tumor and stem cell biology cancer research pituitary ... · mutant Δ29-93 were seeded onto bd...

12
Tumor and Stem Cell Biology Pituitary Tumor Transforming Gene Binding Factor: A New Gene in Breast Cancer Rachel J. Watkins 1 , Martin L. Read 1 , Vicki E. Smith 1 , Neil Sharma 1 , Gary M. Reynolds 2 , Laura Buckley 3 , Craig Doig 4 , Moray J. Campbell 4 , Greg Lewy 1 , Margaret C. Eggo 1 , Laurence S. Loubiere 1 , Jayne A. Franklyn 1 , Kristien Boelaert 1 , and Christopher J. McCabe 1 Abstract Pituitary tumor transforming gene (PTTG) binding factor (PBF; PTTG1IP) is a relatively uncharacterized oncopro- tein whose function remains obscure. Because of the presence of putative estrogen response elements (ERE) in its promoter, we assessed PBF regulation by estrogen. PBF mRNA and protein expression were induced by both dieth- ylstilbestrol and 17β-estradiol in estrogen receptor α (ERα)positive MCF-7 cells. Detailed analysis of the PBF pro- moter showed that the region 399 to 291 relative to the translational start site contains variable repeats of an 18-bp sequence housing a putative ERE half-site (gcccctcGGTCAcgcctc). Sequencing the PBF promoter from 122 normal subjects revealed that subjects may be homozygous or heterozygous for between 1 and 6 repeats of the ERE. Chro- matin immunoprecipitation and oligonucleotide pull-down assays revealed ERα binding to the PBF promoter. PBF expression was low or absent in normal breast tissue but was highly expressed in breast cancers. Subjects with greater numbers of ERE repeats showed higher PBF mRNA expression, and PBF protein expression positively cor- related with ERα status. Cell invasion assays revealed that PBF induces invasion through Matrigel, an action that could be abrogated both by siRNA treatment and specific mutation. Furthermore, PBF is a secreted protein, and loss of secretion prevents PBF inducing cell invasion. Given that PBF is a potent transforming gene, we propose that estrogen treatment in postmenopausal women may upregulate PBF expression, leading to PBF secretion and increased cell invasion. Furthermore, the number of ERE half-sites in the PBF promoter may significantly alter the response to estrogen treatment in individual subjects. Cancer Res; 70(9); 373949. ©2010 AACR. Introduction Breast carcinogenesis requires multiple genetic changes, in- cluding the altered expression and function of tumor suppres- sor genes and oncogenes. Most human breast cancers evolve from normal epithelial cells in terminal duct lobular units through a series of increasingly abnormal stages over long pe- riods of time. Key stages in this progression are hyperplasia, atypical hyperplasia, in situ carcinoma, invasive carcinoma, and, finally, metastatic disease (1). Invasion into surrounding stroma defines the transition from in situ to invasive carcino- ma. However, most defects responsible for the development and progression of malignant disease remain unknown (1). Described in only nine publications (210), four of which are from our own group (2, 3, 6, 8), Pituitary tumor trans- forming gene (PTTG) binding factor (PBF) was identified through its ability to interact with PTTG1, the human securin (4). First isolated in 1997 (11), PTTG1 is an estrogen-regulated gene (12, 13) previously implicated in breast cancer, with highest expression in invasive and metastatic breast cancers (14). Its binding partner PBF has not, however, been studied in the context of breast cancer before. Initially identified in 1998 (10), PBF comprises six exons spanning 24 kb within chromosomal region 21q22.3. The 180amino acid peptide sequence of PBF shares no signifi- cant homology with other human proteins but is highly con- served across a wide diversity of animal species (73% homology to mouse, 67% frog, 60% chicken, 52% zebra fish), suggesting both unique functionality and significant evolu- tionary importance. PBF is ubiquitously expressed (15), but a decade after its cloning, very little has been reported concerning its func- tion. We previously characterized PBF expression in thyroid cancers and showed it to be a transforming gene in vitro and to be tumorigenic in vivo (8). Furthermore, high PBF expres- sion was independently associated with poor prognosis in human thyroid cancer. Most recently, we showed that PBF repressed iodide uptake in thyroid cells, both through tran- scriptional regulation (3) and through altered subcellular trafficking (6). PBF is thus a relatively uncharacterized transforming gene that plays a part in multiple cellular processes, particularly in the setting of endocrine neoplasia. We now present extensive Authors' Affiliations: 1 School of Clinical and Experimental Medicine, 2 Centre for Liver Research, Institute of Biomedical Research, and 3 Cancer Research UK Clinical Trials Unit, School of Cancer Sciences, University of Birmingham, United Kingdom and 4 Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York Corresponding Author: Christopher J. McCabe, Reader in Endocrine Cancer, School of Clinical and Experimental Medicine, Institute for Bio- medical Research, University of Birmingham, Birmingham, B15 2TH, United Kingdom. Phone: 44-121-415-8713; Fax: 44-121-415-8712; E-mail: [email protected]. doi: 10.1158/0008-5472.CAN-09-3531 ©2010 American Association for Cancer Research. Cancer Research www.aacrjournals.org 3739 Research. on December 24, 2020. © 2010 American Association for Cancer cancerres.aacrjournals.org Downloaded from Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

Upload: others

Post on 02-Sep-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

Tumor and Stem Cell Biology

Cancer

Research

Pituitary Tumor Transforming Gene Binding Factor:A New Gene in Breast Cancer

Rachel J. Watkins1, Martin L. Read1, Vicki E. Smith1, Neil Sharma1, Gary M. Reynolds2, Laura Buckley3,Craig Doig4, Moray J. Campbell4, Greg Lewy1, Margaret C. Eggo1, Laurence S. Loubiere1,Jayne A. Franklyn1, Kristien Boelaert1, and Christopher J. McCabe1

Abstract

Authors' A2Centre fo3Cancer ReUniversityPharmacoBuffalo, Ne

CorresponCancer, Scmedical ReUnited KinE-mail: mc

doi: 10.115

©2010 Am

www.aacr

Dow

Pituitary tumor transforming gene (PTTG) binding factor (PBF; PTTG1IP) is a relatively uncharacterized oncopro-tein whose function remains obscure. Because of the presence of putative estrogen response elements (ERE) in itspromoter, we assessed PBF regulation by estrogen. PBF mRNA and protein expression were induced by both dieth-ylstilbestrol and 17β-estradiol in estrogen receptor α (ERα)–positive MCF-7 cells. Detailed analysis of the PBF pro-moter showed that the region −399 to −291 relative to the translational start site contains variable repeats of an 18-bpsequence housing a putative ERE half-site (gcccctcGGTCAcgcctc). Sequencing the PBF promoter from 122 normalsubjects revealed that subjects may be homozygous or heterozygous for between 1 and 6 repeats of the ERE. Chro-matin immunoprecipitation and oligonucleotide pull-down assays revealed ERα binding to the PBF promoter. PBFexpression was low or absent in normal breast tissue but was highly expressed in breast cancers. Subjects withgreater numbers of ERE repeats showed higher PBF mRNA expression, and PBF protein expression positively cor-related with ERα status. Cell invasion assays revealed that PBF induces invasion through Matrigel, an action thatcould be abrogated both by siRNA treatment and specific mutation. Furthermore, PBF is a secreted protein, and lossof secretion prevents PBF inducing cell invasion. Given that PBF is a potent transforming gene, we propose thatestrogen treatment in postmenopausal women may upregulate PBF expression, leading to PBF secretion andincreased cell invasion. Furthermore, the number of ERE half-sites in the PBF promoter may significantly alterthe response to estrogen treatment in individual subjects. Cancer Res; 70(9); 3739–49. ©2010 AACR.

Introduction

Breast carcinogenesis requires multiple genetic changes, in-cluding the altered expression and function of tumor suppres-sor genes and oncogenes. Most human breast cancers evolvefrom normal epithelial cells in terminal duct lobular unitsthrough a series of increasingly abnormal stages over long pe-riods of time. Key stages in this progression are hyperplasia,atypical hyperplasia, in situ carcinoma, invasive carcinoma,and, finally, metastatic disease (1). Invasion into surroundingstroma defines the transition from in situ to invasive carcino-ma. However, most defects responsible for the developmentand progression of malignant disease remain unknown (1).Described in only nine publications (2–10), four of which

are from our own group (2, 3, 6, 8), Pituitary tumor trans-

ffiliations: 1School of Clinical and Experimental Medicine,r Liver Research, Institute of Biomedical Research, andsearch UK Clinical Trials Unit, School of Cancer Sciences,of Birmingham, United Kingdom and 4Department oflogy and Therapeutics, Roswell Park Cancer Institute,w York

ding Author: Christopher J. McCabe, Reader in Endocrinehool of Clinical and Experimental Medicine, Institute for Bio-search, University of Birmingham, Birmingham, B15 2TH,gdom. Phone: 44-121-415-8713; Fax: 44-121-415-8712;[email protected].

8/0008-5472.CAN-09-3531

erican Association for Cancer Research.

journals.org

Researcon Decembecancerres.aacrjournals.org nloaded from

forming gene (PTTG) binding factor (PBF) was identifiedthrough its ability to interact with PTTG1, the human securin(4). First isolated in 1997 (11), PTTG1 is an estrogen-regulatedgene (12, 13) previously implicated in breast cancer, withhighest expression in invasive and metastatic breast cancers(14). Its binding partner PBF has not, however, been studiedin the context of breast cancer before.Initially identified in 1998 (10), PBF comprises six exons

spanning 24 kb within chromosomal region 21q22.3. The180–amino acid peptide sequence of PBF shares no signifi-cant homology with other human proteins but is highly con-served across a wide diversity of animal species (73%homology to mouse, 67% frog, 60% chicken, 52% zebra fish),suggesting both unique functionality and significant evolu-tionary importance.PBF is ubiquitously expressed (15), but a decade after its

cloning, very little has been reported concerning its func-tion. We previously characterized PBF expression in thyroidcancers and showed it to be a transforming gene in vitro andto be tumorigenic in vivo (8). Furthermore, high PBF expres-sion was independently associated with poor prognosis inhuman thyroid cancer. Most recently, we showed that PBFrepressed iodide uptake in thyroid cells, both through tran-scriptional regulation (3) and through altered subcellulartrafficking (6).PBF is thus a relatively uncharacterized transforming gene

that plays a part in multiple cellular processes, particularly inthe setting of endocrine neoplasia. We now present extensive

3739

h. r 24, 2020. © 2010 American Association for Cancer

Page 2: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

Watkins et al.

3740

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

data, outlined below, that suggest PBF represents an entirelynovel gene of direct relevance to breast cancer.

Materials and Methods

Tissues. Breast tumor paraffin-embedded samples ar-ranged on tissue microarrays (TMA) were available from146 patients (16). There were 6 cases of ductal carcinomain situ, 4 cases of medullary carcinoma, 1 atypical medullarycarcinoma, 7 cases of mucinious type, 2 cases of tubular car-cinoma, 22 cases of lobular carcinoma, 1 lobular-papillary,and 1 benign tumor. A further 101 cases were designatedpathologically as being of no special type (breast tumorsnot fitting the histologic categories above but tending to beinvasive ductal carcinomas that could not be further charac-terized on morphologic grounds; ref. 16). Clinical follow-up,encompassing tumor grade, vascular invasion, lymph nodestage, Nottingham prognostic index, and estrogen receptor(ER) status was available for the breast cancer series. Normalbreast paraffin-embedded samples were available from USBiomax (n = 6).Cell lines and hormonal treatments. MCF-7 human

Caucasian breast adenocarcinoma cells were obtained fromthe European Collection of Animal Cell Cultures in April2008. Low passage number cells obtained from the originalstock were maintained in RPMI 1640 (Life Technologies) sup-plemented with 10% fetal bovine serum (FBS). Cells weretreated with diethylstilbestrol (DES) and 17β-estradiol(EST) at final concentrations of 10 nmol/L and 20 nmol/L,respectively, and with ICI 182780 (Faslodex/Fulvestrant)at 100 nmol/L and 1 μmol/L in RPMI 1640 phenol red–freemedium (Life Technologies) supplemented with 10%charcoal-stripped serum.RNA extraction, reverse transcription quantitative PCR,

and Western blot analysis. Total RNA was extracted fromMCF-7 cells using Sigma Trizol kit, as previously described(3). RNA was isolated from paraffin-embedded tissues on mi-croscope slides using the Pinpoint Slide RNA Isolation Systemkit (Zymo Research). RNA was reverse transcribed usingReverse Transcription System (Promega), as previouslydescribed (3). Expression of specific mRNAs was determinedusing 7500 Real-Time PCR System (Applied Biosystems;ref. 8). Western blot analyses were performed as we havedescribed previously (2, 6, 17, 18).Cross-linking chromatin immunoprecipitation. Briefly,

MCF-7 cells were cross-linked by addition of 1% final concen-tration formaldehyde directly to 1.5 × 106 midexponentialcells. After sonication samples were immunoprecipitatedwith Protein A-agarose beads (Upstate Biotech) with 5 μgER antibody (Santa Cruz), alongside an IgG sample as a back-ground control (Abcam). Immunocomplexes were thensequentially washed, and cross-links were removed beforeDNA extraction. PBF promoter primers of sequence 5′-GCA-GCC-CTT-TAG-GAT-GGA-G and 5′-GAG-GAA-AGG-AGC-CTG-GTA-GC were then used with 5 μL of chromatinimmunoprecipitation (ChIP) DNA material for analysis bysemiquantitative PCR.

Cancer Res; 70(9) May 1, 2010

Researcon Decembecancerres.aacrjournals.org Downloaded from

DNA extraction. Genomic DNA from normal and tumor-ous colorectal samples and normal thyroid specimens hadpreviously been extracted (17, 19). Normal DNA was also ob-tained from whole blood from patients with normal thyroidfunction (20). Genomic DNA was isolated from breast tumorand normal tissue, which had been formalin fixed and paraf-fin embedded using the Pinpoint Slide DNA Isolation Systemkit (Zymo Research).PCR. Two primer sets were designed to amplify the region

of the PBF promoter, which contains a repeated 18-bpsequence. Forward (F) and reverse (R) sequences wereprimer set 1(F) 5′-GCG-CTC-CCC-TAG-TCC-CCT-3′, primerset 1(R) 5′-GCG-AGG-AGA-GCG-GCT-GA-3′; primer set 2(F)5′-GCA-GCC-CTT-TAG-GAT-GGA-G-3′, primer set 2(R)5′-GAG-GAA-AGG-AGC-CTG-GTA-GC-3′. Product sizes were220 and 286 bp, respectively. Sequencing was carried out toconfirm estrogen response element (ERE) repeat numbersusing the forward primer of primer set 1 or 2.Luciferase assays. The pGL3_PBFpromoter construct was

created by inserting bases −510 to −211 relative to the trans-lational start site into the pGL3 basic vector (Promega). Cellswere harvested in passive lysis buffer (Promega). The DualLuciferase Reporter Assay System (Promega) was used, anddata were expressed as a ratio of Renilla luciferase activity.Biotinylated oligonucleotide pull-down assay. Pull-down

assays were essentially as described previously (21). Oligonu-cleotides of a consensus ERα binding sequence (22), 5′-GTC-CAA-AGT-CAG-GTC-ACA-GTG-ACC-TGA-TCA-AAG-TT-3′,PBF ERE sequence (half-sites in bold), 5′-CTC-GCC-CCT-CGG-TCA-CGC-CTC-GCC-CCT-CGG-TCA-CGC-CTC-GCC-CCT-CGG-TCA-CGC-CTC-GCC-CCT C-3′ and mutant PBFERE sequence (base changes underlined), 5′-CTC-GCC-CCT-CAA-TTT-CGC-CTC-GCC-CCT-CAA-TTT-CGC-CTC-GCC-CCT-CAA-TTT-CGC-CTC-GCC-CCT-C-3′ were 5′ end-labeledwith biotin and incubated with recombinant ERα protein(P2187; Invitrogen, Ltd.). In competition reactions, recombi-nant protein was additionally incubated with 1 to 2 nmol ofnonbiotinylated oligonucleotide. DNA/protein complexeswere captured with 0.1 mg of magnetic streptavidin beads(Promega). Subsequently, bound proteins were probed withan anti-ERα antibody (Santa Cruz).Immunohistochemistry. Normal and tumorous breast

specimens were immunostained using our specific rabbitpolyclonal antibody to PBF (1:200). For negative controls,the primary antibody was replaced by 2% normal serum. Sec-tions were counterstained with Mayer's hematoxylin, andblinded scoring was carried out according to the intensity(0, not present; +1, least intense; +3, most intense) and per-centage (<25, 25–50, 50–75, >75) of PBF staining.Invasion assays. MCF-7 cells transfected with vector only

(VO), hemagglutinin (HA)–tagged PBF (PBF-HA), or the PBFmutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8 μmol/L). Subsequently, 800 μL of RPMI1640 supplemented with 20% FBS or charcoal-stripped serumwere added to the well below the BD Falcon cell cultureinserts. For knockdown, cells were treated with either50 nmol/L of scrambled (negative control 1, Ambion) orPBF-specific siRNAs 14399 and 147350 (mixed in equal

Cancer Research

h. r 24, 2020. © 2010 American Association for Cancer

Page 3: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

PBF in Breast Cancer

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

quantities; Ambion). After 24 or 48 hours, cells were fixed andstained using Mayer's H&E (Sigma).MTT assays. Cells were transfected or treated with DES

and EST at final concentrations of 10 and 20 nmol/L. At 24and 48 hours post-PBF transfection or 48 hours post-estrogentreatment, 100 μg of MTT were added to each well, as pre-viously described (23).Detection of PBF secretion by Western blotting. Cell

lysates were harvested in radioimmunoprecipitation assay(RIPA) buffer. Cell medium was removed from MCF-7 cellsand centrifuged to remove cellular debris. Supernatantswere then added to three volumes of 100% ethanol andcentrifuged, and the pellet was resuspended in RIPA buffer.Detection of PBF secretion by immunoprecipitating

radiolabeled PBF. Cells were maintained in 1 mL of mediumcontaining 2/3 standard medium and 1/3 leucine-free equi-valent, along with 10 μCi L-leucine [3,4,5-3H] (MP Biomedi-cals). Media extracts were centrifuged to remove floatingcells. To obtain cell lysate fractions, cells were lysed in RIPAbuffer. Rabbit polyclonal anti-PBF antibody (5 μL; ref. 6) ornormal rabbit control serum (5 μL) was added to cell med-ium and lysate fractions. Immunocomplexes were pelleted bycentrifugation at 13,000 × g, and dpm was measured. Western

www.aacrjournals.org

Researcon Decembecancerres.aacrjournals.org Downloaded from

blot analysis of PBF immunoprecipitation confirmed thatPBF was specifically pulled down.Immunofluorescent analysis of PBF vesicular localization.

MCF-7 cells were transfected with 1 μg PBF-HA and 1 μgchromogranin A-GFP (24) on coverslips. Cells were fixedand permeabilized before blocking. Rabbit polyclonal anti-HA (Y-11) antibody (Santa Cruz Biotechnology) was usedas primary antibody, and Alexa Fluor 594–conjugated goatanti-rabbit IgG (Invitrogen) was used as secondary antibody.Hoechst stain was used to visualize nuclei (1:1,000).Statistical analysis. Data were analyzed using Sigma Stat

(SPSS Science Software UK Ltd.). Normally distributed datawere analyzed using a two-sample Student's t test. TheMann-Whitney rank-sum test was used for comparison bet-ween the two groups of nonparametric data. Data containingcategorical information were analyzed using the χ2 test andFisher's exact test.

Results

PBF mRNA and protein expression are induced byestrogen. The binding partner of PBF (PTTG1) is anestrogen-regulated gene implicated in the etiology of breast

Figure 1. A, PBF mRNA expression was induced maximally at 48 h by 20 nmol/L DES and EST in ERα-positive MCF-7 cells compared withvehicle-treated cells. ΔCt values from Taqman RT-PCR with associated SEM are given below. B, PBF protein expression levels were upregulated by20 nmol/L DES and EST. Scanning densitometry (n = 3 experiments) was used to calculate a ratio of expression compared with β-actin. Vehicle treatment(V) = 1.0. C, estrogen (EST) and anti-estrogen ICI 182780 treatment of MCF-7 cells at 48 h. V, vehicle. D, cross-linking ChIP analysis of ERα bindingto the human PBF promoter, yielding an expected product of 286 bp. IgG, immunoglobin control; NTC, no template control. *, P < 0.05; **, P < 0.01;***, P < 0.001.

Cancer Res; 70(9) May 1, 2010 3741

h. r 24, 2020. © 2010 American Association for Cancer

Page 4: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

Watkins et al.

3742

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

cancer (14). We therefore examined whether PBF mightalso be estrogen regulated. PBFmRNA expressionwas induced48 hours after treatment by 10 and 20 nmol/L DES and 10 and20 nmol/L EST in ERα-positive MCF-7 cells (Fig. 1A). PBF pro-tein expression levels were also significantly upregulated 2-foldto 3-fold compared with vehicle controls (Fig. 1B). Further-more, treatment with the anti-estrogen ICI 182780 at 100nmol/L and 1 μmol/L concentrations repressed EST stimula-tion of PBF expression in a dose-dependent manner (Fig. 1C).To investigate whether EST stimulation of PBF expression

occurred directly through the PBF promoter, cross-linkingChIP assays were carried out as described in Materials andMethods. In each case (n = 4 experiments), ERα bound to thehuman PBF promoter, with most pronounced binding occur-ring between 24 and 48 hours post-estrogen treatment inMCF-7 cells (Fig. 1D).

Cancer Res; 70(9) May 1, 2010

Researcon Decembecancerres.aacrjournals.org Downloaded from

The human PBF promoter is polymorphic for ERE half-sites.In silico analysis of the human PBF promoter identifiedthe region −399 to −292 relative to the translational startsite to be replete with putative EREs (Fig. 2A). Preliminarysequencing of this region revealed that it contained variablerepeat numbers of an 18-bp sequence housing a putativeconsensus ERE half-site (gcccctcGGTCAcgcctc; Fig. 2A).PCR and sequencing of the region were additionallyexamined with a separate set of primers, confirming theexistence of polymorphic numbers of 18-bp repeats (Fig. 2A).A panel of 92 genomic DNA samples was available to us,

DNA being prepared from normal thyroid and colon tissueand from tumorous colon. PCR and sequencing revealedthat subjects may be homozygous or heterozygous forbetween one and six repeats of the 18-bp region housingthe ERE (Fig. 2B). We next examined ERE repeat number

Figure 2. Polymorphism of the PBF promoter. A, a hypervariable region between −399 and −292 upstream of the ATG contains between 1 and 6 repeats ofan 18-bp motif, which houses a putative consensus ERE half-site (GGTCA). TSS, transcriptional start site. Bottom, PCR analysis of the PBF promoter,showing negative control (−ve), positive control (+ve; plasmid containing three repeats), and two individuals with 3 and 5 (3/5) and 3 and 6 (3/6) repeats ofthe 18-bp motif. L, ladder. An alternative set of primers (Primer Set 2) was used to amplify the PBF promoter region. B, allele frequencies for one to sixhalf-site repeats in n = 122 subjects. C, estrogen responsiveness of the promoter region −510 to −211 was examined through luciferase assays at24 and 48 h posttransfection in MCF-7 cells with 20 nmol/L DES and EST. Data were corrected for Renilla activity and are expressed relative to vehicletreatment. D, oligonucleotide pull-down assay of recombinant human ERα binding to three ERE half-sites in the human PBF promoter (PBF). NEG, negativecontrol, lacking rPBF oligonucleotide; ERE, biotinylated consensus double stranded ERE oligonucleotide (22); PBF Mut, mutated EREs from the humanPBF promoter. *, P < 0.05; **, P < 0.01.

Cancer Research

h. r 24, 2020. © 2010 American Association for Cancer

Page 5: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

PBF in Breast Cancer

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

in formalin-fixed paraffin-embedded breast TMA samples. Ofthe∼60 tumors assessed, 27 tumors gave informative sequenc-ing data for ERE repeat number, and three of the six normalsyielded unambiguous sequence data. These studies revealedthat breast tumors could either be homozygous (three andthree repeats) or heterozygous (three and five repeats), whereasthe n = 3 normal breast specimens were all homozygous forthree repeats (Fig. 2B). None of the other variantswere detectedin breast DNA. Overall, as each 18-bp repeat houses a putativeconsensus ERE half-site, subjects therefore differ in the numberof EREs present in their PBF promoter.The promoter region −399 to −292 is estrogen responsive.

Having determined that PBF is regulated by estrogen and thatERα binds to the PBF promoter in ChIP assays and havingidentified a polymorphic region in the human PBF promotercontaining variable numbers of putative ERE half-sites, wenext examined the estrogen responsiveness of this fragmentof the promoter. DES and EST (20 nmol/L) both induced sig-nificant luciferase activity compared with vehicle-treatedcells after 24 hours (Fig. 2C). At 48 hours, the effect was morepronounced (DES, 1.8 ± 0.2-fold, P < 0.01 compared with ve-hicle, n = 3; EST, 1.7 ± 0.3-fold, P < 0.05 compared with vehicle,n = 3; Fig. 2C).Next, we investigated whether the polymorphic ERE half-

sites identified within the promoter region −399 to −292 werecapable of binding ERα directly. We performed pull-down as-says using oligonucleotides containing either a series of threewild-type (WT) or three mutated PBF ERE half-site repeats.Recombinant human ERα bound specifically to the biotiny-lated double-stranded PBF ERE oligonucleotide (Fig. 2D),which could be competed out by incubation with increasingexcesses of unlabeled PBF oligonucleotide. Furthermore,recombinant ERα bound a biotinylated consensus double-stranded ERE oligonucleotide, and binding was repressedby competition with unlabeled WT PBF ERE oligonucleotidebut not by the mutated PBF oligonucleotide in which theEREs had been abolished. As PBF mRNA was induced 1.5-foldto 3-fold at identical time points and at identical doses ofestrogen (Fig. 1), these data indicate that the short promoterregion −510 to −211 is positively regulated by estrogen andconfers most, but not all, of PBF's responsiveness to DESand EST. In addition, the specific ERE half-site region iden-tified within the promoter region −399 to −292 is capable ofbinding ERα, suggesting that the main mechanism of estro-gen regulation of PBF is directly via the polymorphic EREs ofthe proximal promoter.PBF expression and correlation in a breast tumor series.

We next investigated whether PBF was expressed in humanbreast tumors. Initial mRNA investigations used 20 breastTMA specimens and 6 normal breast specimens. PBF mRNAexpression was apparent in RNA extracted from 18 of 20 TMAtumor samples, with a mean Taqman reverse transcription–PCR (RT-PCR)ΔCt of∼8 to 10, suggesting robustly detectablelevels of expression (Fig. 3A). Reverse transcriptase negativecontrols confirmed that amplification in the tumor samplesexamined was not an artifact of genomic DNA contamination(data not shown). In contrast, PBF mRNA was not detected inany of the six normal breast specimens analyzed (Fig. 3A).

www.aacrjournals.org

Researcon Decembecancerres.aacrjournals.org Downloaded from

PBF protein expression was next quantified in a largerseries of normal breast specimens (n = 8) and TMA tumorsamples (n = 146) through immunohistochemistry, usingour rabbit polyclonal antibody (6). Examples of scoring in-tensities are provided in Fig. 3B. PBF expression was lowor absent in normal breast tissue, whereas it was stronglyexpressed in epithelial cells of all tumor types and gradesof breast tumor assessed (Fig. 3C). Specificity of stainingwas confirmed in negative control experiments (data notshown). Importantly, ERα status positively and significantlycorrelated with the percentage of PBF protein expression(P < 0.001). However, the remaining phenotypic end-pointsexamined (tumor grade, vascular invasion, lymph nodestage, or Nottingham prognostic index) were not associatedwith PBF staining intensity or percentage expression.Next, we investigated the relationship between ERE num-

ber and PBF mRNA expression. Eight fixed tumor specimensand three normal breast samples yielded both mRNA andpromoter sequencing data. PBF expression was apparent in86% of three and five heterozygotes but only 25% of three andthree homozygotes (P = 0.044). Thus, PBF is estrogen regula-ted, its expression is higher in ER-positive than ER-negativetumors, and a greater number of ERE repeats is associatedwith higher PBF expression.PBF and estrogen both induce invasiveness in MCF-7

cells. The exact function of PBF in cell transformation isnot known (8). Given that PBF is induced in breast cancerand that invasion and metastasis are critical processes inbreast cancer progression, we next assessed whether PBFmight play a role in cell invasion.When 1 × 105 cells per well were seeded in invasion assays,

PBF overexpression was associated with a 2.5 ± 0.6-foldincreased cell invasion compared with VO treatment at24 hours (n = 6, P < 0.01) and a 6.1 ± 2.9-fold increase at48 hours (n = 6, P < 0.01; Fig. 4A). At a density of 2 × 105 cellsper well, PBF induced a 2.5 ± 0.3-fold increase in cell invasionafter 48 hours (n = 6, P < 0.01).To determine whether the enhanced invasiveness of

MCF-7 cells following PBF transfection reflected an increasein cell proliferation, MCF-7 cells were transfected with VO orPBF, and cell number estimated after 24 and 48 hours usingMTT assays (Fig. 4B). PBF did not significantly increase theproliferation of MCF-7 cells after 24 hours but did marginally(∼10%) increase cell number after 48 hours. These datasuggest that increased proliferation does not explain PBF'sinfluence upon invasion.PBF confers estrogen induction of MCF-7 cell invasion.

As estrogen induces PBF and MCF-7 cells are well documen-ted as an estrogen-responsive cell line, we next examined theinfluence of treatment with 10 nmol/L DES on cell invasion.DES treatment induced MCF-7 cell invasion by ∼2.3-foldcompared with vehicle treatment (n = 3, P < 0.001; Fig. 5A).DES and EST (10 and 20 nmol/L) treatment did not signifi-cantly alter MCF-7 cell proliferation (data not shown).Subsequently, we validated transient knockdown of PBF in

MCF-7 cells using siRNA. PBF-specific siRNA (50 nmol/L) eli-cited ∼80% to 90% knockdown compared with a scrambledcontrol (Fig. 5B). Cell invasion assays were then repeated

Cancer Res; 70(9) May 1, 2010 3743

h. r 24, 2020. © 2010 American Association for Cancer

Page 6: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

Watkins et al.

3744

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

Figure 3. A, expression of PBF mRNA relative to 18 s rRNA (ΔCt values) in 20 TMA samples of breast tumors compared with normal breast. ND, notdetected after 40 cycles of PCR. B, representative immunohistochemical examination of PBF staining in one normal breast sample (US BioMax) andseven tumor samples from TMA sections. Columns 1 to 4 represent the different staining intensities observed, from 0 (absent) to +3 (intense). Values in thebottom right-hand corners indicate the percentage of PBF expression observed in the whole section, with original magnifications annotated next totumor type. C, representative immunohistochemical examination of PBF staining in three normal breast samples (N1–N3; US BioMax; ×40 originalmagnification) and three tumor samples from TMA sections. T1, Grade I; T2, Grade II; T3, Grade III; all ×40 original magnification.

Cancer Res; 70(9) May 1, 2010 Cancer Research

Research. on December 24, 2020. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

PBF in Breast Cancer

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

after 24 hours in the presence of 10 nmol/L DES or vehicle.Critically, the increased cell invasion observed after DEStreatment in the presence of a scrambled control (103 ± 17cells, P = 0.002, n = 2) was abolished when PBF was simul-taneously knocked down using a PBF-specific siRNA (27 ±14 cells, P = not significant, n = 2; Fig. 5C). These data suggestthat estrogen induction of MCF-7 cell invasion throughMatrigel is mediated via PBF.PBF is a secreted protein. As the induction of invasion

by PBF could not be explained by increased cell number

www.aacrjournals.org

Researcon Decembecancerres.aacrjournals.org Downloaded from

and because cell invasion is a process frequently associatedwith secretion, we assessed whether PBF is a secreted protein.MCF-7 cells were grown in the presence of L-leucine [3,4,5-3H],cell medium fractions were harvested after 24 hours, and im-munoprecipitations were carried out with our PBF antibody(Fig. 6). Immunoprecipitation of labeled PBF revealed thatPBF is indeed a secreted protein with vector-only transfectedMCF-7 cells showing that ∼20% of total cellular PBF is sec-reted over the experimental time frame of 24 hours (Fig. 6A).This was significantly enhanced by transient overexpression

Figure 4. A, PBF overexpressionsignificantly increased the invasivenessof MCF-7 cells after both 24 and48 h compared with VO. Representativeimages of invading cells (arrow) stainedwith Mayer's hematoxylin (nuclear, blue)and eosin (cytoplasm, pink) are presentedabove. **, P < 0.01. B, MCF-7 cellnumber following transfection with VOor PBF assessed through MTT assays.*, P < 0.05, n = 4.

Cancer Res; 70(9) May 1, 2010 3745

h. r 24, 2020. © 2010 American Association for Cancer

Page 8: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

Watkins et al.

3746

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

of PBF (49 ± 3% secretion, n = 3, P < 0.05 compared with VO).Furthermore, a mutant of PBF lacking amino acids 29 to 93(and hence, a functional signal peptide and two potential pu-tative glycosylation sites; ref. 6) showed reduced secretion intothe cell medium comparedwithWT (28 ± 7%, n = 3) but did notdiffer statistically from VO (Fig. 6A).To confirm that PBF is secreted into the cell medium, we

further carried out Western blotting for PBF-HA. Cell ly-sates showed successful transfection of PBF-HA (Fig. 6B).Wild-type PBF is a putative glycoprotein (10), which runsas a doublet at around 25 to 30 kDa. Mutant Δ29-93 was,as anticipated, smaller and ran at ∼20 kDa (Fig. 6B).Whereas WT PBF was easily detectable in cell media, con-firming that PBF is a secreted protein, mutant Δ29-93 wasnot apparent.To investigate the mechanism by which PBF is secreted,

MCF-7 cells were cotransfected with PBF-HA and chromo-granin A-GFP. Chromogranin A-GFP is a chimeric photopro-tein that is transported via the regulated pathway forexocytosis (24) and has recently been used as a marker forsecretory granules in MCF-7 cells (25). As shown previouslyin other cell lines, PBF-HA was found within intracellular ve-sicles, often detected toward the periphery of cells (6), in

Cancer Res; 70(9) May 1, 2010

Researcon Decembecancerres.aacrjournals.org Downloaded from

keeping with a secretory phenotype (Fig. 6C). Although thesevesicles were localized in a similar pattern to the chromog-ranin A-GFP–labeled secretory granules, no colocalizationwith PBF was observed. This suggests that PBF is secretedvia the constitutive pathway of secretion rather than viaregulated secretion. Overall then, PBF is secreted by MCF-7cells, higher expression results in increased secretion, and de-letion of the amino acid region Δ29-93 results in significantlyattenuated secretion into the cell medium.Induction of MCF-7 cell invasion is modulated by PBF

secretion. Having shown that PBF is secreted by MCF-7 cells,we investigated the relationship between secretion and inva-sion. As before, PBF induced significant cell invasion whenoverexpressed (269 ± 52 invading cells, n = 10 experiments,P = 0.018 compared with VO control; Fig. 6C). However, mu-tant Δ29-93 failed to induce cell invasion compared with VO(161 ± 45 invading cells, n = 10 experiments, P = 0.518 com-pared with VO). Thus, secretion of PBF contributes signifi-cantly to its induction of cell invasion.In summary, PBF is a relatively uncharacterized proto-

oncogene that is induced by estrogen in MCF-7 cells, showsincreased expression in breast cancer, and stimulates cellinvasion, at least in part through secretion. On this basis,

h. r 24, 2020. © 2010 American

Figure 5. A, 10 nmol/Ldiethylstilbesterol treatment of1 × 105 native MCF-7 cells inducedsignificant invasion throughMatrigel at 24 h. B, siRNAknockdown of PBF. DES (10 nmol/L)induced PBF in the presenceof a scrambled siRNA controlcompared with vehicle treatment.However, 50 nmol/L PBF siRNAyielded 80% to 90% knockdown ofprotein, which was not altered byDES treatment. C, invasion assayscarried out in parallel toknockdown experiments. V,vehicle. Arrows, invading cells.

Cancer Research

Association for Cancer

Page 9: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

PBF in Breast Cancer

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

Figure 6. Effect of WT and mutant PBF overexpression on the secretion and invasiveness of MCF-7 cells. A, the percentage of total PBF secreted intothe medium extracted from MCF-7 cells. Mutant Δ29-93 lacks amino acids 29 to 93. B, Western blot of cell medium and whole-cell lysate extracted fromMCF-7 cells transfected with VO, WT PBF, and the Δ29-93 PBF mutant. PBF constructs were HA-tagged. Whereas WT PBF is detectable in the cellmedium, mutant Δ29-93 is not. C, immunofluorescent subcellular analysis of MCF-7 cells cotransfected with PBF-HA and chromogranin A-GFP. i and iv,vesicular PBF-HA expression (red); ii and v, chromogranin A-GFP expression; iii and vi, merged image of PBF-HA and chromogranin A-GFP. D, cell invasionassays. In contrast to WT PBF, mutant Δ29-93 failed to induce statistically significant cell invasion. Columns, mean values; bars, SEM. n = 10.

Cancer Res; 70(9) May 1, 2010www.aacrjournals.org 3747

Research. on December 24, 2020. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

Watkins et al.

3748

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

PBF warrants further and intensive study in the context ofbreast cancer initiation and progression.

Discussion

Numerous genetic changes governing the initiation, pro-gression, and metastasis of breast cancer have already beendescribed, but new genetic markers and therapeutic targetsare vital for continued progress in addressing the approxi-mately half-million global deaths annually from the disease.Based on our investigations, we propose four principle linesof evidence suggesting a critical role of PBF in breast can-cer. First, PBF is highly expressed in breast tumors, whereits expression correlates with ER positivity. Second, it isregulated by DES and EST, both at the mRNA and proteinlevel. Third, PBF upregulation results in significant MCF-7cell invasion through Matrigel, a phenomenon highly de-pendent upon PBF secretion. Fourth, repressing PBF ex-pression in the face of estrogen stimulation preventsestrogen-mediated induction of cell invasion. Hence, estro-gen regulates PBF expression in MCF-7 cells, tumors showincreased levels of the protein, and its functional propertyin promoting invasion can be ameliorated both throughmutation and knockdown.Although highly conserved, the 180–amino acid peptide se-

quence of PBF shares no significant homology with other hu-man proteins. Its exact mechanisms of action have not beendivined, but it has been shown to induce tumors in nudemice (8), to regulate expression and function of the sodiumiodide symporter (3, 6), and to interact with the human se-curin PTTG (4). Our experiments are the first to report a rolefor PBF in the breast and to elucidate a function in cell inva-sion. We now propose that estrogen regulates PBF mRNAand protein expression and suggest that this is mediated pre-dominantly by a cluster of ERE half-sites ∼300 bp upstreamof the translational start site.One surprising finding of our preliminary sequence ana-

lysis was that the human PBF promoter is polymorphic foran 18-bp repeat housing a putative ERE half-site. Impor-tantly, a higher number of putative EREs were statisticallyassociated with greater PBF mRNA expression in the breastand ER-positive tumors having significantly increased PBFprotein expression compared with ER-negative tumors. Al-though these findings would need to be confirmed in a lar-ger series of samples, a higher number of EREs in a breasttumor would therefore predict a greater response to circu-lating estrogen and hence increased expression of a knowntransforming gene.Estrogen has previously been shown to enhance expres-

sion of PTTG, the binding partner of PBF (12, 13), andPTTG upregulation has been described in breast cancer(14). Whereas it was not the focus of the current investiga-tion, it would be interesting to correlate PTTG and PBF ex-pression in individual breast tumors. The functionalimplications of simultaneous overexpression are hard togauge, given that both proteins are inherently multifunc-tional and likely to have dependent and independent modesof action. However, future studies may delineate the

Cancer Res; 70(9) May 1, 2010

Researcon Decembecancerres.aacrjournals.org Downloaded from

individual contributions of each gene to breast cancer ini-tiation and progression.Our in vitro experiments were predominantly carried out in

MCF-7 cells, which remain the “gold standard” ERα-positiveand estrogen-sensitive breast cell line (26). Furthermore,MCF-7 cells are weakly invasive (27), allowing us to performphysiologically relevant invasion assays.Our wider clinical associations did not reveal striking as-

sociations between PBF expression and tumor phenotype,which was unexpected. Clinical associations were hamperedby a lack of a matched normal/tumor cohort, which wouldallow a more detailed interrogation of the association bet-ween promoter polymorphism and clinical outcome. Fur-thermore, fresh tissue would have allowed us to performWestern blotting for PBF expression, which would haveprovided more quantitative expression data than throughimmunohistochemistry on FFPE slides.The mechanism by which PBF induces cell transformation

is not known. Our current study suggests that this might bevia the induction of cell invasion. MCF-7 cells showed po-tent increases in invasion through Matrigel in response toPBF transfection. A mutant PBF, which was not secretedinto the cell medium, lost this phenotype. Furthermore, aspecific siRNA entirely blocked estrogen induction of cellinvasion. Thus, we hypothesize that estrogen induces PBF,which in turn drives breast cell invasion, at least in partthrough secretion. As estrogen stimulation of PBF expres-sion was abrogated by cotreatment with the anti-estrogenICI 182780, ER antagonists might be useful in treatingtumors with high PBF expression to block potential cellinvasion mediated by PBF.In summary, we present evidence that the poorly charac-

terized proto-oncogene PBF has particular relevance tobreast tumorigenesis, particularly with respect to progres-sion. Based on our in vitro and ex vivo findings, we predictthat estrogen treatment in postmenopausal women mayupregulate PBF expression, leading to increased PBF secre-tion. Given that individuals have different numbers of ERErepeats, it is likely that estrogen will stimulate PBF expres-sion variably, with a potentially critical effect upon breastcell invasion.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We thank Prof. Richard Cheney (University of North Carolina) for providingthe chromogranin A-GFP construct.

Grant Support

IDA Cooper Foundation and Medical Research Council.The costs of publication of this article were defrayed in part by the payment

of page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received 09/28/2009; revised 02/05/2010; accepted 02/13/2010; publishedOnlineFirst 4/20/2010.

Cancer Research

h. r 24, 2020. © 2010 American Association for Cancer

Page 11: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

PBF in Breast Cancer

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531

References

1. Allred DC, Medina D. The relevance of mouse models to understan-

ding the development and progression of human breast cancer.J Mammary Gland Biol Neoplasia 2008;13:279–88.

2. Boelaert K, Tannahill LA, Bulmer JN, et al. A potential role for PTTG/securin in the developing human fetal brain. FASEB J 2003;17:1631–9.

3. Boelaert K, Smith VE, Stratford AL, et al. PTTG and PBF repress thehuman sodium iodide symporter. Oncogene 2007;26:4344–56.

4. Chien W, Pei L. A novel binding factor facilitates nuclear transloca-tion and transcriptional activation function of the pituitary tumor-transforming gene product. J Biol Chem 2000;275:19422–7.

5. Mo Z, Zu X, Xie Z, et al. Antitumor effect of F-PBF(β-TrCP)-inducedtargeted PTTG1 degradation in HeLa cells. J Biotechnol 2009;139:6–11.

6. Smith VE, Read ML, Turnell AS, et al. A novel mechanism of sodiumiodide symporter repression in differentiated thyroid cancer. J CellSci 2009;122:3393–402.

7. Stock M, Schafer H, Fliegauf M, Otto F. Identification of novel genesof the bone-specific transcription factor Runx2. J Bone Miner Res2004;19:959–72.

8. Stratford AL, Boelaert K, Tannahill LA, et al. Pituitary tumor trans-forming gene binding factor: a novel transforming gene in thyroidtumorigenesis. J Clin Endocrinol Metab 2005;90:4341–9.

9. Tfelt-Hansen J, Yano S, Bandyopadhyay S, Carroll R, Brown EM,Chattopadhyay N. Expression of pituitary tumor transforming gene(PTTG) and its binding protein in human astrocytes and astrocytomacells: function and regulation of PTTG in U87 astrocytoma cells.Endocrinology 2004;145:4222–31.

10. Yaspo ML, Aaltonen J, Horelli-Kuitunen N, Peltonen L, Lehrach H.Cloning of a novel human putative type Ia integral membrane proteinmapping to 21q22.3. Genomics 1998;49:133–6.

11. Pei L, Melmed S. Isolation and characterization of a pituitary tumor-transforming gene (PTTG). Mol Endocrinol 1997;11:433–41.

12. Heaney AP, Horwitz GA, Wang Z, Singson R, Melmed S. Early in-volvement of estrogen-induced pituitary tumor transforming geneand fibroblast growth factor expression in prolactinoma pathogene-sis. Nat Med 1999;5:1317–21.

13. Heaney AP, Fernando M, Melmed S. Functional role of estrogen inpituitary tumor pathogenesis. J Clin Invest 2002;109:277–83.

14. Ogbagabriel S, Fernando M, Waldman FM, Bose S, Heaney AP.Securin is overexpressed in breast cancer. Mod Pathol 2005;18:985–90.

15. Chen L, Puri R, Lefkowitz EJ, Kakar SS. Identification of the human

www.aacrjournals.org

Researcon Decembecancerres.aacrjournals.org Downloaded from

pituitary tumor transforming gene (hPTTG) family: molecular struc-ture, expression, and chromosomal localization. Gene 2000;248:41–50.

16. Murray PG, Lissauer D, Junying J, et al. Reactivity with A monoclonalantibody to Epstein-Barr virus (EBV) nuclear antigen 1 defines asubset of aggressive breast cancers in the absence of the EBVgenome. Cancer Res 2003;63:2338–43.

17. Kim D, Pemberton H, Stratford AL, et al. Pituitary tumour transform-ing gene (PTTG) induces genetic instability in thyroid cells.Oncogene 2005;24:4861–6.

18. Kim DS, Franklyn JA, Stratford AL, et al. Pituitary tumor-transforminggene regulates multiple downstream angiogenic genes in thyroidcancer. J Clin Endocrinol Metab 2006;91:1119–28.

19. Kim DS, Franklyn JA, Smith VE, et al. Securin induces geneticinstability in colorectal cancer by inhibiting double-stranded DNArepair activity. Carcinogenesis 2007;28:749–59.

20. Simmonds MJ, Heward JM, Carr-Smith J, Foxall H, Franklyn JA,Gough SC. Contribution of single nucleotide polymorphisms withinFCRL3 and MAP3K7IP2 to the pathogenesis of Graves' disease.J Clin Endocrinol Metab 2006;91:1056–61.

21. Liu Y, Asch H, Kulesz-Martin MF. Functional quantification of DNA-binding proteins p53 and estrogen receptor in cells and tumor tissuesby DNA affinity immunoblotting. Cancer Res 2001;61:5402–6.

22. Vyhlidal C, Samudio I, Kladde MP, Safe S. Transcriptional activationof transforming growth factor α by estradiol: requirement for both aGC-rich site and an estrogen response element half-site. J MolEndocrinol 2000;24:329–38.

23. James SR, Franklyn JA, Reaves BJ, et al. Monocarboxylate trans-porter 8 (MCT8) in neuronal cell growth. Endocrinology 2008.

24. Taupenot L, Harper KL, Mahapatra NR, Parmer RJ, Mahata SK,O'Connor DT. Identification of a novel sorting determinant for theregulated pathway in the secretory protein chromogranin A. J CellSci 2002;115:4827–41.

25. Jacobs DT, Weigert R, Grode KD, Donaldson JG, Cheney RE. MyosinVc is a molecular motor that functions in secretory granule trafficking.Mol Biol Cell 2009;20:4471–88.

26. Brooks SC, Skafar DF. From ligand structure to biological activity:modified estratrienes and their estrogenic and antiestrogenic effectsin MCF-7 cells. Steroids 2004;69:401–18.

27. Hazan RB, Phillips GR, Qiao RF, Norton L, Aaronson SA. Exogenousexpression of N-cadherin in breast cancer cells induces cell migra-tion, invasion, and metastasis. J Cell Biol 2000;148:779–90.

Cancer Res; 70(9) May 1, 2010 3749

h. r 24, 2020. © 2010 American Association for Cancer

Page 12: Tumor and Stem Cell Biology Cancer Research Pituitary ... · mutant Δ29-93 were seeded onto BD Falcon cell culture in-serts (pore size, 8μmol/L). Subsequently, 800μLofRPMI 1640

2010;70:3739-3749. Published OnlineFirst April 20, 2010.Cancer Res   Rachel J. Watkins, Martin L. Read, Vicki E. Smith, et al.   Gene in Breast CancerPituitary Tumor Transforming Gene Binding Factor: A New

  Updated version

  10.1158/0008-5472.CAN-09-3531doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/70/9/3739.full#ref-list-1

This article cites 26 articles, 7 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/70/9/3739.full#related-urls

This article has been cited by 8 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/70/9/3739To request permission to re-use all or part of this article, use this link

Research. on December 24, 2020. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3531