theTFF1 gene at 24 hours seem to be due to indirect effects mostlikely caused by changes in the stability or expression of agene-specific cofactor. Consistent with the absence of aneffect of bortezomib on the fulvestrant-induced decrease inERa protein levels, we observed no effect of bortezomib onthe fulvestrant-induced decreases in ERa-dependent genetranscription and ERa binding (Supplementary Fig. S3).Further microarray studies conducted after 6 hours oftreatment (Supplementary Fig. S4) showed similar, albeitweaker, effects of bortezomib treatment on estrogen-induced transcription. Therefore, we conclude that protea-some inhibition most likely decreases ERa-dependent estro-gen-induced gene transcription by a direct mechanism in amanner distinct from that of the antiestrogen fulvestrant.

GR activity is not affected by bortezomib treatmentThe laboratory proteasome inhibitor MG132 has significant

effects on GR-dependent gene expression (13). Thus, we alsotested the specificity of proteasome inhibition toward ERa-dependent gene transcription by treating the glucocorticoid-

responsive A549 lung cancer cell line with bortezomib anddexamethasone. In contrast to the effects observed with ERa,we observed no effects of bortezomib treatment on the rapidinduction of the 4 glucocorticoid-responsive genes investi-gated (FKBP5, GILZ, SGK1, and SLC19A2; Fig. 3A). Further-more, no effect of bortezomib on GR binding to these geneswas observed (Fig. 3B).

Bortezomib treatment does not affect ERa binding orcofactor recruitment to estrogen-induced genes

On the basis of the results of a previous study investigatingthe effects of MG132 on the TFF1 gene (10), we utilized ChIPanalyses to test whether proteasome inhibition by bortezomibaffects ERa binding to endogenous estrogen-induced genesand the subsequent assembly of factors involved in transcrip-tional activation. However, no significant decrease in ERabinding to any of the 10 investigated binding sites wasobserved (Fig. 4A; Supplementary Figs. S5 and 6A). Giventhe importance of the p160 family of nuclear receptor coacti-vators in controlling estrogen-induced gene transcription andcellular proliferation (28), we also investigated whether thebinding of GRIP1 (also called SRC2 and TIF2) was affected bybortezomib treatment. Like ERa, GRIP1 showed an increasedrecruitment to several ERa binding sites following estrogentreatment, and this recruitment was unaffected by bortezomibtreatment (Fig. 4B, Supplementary Fig. S6B).

The induction of long-range chromosomal interactionsdoes not require proteasome activity

Unbiased chromosome- and genome-wide studies showedthat a large fraction of ERa binding sites are greater than 10 kbaway from the genes that they regulate (29). Consistently, ERafrequently colocalizes with the cohesin component STAG1(30) and thereby induces dramatic changes in higher orderchromatin structure by inducing long-range chromosomalinteractions between different ERa binding sites (25). Com-ponents of the mediator complex, which has been shown toplay an essential role in regulating ERa function (31), alsocolocalize and cooperate with cohesin to regulate tissue-specific gene expression (32). Therefore, we also examinedthe recruitment of MED12 (Fig. 4C, Supplementary Fig. S6C)and STAG1 (Fig. 4D, Supplementary Fig. S6D) to ERa-bindingsites which nucleate long-range chromosomal interactions(25) and were previously shown to colocalize with cohesin(30). These studies revealed that neither MED12 nor STAG1recruitment was affected by bortezomib treatment. Consis-tent with the preserved recruitment of both MED12 andcohesin to ERa binding sites, we also observed no effect ofbortezomib treatment on estrogen-induced changes in long-range chromosomal interactions as assessed by 3C analyseson the CXCL12, GREB1, and TFF1 genes (Fig. 4E; Supplemen-tary Figs. S6E and F and S7).

Proteasome inhibition does not affect transcriptionalinitiation steps but decreases efficient transcriptionalelongation

On the basis of the normal recruitment of ERa, transcrip-tional coactivators, and the induction of long-range chromo-

Bortezomib Reduces ERa Activity by Decreasing H2Bub1

www.aacrjournals.org Cancer Res; 71(17) September 1, 2011 5743

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 6: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

Prenzel et al.

Cancer Res; 71(17) September 1, 2011 Cancer Research5744

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 7: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

somal interactions, we next tested which subsequent transcrip-tional stepmay be affected by proteasome inhibition. Given thesignificant role of histone H3 acetylation (H3Ac) during tran-scriptional initiation, we investigated whether estrogen-induced changes in this epigenetic mark near the transcrip-tional start site (TSS) of ERa target genes may be affected bybortezomib treatment. However, no effect of bortezomib onH3Ac was observed on any of the investigated genes (Fig. 4F;Supplementary Fig. S8A). Despite the significant decreaseobserved in their mRNA levels (Fig. S2D), the recruitment ofRNAPII to the TSS was also unaffected on both the CXCL12 andGREB1 genes andonlymildly affected on theTFF1 gene (Fig. 4G;Supplementary Fig. S8B). These results are supported by aprevious study which concluded that many ERa target genesare regulated primarily at the level of transcriptional elongation(33). Therefore, we concluded that a posttranscriptional initia-tion step such as transcriptional elongation likely leads to thedecrease in mRNA expression observed on most ERa targetgenes following bortezomib treatment.Phosphorylation of RNAPII at serine 2 of its CTD by cyclin-

dependent kinase-9 (CDK9) plays a critical role during tran-scriptional elongation by serving as a scaffold to promote thebinding of transcriptional regulatory and chromatin-modify-ing proteins (34). Therefore, we investigated the effects ofbortezomib treatment on CDK9 recruitment to the TSS ofCXCL12, GREB1, and TFF1 following estrogen, bortezomib, orcombined treatments in MCF7 cells. As shown in Figure 4Hand Supplementary Figure S8C, bortezomib had little or noeffect on CDK9 recruitment in the presence of estrogen. Thus,we next tested whether the ability of RNAPII to traverse ERatarget genes was affected by proteasome inhibition. Indeed, asshown in Figure 4I and Supplementary Figure S8D, weobserved a decrease in the elongating (Ser2 phosphorylated)form of RNAPII at the 30 end of the investigated estrogen-regulated genes, whereas the total levels of Ser2 phosphoryla-tion were unchanged (see Fig. 1A).

Proteasome inhibition decreases H2Bmonoubiquitination and affects histone exchangeOn the basis of the normal recruitment of CDK9 but

decreased ability of RNAPII to transcribe the entire lengthof the investigated ERa target genes, we hypothesized thatbortezomib-induced changes in chromatin structure mayprevent transcriptional elongation. One histone modificationwhich is associated with the transcribed regions of activegenes (35) is dependent upon Ser2 phosphorylation of the

RNAPII CTD (20) and has been implicated in transcriptionalelongation is histone H2Bub1 (36). Furthermore, proteasomeinhibition causes a rapid decrease in H2Bub1 within minutesof treatment (35, 37). In contrast to the stabilizing effect ofproteasome inhibition on many ubiquitinated proteins, theeffect on H2Bub1 is related to the transient nature ofH2Bub1 and is a consequence of a rapid depletion of thepool of free ubiquitin in the nucleus (37, 38). Consistent withthe previously published results for other proteasome inhi-bitors (35, 37), we observed a significant decrease in H2Bub1following bortezomib treatment (Fig. 5A). These effects werespecific for H2Bub1 because other histone modificationsassociated with active genes and mark transcriptional initia-tion (H3 lysine 9/14 acetylation; H3Ac) and elongation (H3lysine 36 trimethylation; H3K36me3) were unaffected bybortezomib treatment.

H2Bub1 functions during transcriptional elongation bycooperating with the facilitates chromatin transcription(FACT) histone chaperone complex to increase histoneexchange and open chromatin structure to allow passage ofRNAPII through chromatin during transcription (36). Impor-tantly, H2Bub1 is localized exclusively in a chromatin-asso-ciated form (39) and seems to be a prerequisite for bothtranscription-associated (36) and DNA repair–associated his-tone exchange (39). Therefore, we tested whether the borte-zomib-induced decrease in H2Bub1 levels also leads todecreased recruitment of the FACT complex to chromatinby separating the nucleoplasmic and chromatin-bound frac-tions from MCF7 cells treated with estrogen, bortezomib, orcombined treatments. Indeed, as shown in Figure 5B andconsistent with the global decrease in H2Bub1 levels, therecruitment of the FACT component SSRP1 to chromatinwas significantly impaired by bortezomib treatment.

The positive effect of histone chaperones such as FACTduring transcriptional elongation and other DNA-associatedprocesses is achieved by the removal and reinsertion ofcomponents of the core nucleosome. This effect can beobserved by analyzing the soluble fraction (i.e., "non-chroma-tin bound fraction") of core histones such as H3. A recentreport showed that H2Bub1 is essential for DNA damage–induced changes in the pool of free H3 (39). Consistently, wealso observed that the bortezomib-induced loss of total(Fig. 5A) and chromatin-bound H2Bub1 (Fig. 5B) resultedin decreased nucleoplasmic H3 (Fig. 5B). Thus, bortezomibseems to decrease histone exchange by blocking H2B mono-ubiquitination.

Figure 2. The bulk of estrogen-regulated genes is negatively influenced by proteasome inhibition using bortezomib. A, mRNA expression profiling ofestrogen-regulated genes after 24-hour treatment. The heatmap shows log2 fold changes (FC) in experiments 17b-estradiol versus control (E2), bortezomibþ17b-estradiol versus 17b-estradiol (Bort þ E2), and PSMB siRNA þ 17b-estradiol versus control siRNA þ 17b-estradiol (siRNA PSMB þ E2; columns)for genes which are significantly (q < 0.05; q ¼ P values adjusted to false discovery rate) regulated by estrogen [FC < �log2 (1.5) or FC > log2 (1.5) rows]. Thecolor key ranges from red marking downregulated to blue marking upregulated genes; mean values, n¼ 3. PSMB knockdown samples depict all genes whichwere either up- or downregulated in all 4 following comparisons: PSMB3 versus control 1, PSMB3 versus control 2, PSMB5 versus control 1, and PSMB5versus control 2 siRNAs. B, effect of proteasome inhibition and knockdown on genes which are significantly influenced by estrogen. Numbers ofgenes depicted in the heatmapwhich are affected by estrogen, bortezomib, or the knockdown of 20S b subunits. E2 genes: # ¼ q < 0.05, FC <�log2 (1.5); " ¼ q< 0.05, FC > log2 (1.5); Bortþ E2/PSMBþ E2 genes: # ¼ q < 0.1, negative FC; " ¼ q < 0.1, positive FC. C, Venn diagram showing the overlap of genes that aresignificantly upregulated by estrogen [q < 0.05, FC > log2 (1.5)] and that are downregulated by bortezomib and/or knockdown of PSMB3 and PSMB5(negative FC). D, total mRNA was extracted from MCF7 cells, pretreated for 15 minutes with 50 nmol/L bortezomib (Bort) or vehicle (ethanol, Cont) andincubated with 10 nmol/L 17b-estradiol (E2) for 2, 6, or 24 hours. The expression levels of estrogen target genes CXCL12, GREB1, TFF1, PGR, PKIB, andWISP2were normalized to 28S ribosomal RNA graphed relative to the control sample and expressed as "relativemRNA expression"; mean values� SD, n¼ 2.

Bortezomib Reduces ERa Activity by Decreasing H2Bub1

www.aacrjournals.org Cancer Res; 71(17) September 1, 2011 5745

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 8: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

Depletion of the histone H2B ubiquitin ligase RNF40specifically results in decreased estrogen-dependentgene transcription and enables estrogen-independentcell proliferation

Histone H2B monoubiquitination is carried out by a het-erodimeric ubiquitin ligase complex containing RNF20 andRNF40 (36, 40) in which both proteins are essential for

maintaining H2Bub1 levels (20). Given the association ofH2Bub1 with active gene transcription and its connectionto transcriptional elongation, we tested whether a loss ofH2Bub1 following RNF40 knockdown in MCF7 cells impairsestrogen-regulated gene transcription in a manner similar tobortezomib treatment. Indeed, RNF40 knockdown signifi-cantly decreased the induction of CXCL12, GREB1, and

Figure 3. Proteasome inhibitionusing bortezomib has neither asignificant effect on GR targetgene expression nor on GRrecruitment to these target genes.Following a 15-minutepretreatment with 50 nmol/Lbortezomib (Bort) or vehicle(ethanol, Cont), A549 cells wereincubated with 100 nmol/Ldexamethasone (Dex) for 2 hours.A, total RNA was harvested andthe expression levels of GR targetgenes FKBP5, GILZ, SGK, andSLC were normalized to 28Sribosomal mRNA and graphed asdescribed in Figure 2D; meanvalues � SD, n ¼ 2. B, GR bindingto the glucocorticoid responseelements (GRE) of these targetgenes was analyzed via ChIPanalysis using a specific GRantibody. Nonspecific IgG wasused to distinguish betweenspecific and background binding(depicted as a dotted line). ChIPsamples were normalized to inputsamples and expressed as "%input"; mean values � SD, n ¼ 3.

Prenzel et al.

Cancer Res; 71(17) September 1, 2011 Cancer Research5746

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 9: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

TFF1 expression following estrogen treatment (Fig. 5C; Sup-plementary Fig. S9A), without decreasing the protein levels ofany of the investigated cofactors MED12, GRIP1, or CDK9(Supplementary Fig. S9B). This effect was specific for ERa-regulated transcription, as the rapid induction of several GR

target genes following dexamethasone treatment of A549 cellswas unaffected by RNF40 knockdown (Fig. 5D; SupplementaryFig. S9C).

Our previous work indicated a potential tumor suppressorrole for H2Bub1 (41). Therefore, we tested whether a loss of

Figure 4. Bortezomib treatment has no effect on ERa, GRIP1, MED12, and STAG1 recruitment to target genes, does not influence estrogen-inducedlong-range interaction, shows no effect on the transcriptional initiation complex assembly but decreases RNAPII elongation on estrogen target genes. A–D andF–I, after a 15-minute pretreatment with 50 nmol/L bortezomib (Bort) or vehicle (ethanol, Cont), MCF7 cells were incubatedwith 10 nmol/L 17b-estradiol (E2) for2 hours. ERa (A), GRIP1 (B), MED12 (C), STAG1 (D), H3Ac (F), RNAPII (G), CDK9 (H), and RNAPII p-Ser2 (I) binding to the indicated CXCL12 gene siteswas analyzed via ChIP analysis using specific antibodies. ChIP analysis was conducted as in Figure 3B; mean values � SD, n ¼ 3. E, graphical schemeof the tested 3C sites on the CXCL12 locus. The interacting sites were chosen on the basis of their published ChIA-PET interactions by Fullwood andcolleagues (25) and are labeled according to published ChIP-on-chip data by Carroll and colleagues (29). 3C assay was done as described in the Methodssection. Purified 3C DNA samples were quantified by qPCR using a standard curve from BAC DNA containing the CXCL12 locus which was digested andligated as for 3C samples. 3C values were normalized to values from an internal control site that lies between restriction enzyme sites, graphed relative to thecontrol sample (set to 1), and represented as "normalized relative interaction"; mean values � SD, n ¼ 3.

Bortezomib Reduces ERa Activity by Decreasing H2Bub1

www.aacrjournals.org Cancer Res; 71(17) September 1, 2011 5747

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 10: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

Figure 5. Proteasome inhibition using bortezomib decreases histone H2B monoubiquitination. Knockdown of the H2B ubiquitin ligase RNF40decreases estrogen-induced target gene expression but enhances estrogen-independent proliferative capacity of breast cancer cells. A, after growingMCF7 cells in hormone-deprived medium for 3 days, cells were pretreated with either vehicle (ethanol) or 50 nmol/L bortezomib (Bort) for 15 minutesbefore incubating with 10 nmol/L 17b-estradiol (E2) for 2 hours. Whole protein extracts were analyzed via Western blotting with the indicatedantibodies. b-Actin is shown as loading control. B, MCF7 cells were kept in hormone-depleted medium for 2 days and then treated (as in A). Nuclearprotein extracts were separated into soluble and insoluble (chromatin) fractions and analyzed by Western blotting for SSRP1, histone H3, H2Bub1, and H2Bprotein levels. MCF7 (C) and A549 (D) cells transfected with RNF40 siRNAs for 24 hours were grown in hormone-free medium for an additional 48 hoursbefore stimulation with either 10 nmol/L 17b-estradiol (E2; C) or 100 nmol/L dexamethasone (Dex; D) for 2 hours. Hormone-induced gene expressionwas normalized to hnRNPK (C) or 28S ribosomal RNA (D) levels and graphically represented as in Figure 2D; mean values � SD, n ¼ 3. E and F, clonogenicassay after RNF40 knockdown. MCF7 cells were transfected with control or RNF40 siRNA. The next day, cells were replated into 6-well plates.After attachment, growth medium was changed to hormone-deprived 5% CSS growth medium and cells stimulated with 10 nmol/L 17b-estradiol (E2) for7 days (E). Efficient knockdown of RNF40 was verified and ERa, p-AKT, AKT, p-ERK, ERK, and H2Bub1 protein levels were analyzed byWestern blotting. H2Bwas used as loading control (F).

Prenzel et al.

Cancer Res; 71(17) September 1, 2011 Cancer Research5748

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 11: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

H2Bub1 mediated by knockdown of RNF40 might affectestrogen-dependent cellular proliferation. Interestingly,RNF40 knockdown led to estrogen-independent proliferationof MCF7 cells (Fig. 5E and F). This effect was not furtherpotentiated by estrogen treatment. In contrast, control trans-fected cells depended upon estrogen treatment of prolifera-tion (Fig. 5E, top).ERa-independent proliferation is frequently associated

with increased activation of the phosphoinositide-3-kinase(PI3K)/AKT and extracellular signal–regulated kinase(ERK)/mitogen-activated protein kinase (MAPK) signalingpathways (42). To determine whether a loss of RNF40expression also leads to the activation of estrogen-independent cell proliferation signaling, we examined theactivation of both AKT and ERK. Interestingly, RNF40 knock-down was sufficient to significantly activate both signalingpathways (Fig. 5F).

H2Bub1 decreases during tumor progressionAlthough our previous study indicated a potential tumor

suppressor role for RNF20 (41), no study to date has inves-tigated the presence of H2Bub1 in normal and canceroustissues. Therefore, we developed a newH2Bub1-specificmono-clonal antibody using a previously published branched-peptideantigen strategy (35) and carried out immunohistochemicalanalyses of tissue microarrays containing normal mammaryepithelial tissue as well as benign, malignant, and metastaticbreast cancer samples. These studies yielded a total of 109interpretable results in which the nuclei of both normalmammary ductal epithelial cells (Fig. 6A) and all 18 benigntumor samples stained positive for H2Bub1 (Fig. 6B and C).In contrast, only 21 of 64 malignant breast cancer samples(32%) stained positive for H2Bub1 and only 3 of the 19metastatic tumor samples stained positive for H2Bub1(Fig. 6B and C; Table 1). As a control, we also stained normaland tumor samples for (total) H2B (Supplementary Fig. S10).As expected, all samples stained positive for H2B, confirmingthat the decreased H2Bub1 staining during breast cancerprogression is due to decreased monoubiquitination of H2Band not a loss of nuclear histones. Thus, decreased H2Bmonoubiquitination correlates with breast cancer progres-sion and metastasis in a small cohort of tumor samples.Furthermore, on the basis of these results and the impor-tance of cell migration for metastasis, we also investigatedthe effects of RNF40 knockdown on the migratory potentialof a normal mammary epithelial cell line. Consistent with acentral role for H2Bub1 in suppressing metastasis, increasedAKT and ERK activity following RNF40 knockdown, and withour previous study with RNF20 (41), siRNA-mediated knock-down of RNF40 resulted in increased migration of MCF10Acells (Fig. 6D).

Discussion

Inhibition of proteasome activity has been shown to beeffective in treating some types of cancers including multiplemyeloma and mantle cell lymphoma. However, its effective-ness in the treatment of breast cancer is not clear. Moreover,

its utility for different stages and subtypes of breast cancerneeds to be determined. On the basis of the proposed neces-sity of proteasome activity for ERa-induced gene transcription(9, 10), we sought to determine the effects of the clinicalproteasome inhibitor bortezomib on estrogen-regulated genetranscription and investigated its mechanism of action. Weprovide the first evidence that bortezomib-mediated protea-some inhibition functions to limit transcriptional elongationof estrogen-induced genes by causing a global decrease inH2Bub1 levels. This in turn prevents histone chaperonerecruitment to chromatin and increased nucleosomedynamics on ERa target genes.

This "nonproteolytic" effect of proteasome inhibition chal-lenges the general assumption that proteasome inhibitionfunctions primarily by preventing the degradation of specificproteins required for essential cellular processes (43, 44).Although these mechanisms likely influence the ultimateinhibition of cell-cycle progression and the induction ofapoptosis after prolonged bortezomib treatment in MCF7cells, they do not seem to be responsible for the rapid effectson estrogen-induced gene transcription that we haveobserved.

A recent study reported a combinatorial effect of fulvestrantand bortezomib on the induction of apoptosis and laid theground work for ongoing clinical trials combining these 2treatments (16). In this study, we were unable to see anyeffects of bortezomib treatment on the rapid fulvestrant-induced ERa downregulation and the repression of ERa targetgene transcription or ERa binding. Thus, as suggested by thereported induction of an unfolded protein response, the long-term effects of combined bortezomib and fulvestrant treat-ment are probably not due to changes in ERa-dependent genetranscription but rather a secondary effect induced by theaccumulation of a highly polyubiquitinated protein. However,additional experiments are clearly needed to address whetherand how these 2 therapies work synergistically, their mechan-ism of action, and the contribution of altering ERa transcrip-tional activity in eliciting their effects.

According to our data, we propose a new mechanismwhereby bortezomib treatment decreases the ability of ERato activate estrogen-responsive genes by leading to a rapid andglobal loss of H2Bub1. This effect is due to a rapid depletion offree ubiquitin in the nucleus (37, 38). On the basis of the verydynamic nature of H2B monoubiquitination (20), the deple-tion of nuclear ubiquitin prevents new ubiquitination of H2Band causes a rapid loss of H2Bub1 levels. This is in starkcontrast to most previously proposed models which invokedthe degradation of specific transcriptional complexes as beingessential for ERa activity. Although a stabilization of somegene-specific coactivator proteins following bortezomib treat-ment likely influences the regulation of some estrogen-respon-sive genes at later time points (e.g., TFF1), our data suggestthat H2Bub1-dependent recruitment of the FACT histonechaperone complex and the subsequent changes in nucleo-some dynamics which allow passage of transcribing RNAPIIthrough ERa target genes is a likely mechanism wherebybortezomib very rapidly decreases the majority of estrogen-induced gene transcription.

Bortezomib Reduces ERa Activity by Decreasing H2Bub1

www.aacrjournals.org Cancer Res; 71(17) September 1, 2011 5749

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 12: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

Figure 6. H2Bub1 expression in human normal breast tissue, tumors, and metastasis. A, representative immunohistochemical image displayingH2Bub1 protein expression in normal breast tissue; bar ¼ 100 mm. B, bar graph representing the immunohistochemical analysis of the H2Bub1 staining innormal adjacent tissue, the indicated breast tumors, or metastasis. Shown are relative numbers in "% samples" as well as the absolute numbers of samples onthe bottom of each bar. C, representative sample pictures of a tissue microarray analysis showing immunohistochemical H2Bub1 staining in benign,malignant, and metastatic breast tumor samples. Samples were categorized into 2 groups as either negative or positive for H2Bub1 (shown in left top cornersof the images); bar¼ 100 mm. D, cell migration assay after RNF40 knockdown. Control or RNF40 depleted MCF10A cells were seeded onto cell culture insertsand allowed to migrate for 48 hours. Representative images from an experiment are shown, bar ¼ 500 mm.

Prenzel et al.

Cancer Res; 71(17) September 1, 2011 Cancer Research5750

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 13: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

Consistent with the data presented here on estrogen-regu-lated genes, we and others have shown that H2Bub1 isprobably most important for signal-specific induction of geneexpression (36, 41). These effects are likely to be due to theneed for major changes in chromatin structure caused bydisruption of the 30-nm chromatin fiber by monoubiquitina-tion of H2B (45). Genes which are already active or repressedmay not require the presence or absence of H2Bub1 to remainin these states. In contrast, the activation of silent or lowlyexpressed genes which lie within a specific chromatin contextis more likely to require the addition or loss of H2Bub1because the activation of these genes will require dynamicalterations in the structure of the surrounding chromatin.Interestingly, these effects seem to be signal specific becausethe activation of gene transcription by GR did not requireRNF40 expression.A particularly striking and potentially therapy-relevant

finding of this study is the correlation of a loss of H2Bub1with tumor progression and the effects of RNF40 knock-down on both cellular proliferation and migration. Despitea significant decrease in the induction of estrogen-regulated gene transcription, we observed no significanteffect of RNF40 knockdown on cellular proliferation inthe presence of estrogen. Instead, RNF40 knockdownallowed for estrogen-independent proliferation of MCF7cells. These paradoxical findings were initially very surpris-ing. However, our findings that the AKT and ERK cellsurvival and proliferative signaling pathways are activatedsuggest that the effects of RNF40 knockdown may extendbeyond its effects on chromatin structure. Consistent withthis hypothesis, the obligate heterodimeric partner ofRNF40, RNF20, has been shown to interact with both iso-forms of EBP1 (46) which has been implicated in directlycontrolling the activity of nuclear AKT (47) and the EGFreceptor (48). Although, it is likely that the increasedactivation of the ERK and AKT pathways may be relatedto this interaction, it is presently unknown how or if RNF40affects the activity of EBP1. Thus, a loss of RNF40 or RNF20activity during tumor progression may be an importantstep in the development of hormone-independent breastcancer by simultaneously decreasing ERa-dependent celldifferentiation and activating cell survival and proliferativesignaling.

We hypothesize that the loss of H2Bub1 during tumorprogression and metastasis may lead to decreased cellulardifferentiation and reversion to a stem cell–like phenotype. Insupport of this, the mRNA levels of the H2B deubiquitinatingenzyme USP22 correlate with an undifferentiated stem-likephenotype in multiple types of cancer (49), and its proteinexpression was shown to correlate with breast cancer pro-gression and patient outcome (50). Thus, multiple mechan-isms which diminish H2B monoubiquitination such asdecreased RNF20 or RNF40 expression or increased USP22expression may similarly lead to a loss of H2Bub1 and rever-sion to a stem cell–like phenotype.

In this study, we have uncovered a novel mechanismwhereby proteasome inhibition blocks ERa-activated geneexpression by interfering with histone H2B monoubiquitina-tion and histone exchange. Moreover, we provide the firstevidence that H2Bub1 decreases during tumorigenesis. Addi-tional studies into the various mechanisms of proteasomeinhibitor action need to be conducted to determine whichtumors may respond best to bortezomib treatment. Forexample, more differentiated ERa/H2Bub1-positive tumorsmight be predicted to respond to H2Bub1-reducing therapiessuch as bortezomib which simultaneously decrease ERa-dependent gene transcription and induce apoptosis. In con-trast, more advanced, less differentiated, ERa/H2Bub1-nega-tive tumors may benefit from therapies which increaseH2Bub1 levels and lead to a more differentiated phenotype.To address these aspects, additional immunohistochemicalstudies investigating the presence of H2Bub1 in a larger cohortof patients with more extensive immunohistologic character-ization, patient follow-up information and hormone respon-siveness as well as in vivo animal model studies should beconducted. We hypothesize that H2Bub1 may represent animportant new diagnostic marker and potential therapeutictarget in breast cancer.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

The authors to thank G. Salinas-Riester, S. Luthin, S. Le Bihan, and A. Haegertfor carrying out microarray experiments; P. R€ake-K€ugler for outstanding

Table 1. Statistical analysis of tissue microarray analyses: Fisher's exact test

P < 0.0001

P < 0.0001

Normal adjacent tissue Benign tumor Malignant tumor Metastasis

Positive 8 18 21 3Negative 0 0 43 16

P ¼ 0.0004

P < 0.0001

Bortezomib Reduces ERa Activity by Decreasing H2Bub1

www.aacrjournals.org Cancer Res; 71(17) September 1, 2011 5751

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 14: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

secretarial support; T. Pukrop and H. Reichardt for advice and thoughtfulsuggestions throughout these studies; and all the members of the Johnsengroup for helpful discussions and proofreading.

Grant Support

This work was funded by an ERC Starting Grant to M. Simons; grants fromthe German Research Foundation (DFG), Transregional CollaborativeResearch Group (SFB) TR5 to D. Eick; the German Research Foundation

(DFG) Research Group 942 to T. Beissbarth; and the Deutsche Krebshilfe(109088), the State of Lower Saxony, Hannover, Germany (VWZN2562), andthe Forschungsf€orderungs program at University of G€ottingen MedicalFaculty to S.A. Johnsen.

The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received June 6, 2011; revised July 12, 2011; accepted July 15, 2011;published OnlineFirst August 23, 2011.

References1. Deroo BJ, Korach KS. Estrogen receptors and human disease. J Clin

Invest 2006;116:561–70.2. Ali S, Coombes RC. Endocrine-responsive breast cancer and strate-

gies for combating resistance. Nat Rev Cancer 2002;2:101–12.3. Musgrove EA, Sutherland RL. Biological determinants of endocrine

resistance in breast cancer. Nat Rev Cancer 2009;9:631–43.4. Johnsen SA, Kangaspeska S, Reid G, Gannon F. Interfering with the

dynamics of estrogen receptor-regulated transcription. Ernst Scher-ing Found Symp Proc 2006;1–12.

5. Hoeller D, Dikic I. Targeting the ubiquitin system in cancer therapy.Nature 2009;458:438–44.

6. Ciechanover A. Proteolysis: from the lysosome to ubiquitin and theproteasome. Nat Rev Mol Cell Biol 2005;6:79–87.

7. Johnsen SA, G€ung€or C, Prenzel T, Riethdorf S, Riethdorf L, Taniguchi-Ishigaki N, et al. Regulation of estrogen-dependent transcription bythe LIM cofactors CLIM and RLIM in breast cancer. Cancer Res2009;69:128–36.

8. Fan M, Nakshatri H, Nephew KP. Inhibiting proteasomal proteolysissustains estrogen receptor-alpha activation. Mol Endocrinol 2004;18:2603–15.

9. Lonard DM, Nawaz Z, Smith CL, O’Malley BW. The 26S proteasome isrequired for estrogen receptor-alpha and coactivator turnover andfor efficient estrogen receptor-alpha transactivation. Mol Cell 2000;5:939–48.

10. Reid G, Hubner MR, Metivier R, Brand H, Denger S, Manu D, et al.Cyclic, proteasome-mediated turnover of unliganded and ligandedERalpha on responsive promoters is an integral feature of estrogensignaling. Mol Cell 2003;11:695–707.

11. Stenoien DL, Patel K, Mancini MG, Dutertre M, Smith CL, O’MalleyBW, et al. FRAP reveals that mobility of oestrogen receptor-alpha isligand- and proteasome-dependent. Nat Cell Biol 2001;3:15–23.

12. Zhang H, Sun L, Liang J, Yu W, Zhang Y, Wang Y, et al. The catalyticsubunit of the proteasome is engaged in the entire processof estrogen receptor-regulated transcription. EMBO J 2006;25:4223–33.

13. Kinyamu HK, Archer TK. Proteasome activity modulates chromatinmodifications and RNA polymerase II phosphorylation to enhanceglucocorticoid receptor-mediated transcription. Mol Cell Biol2007;27:4891–904.

14. Yang CH, Gonzalez-Angulo AM, Reuben JM, Booser DJ, Pusztai L,Krishnamurthy S, et al. Bortezomib (VELCADE) in metastatic breastcancer: pharmacodynamics, biological effects, and prediction ofclinical benefits. Ann Oncol 2006;17:813–7.

15. Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, IrwinD, et al. A phase 2 study of bortezomib in relapsed, refractorymyeloma. N Engl J Med 2003;348:2609–17.

16. Ishii Y, Papa L, Bahadur U, Yue Z, Aguirre-Ghiso J, Shioda T, et al.Bortezomib enhances the efficacy of fulvestrant by amplifying theaggregation of the estrogen receptor, which leads to a proapoptoticunfolded protein response. Clin Cancer Res 2011;17:2292–300.

17. Mendez J, Stillman B. Chromatin association of human origin recog-nition complex, cdc6, and minichromosome maintenance proteinsduring the cell cycle: assembly of prereplication complexes in latemitosis. Mol Cell Biol 2000;20:8602–12.

18. Begus-Nahrmann Y, Lechel A, Obenauf AC, Nalapareddy K, Peit E,Hoffmann E, et al. p53 deletion impairs clearance of chromosomal-instable stem cells in aging telomere-dysfunctional mice. Nat Genet2009;41:1138–43.

19. R Development Core Team. R: a language and environment forstatistical computing. Vienna, Austria: R Foundation for StatisticalComputing; 2011.

20. Pirngruber J, Shchebet A, Schreiber L, Shema E, Minsky N, ChapmanRD, et al. CDK9 directs H2B monoubiquitination and controls replica-tion-dependent histone mRNA 30 end processing. EMBO Rep2009;10:894–900.

21. Bolstad BM, Irizarry RA, Astrand M, Speed TP. A comparison ofnormalization methods for high density oligonucleotide array databased on variance and bias. Bioinformatics 2003;19:185–93.

22. Smyth GK. Linear models and empirical Bayes methods for assessingdifferential expression in microarray experiments. Stat Appl GenetMolBiol 2004;3:Article3.

23. Benjamini Y, Hochberg Y. Controlling the false discovery rate - apractical and powerful approach to multiple testing. J R Statist Soc B1995;57:289–300.

24. Miele A, Dekker J. Mapping cis- and trans- chromatin interactionnetworks using chromosome conformation capture (3C). MethodsMol Biol 2009;464:105–21.

25. Fullwood MJ, Liu MH, Pan YF, Liu J, Xu H, Mohamed YB, et al. Anoestrogen-receptor-alpha-bound human chromatin interactome. Nat-ure 2009;462:58–64.

26. Powers GL, Ellison-Zelski SJ, Casa AJ, Lee AV, Alarid ET. Proteasomeinhibition represses ERalpha gene expression in ERþ cells: a new linkbetween proteasome activity and estrogen signaling in breast cancer.Oncogene 2010;29:1509–18.

27. Nawaz Z, Lonard DM, Dennis AP, Smith CL, O’Malley BW. Protea-some-dependent degradation of the human estrogen receptor. ProcNatl Acad Sci U S A 1999;96:1858–62.

28. Cavarretta IT, Mukopadhyay R, Lonard DM, Cowsert LM, Bennett CF,O’Malley BW, et al. Reduction of coactivator expression by antisenseoligodeoxynucleotides inhibits ERalpha transcriptional activity andMCF-7 proliferation. Mol Endocrinol 2002;16:253–70.

29. Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, Eeckhoute J, et al.Genome-wide analysis of estrogen receptor binding sites. Nat Genet2006;38:1289–97.

30. Schmidt D, Schwalie PC, Ross-Innes CS, Hurtado A, Brown GD,Carroll JS, et al. A CTCF-independent role for cohesin in tissue-specific transcription. Genome Res 2010;20:578–88.

31. Kang YK, Guermah M, Yuan CX, Roeder RG. The TRAP/Mediatorcoactivator complex interacts directly with estrogen receptors alphaandbeta through theTRAP220subunit anddirectly enhancesestrogenreceptor function in vitro. Proc Natl Acad Sci U S A 2002;99:2642–7.

32. Kagey MH, Newman JJ, Bilodeau S, Zhan Y, Orlando DA, van BerkumNL, et al. Mediator and cohesin connect gene expression and chro-matin architecture. Nature 2010;467:430–5.

33. Kininis M, Isaacs GD, Core LJ, Hah N, Kraus WL. Post-recruitmentregulation of RNA polymerase II directs rapid signaling responsesat the promoters of estrogen target genes. Mol Cell Biol 2009;29:1123–33.

34. Egloff S, Murphy S. Cracking the RNA polymerase II CTD code.Trends Genet 2008;24:280–8.

35. Minsky N, Shema E, Field Y, Schuster M, Segal E, Oren M. Mono-ubiquitinated H2B is associated with the transcribed region of highlyexpressed genes in human cells. Nat Cell Biol 2008;10:483–8.

36. Pavri R, Zhu B, Li G, Trojer P, Mandal S, Shilatifard A, et al. HistoneH2B monoubiquitination functions cooperatively with FACT to reg-ulate elongation by RNA polymerase II. Cell 2006;125:703–17.

Prenzel et al.

Cancer Res; 71(17) September 1, 2011 Cancer Research5752

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 15: Estrogen-Dependent Gene Transcription in Human Breast ... · assessing the molecular mechanisms involved and determin-ing which types of tumors (i.e., nonmetastatic vs. metastatic;

37. Mimnaugh EG, Chen HY, Davie JR, Celis JE, Neckers L. Rapiddeubiquitination of nucleosomal histones in human tumor cellscaused by proteasome inhibitors and stress response inducers:effects on replication, transcription, translation, and the cellular stressresponse. Biochemistry 1997;36:14418–29.

38. Dantuma NP, Groothuis TA, Salomons FA, Neefjes J. A dynamicubiquitin equilibrium couples proteasomal activity to chromatin remo-deling. J Cell Biol 2006;173:19–26.

39. Nakamura K, Kato A, Kobayashi J, Yanagihara H, Sakamoto S,Oliveira DV, et al. Regulation of homologous recombination byRNF20-dependent H2B ubiquitination. Mol Cell 2011;41:515–28.

40. Zhu B, Zheng Y, Pham AD, Mandal SS, Erdjument-Bromage H,Tempst P, et al. Monoubiquitination of human histone H2B: thefactors involved and their roles in HOX gene regulation. Mol Cell2005;20:601–11.

41. Shema E, Tirosh I, Aylon Y, Huang J, Ye C, Moskovits N, et al. Thehistone H2B-specific ubiquitin ligase RNF20/hBRE1 acts as a putativetumor suppressor through selective regulation of gene expression.Genes Dev 2008;22:2664–2.

42. Frogne T, Benjaminsen RV, Sonne-Hansen K, Sorensen BS, Nexo E,Laenkholm AV, et al. Activation of ErbB3, EGFR and Erk is essential forgrowth of human breast cancer cell lines with acquired resistance tofulvestrant. Breast Cancer Res Treat 2009;114:263–75.

43. Hideshima T, Chauhan D, Richardson P, Mitsiades C, Mitsiades N,Hayashi T, et al. NF-kappa B as a therapeutic target in multiplemyeloma. J Biol Chem 2002;277:16639–47.

44. Ling YH, Liebes L, Jiang JD, Holland JF, Elliott PJ, Adams J, et al.Mechanisms of proteasome inhibitor PS-341-induced G(2)-M-phasearrest and apoptosis in human non-small cell lung cancer cell lines.Clin Cancer Res 2003;9:1145–54.

45. Fierz B, Chatterjee C, McGinty RK, Bar-Dagan M, Raleigh DP,Muir TW. Histone H2B ubiquitylation disrupts local andhigher-order chromatin compaction. Nat Chem Biol 2011;7:113–9.

46. Liu Z, Oh SM, Okada M, Liu X, Cheng D, Peng J, et al. Human BRE1 isan E3 ubiquitin ligase for Ebp1 tumor suppressor. Mol Biol Cell2009;20:757–68.

47. Ahn JY, Liu X, Liu Z, Pereira L, Cheng D, Peng J, et al. Nuclear Aktassociates with PKC-phosphorylated Ebp1, preventing DNA frag-mentation by inhibition of caspase-activated DNase. EMBO J2006;25:2083–95.

48. Yoo JY, Wang XW, Rishi AK, Lessor T, Xia XM, Gustafson TA, et al.Interaction of the PA2G4 (EBP1) protein with ErbB-3 and regulation ofthis binding by heregulin. Br J Cancer 2000;82:683–90.

49. Glinsky GV, Berezovska O, Glinskii AB. Microarray analysis iden-tifies a death-from-cancer signature predicting therapy failurein patients with multiple types of cancer. J Clin Invest 2005;115:1503–21.

50. Zhang Y, Yao L, Zhang X, Ji H, Wang L, Sun S, et al. Elevatedexpression of USP22 in correlation with poor prognosis in patientswith invasive breast cancer. J Cancer Res Clin Oncol 2011;137:1245–53.

Bortezomib Reduces ERa Activity by Decreasing H2Bub1

www.aacrjournals.org Cancer Res; 71(17) September 1, 2011 5753

Cancer Research. by guest on August 22, 2020. Copyright 2011 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from