Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Tumor and Stem Cell Biology

Androgen and Estrogen Receptors in Breast Cancer Coregulate Human UDP-Glucuronosyltransferases 2B15 and 2B17

Dong G. Hu, Luke A. Selth, Gerard A. Tarulli, Robyn Meech, Dhilushi Wijayakumara, Apichaya Chanawong, Roslin Russell, Carlos Caldas, Jessica L.L. Robinson, Jason S. Carroll, Wayne D. Tilley, Peter I. Mackenzie and Theresa E. Hickey
Dong G. Hu
1Department of Clinical Pharmacology, Flinders University School of Medicine, Flinders Medical Centre, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Luke A. Selth
2Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide, South Australia, Australia.
3Freemasons Foundation Centre for Men's Health, School of Medicine, The University of Adelaide, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gerard A. Tarulli
2Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Robyn Meech
1Department of Clinical Pharmacology, Flinders University School of Medicine, Flinders Medical Centre, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Dhilushi Wijayakumara
1Department of Clinical Pharmacology, Flinders University School of Medicine, Flinders Medical Centre, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Apichaya Chanawong
1Department of Clinical Pharmacology, Flinders University School of Medicine, Flinders Medical Centre, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Roslin Russell
4Breast Cancer Genomics Group, Cancer Research UK, Cambridge Institute, Cambridge University, Cambridge, United Kingdom.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Carlos Caldas
4Breast Cancer Genomics Group, Cancer Research UK, Cambridge Institute, Cambridge University, Cambridge, United Kingdom.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jessica L.L. Robinson
5Nuclear Transcription Factor Laboratory, Cancer Research UK, Cambridge Institute, Cambridge University, Cambridge, United Kingdom.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jason S. Carroll
5Nuclear Transcription Factor Laboratory, Cancer Research UK, Cambridge Institute, Cambridge University, Cambridge, United Kingdom.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wayne D. Tilley
2Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide, South Australia, Australia.
3Freemasons Foundation Centre for Men's Health, School of Medicine, The University of Adelaide, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Peter I. Mackenzie
1Department of Clinical Pharmacology, Flinders University School of Medicine, Flinders Medical Centre, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Theresa E. Hickey
2Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide, South Australia, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: theresa.hickey@adelaide.edu.au
DOI: 10.1158/0008-5472.CAN-15-3372 Published October 2016
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Glucuronidation is an enzymatic process that terminally inactivates steroid hormones, including estrogens and androgens, thereby influencing carcinogenesis in hormone-dependent cancers. While estrogens drive breast carcinogenesis via the estrogen receptor alpha (ERα), androgens play a critical role as prohormones for estrogen biosynthesis and ligands for the androgen receptor (AR). In this study, the expression and regulation of two androgen-inactivating enzymes, the UDP-glucuronosyltransferases UGT2B15 and UGT2B17, was assessed in breast cancer. In large clinical cohorts, high UGT2B15 and UGT2B17 levels positively influenced disease-specific survival in distinct molecular subgroups. Expression of these genes was highest in cases positive for ERα. In cell line models, ERα, AR, and the transcription factor FOXA1 cooperated to increase transcription via tandem binding events at their proximal promoters. ERα activity was dependent on FOXA1, facilitated by AR activation, and potently stimulated by estradiol as well as estrogenic metabolites of 5α-dihydrotestosterone. AR activity was mediated via binding to an estrogen receptor half-site 3′ to the FOXA1 and ERα-binding sites. Although AR and FOXA1 bound the UGT promoters in AR-positive/ERα-negative breast cancer cell lines, androgen treatment did not influence basal transcription levels. Ex vivo culture of human breast tissue and ERα+ tumors provided evidence for upregulation of UGT2B15 and UGT2B17 by estrogen or androgen treatment. ERα binding was evident at the promoters of these genes in a small cohort of primary tumors and distant metastases. Collectively, these data provide insight into sex steroid receptor-mediated regulation of androgen-inactivating enzymes in ERα+ breast cancer, which may have subtype-specific consequences for disease progression and outcomes. Cancer Res; 76(19); 5881–93. ©2016 AACR.

Introduction

Breast and prostate cancer are the most common forms of hormone-dependent cancer and account for a major proportion of cancer-related deaths in women and men, respectively. Breast cancer is predominantly driven by aberrant estrogen receptor alpha (ERα) signaling and prostate cancer by aberrant androgen receptor (AR) signaling. These diseases display multiple similarities of etiology and pathology (1). There has been mounting interest in the role of AR in breast cancer following identification of a subgroup (∼12%) that lacks expression of ERα but is AR+ and displays features reminiscent of prostate cancers (2, 3). However, the large majority (>70%) of breast cancers express both sex hormone receptors, and the role of AR in ERα+ breast cancer may be antiestrogenic and, thereby, tumor suppressive (4).

Inactivation and disposal of steroids regulates target cell exposure to steroid receptor agonists, and derangement of these pathways is evident in the pathology of hormone-dependent cancers (5, 6). Several enzymatic pathways inactivate steroids, but glucuronidation is the only irreversible event. Uridine 5′-diphospho (UDP) glucuronosyltransferases (UGT) create steroid conjugates incapable of binding to cognate receptors. The resulting steroid glucuronides are more hydrophilic and more readily excreted from tissues than unconjugated steroids. Therefore, glucuronidation is a vital mechanism that regulates steroid receptor activity within hormone-responsive tissues. Metabolism of estrogenic hormones has been widely investigated in breast cancer (6), but metabolism of androgenic hormones has yet to be extensively investigated in relation to this disease.

Among 19 human UGTs, only a few glucuronidate sex steroids to any significant extent (7, 8). The most potent natural androgens, testosterone and 5α-dihydrotestosterone (DHT), are glucuronidated by UGT2B15 and UGT2B17. Testosterone is a major precursor for synthesis of 17β-estradiol (E2), the most potent natural ERα ligand, while testosterone and DHT are AR ligands. Hence, the activity of UGT2B15 and UGT2B17 functionally impacts ERα and AR activity, but the significance of UGT2B15 and UGT2B17 expression has not been examined in relation to the different molecular subtypes and clinical outcomes of breast cancer.

Primary breast tissues express UGT2B15 and UGT2B17 (9), with evidence of transcriptional upregulation by ERα signaling in breast cancer cell lines (10, 11). One study also reported that DHT weakly stimulated expression of these genes in an ERα-dependent manner (10). The latter finding is feasibly due to metabolism of DHT into 3α-androstanediol (3α-diol) or 3β-androstanediol (3β-diol), compounds that have some estrogenic activity (12), but this has not been tested. Finally, regulation of UGT2B15 and UGT2B17 by AR signaling has not been thoroughly investigated in an ERα+ context and never in an AR+, ERα− breast cancer context, in which the consequences of androgen-metabolizing capacity may markedly differ. The current study was undertaken to fill these gaps in knowledge by examining expression in large clinical cohorts and elucidating mechanisms of transcriptional regulation by sex steroid receptor signaling in breast cancer, taking into account the molecular complexity of this disease.

Materials and Methods

Chemicals and cell culture

Steroids and steroid receptor antagonists were purchased from Sigma-Aldrich, including E2, DHT, 5α-androstane-3α,17β-diol (3α-diol), 5α-androstane-3β,17β-diol (3β-diol), methyltrienolone (R1881), 4-hydroxy-flutamide (Flut), fulvestrant (ICI 182,780), and bicalutamide. Enobosarm was provided by the manufacturer (GTx, Inc). The MCF7, ZR75-1, and MDA-MB-453 breast cancer cell lines were obtained from the ATCC and the MFM223 line from the German Collection of Microorganisms and Cell Cultures (DMSZ); these were regularly verified for identity via short tandem repeat analyses. MCF7 and ZR75-1 cells were maintained in RPMI1640 medium (Invitrogen), MDA-MB-453 cells in DMEM (Gibco), and MFM223 cells in EMEM (Gibco) containing either 5% (v/v; MCF7, ZR75-1, MDA-MB-453) or 10% (v/v; MFM223) FBS (Gibco) at 37°C and 5% CO2.

Clinical tissues and ex vivo culture

Normal breast tissues from reduction mammoplasties were obtained from Flinders Medical Centre (Adelaide, Australia). Primary ERα+ breast tumor samples were obtained from the Burnside Private Hospital (Adelaide, Australia). All tissues were collected after provision of informed, written consent by the donors. This study was approved by the University of Adelaide Human Research Ethics Committee (approval numbers: H-065-2005; H-169-2011). Tissue pieces were cultured on hydrated gelatin sponges (3–4/sponge) as described previously (13, 14). After a 36-hour preincubation period, hormones were added to the media as indicated and tissues harvested 24 hours later for RNA extraction. Representative pieces of uncultured tissue were fixed in 4% formalin in PBS at 4°C overnight, then processed into paraffin blocks. Sections (2 μm) were (i) stained with hematoxylin and eosin and examined by a pathologist to quantify the relative proportion of tumor cells; and (ii) immunostained for ERα and AR, as described previously (14). Treatment conditions included vehicle (0.01% ethanol), E2 (10 nmol/L), DHT (10 nmol/L), enobosarm (100 nmol/L).

RNA extraction and transcriptional analyses

Cell lines were cultured for 3 days in phenol red-free medium supplemented with 5% dextran-coated charcoal (DCC)-stripped FBS, then seeded under the same conditions into 6-well plates at 5 × 105 cells per well. Three days later, cells were treated for 24 hours with treatments as indicated. RNA extraction, reverse transcription, and qRT-PCR were performed as previously reported (11, 15, 16). For tissue samples, replicate pieces representing one condition were snap frozen in one tube containing RNAlater (Qiagen) and RNA isolated using RNeasy Kits (Qiagen) according to the manufacturer's protocol. Reverse transcription was performed using total RNA (500 ng–1 μg) and an iScript cDNA Synthesis Kit (Bio-Rad). The resulting cDNA was diluted 1:10 and used for transcriptional analysis as above.

Testosterone glucuronidation assay

MCF7 cells were precultured for 72 hours in phenol red-free RPMI1640 supplemented with 5% DCC–FBS in T175 flasks (Gibco), then treated with vehicle control (0.01% ethanol), E2 (10 nmol/L), DHT (10 nmol/L), 3α-diol (100 nmol/L), or 3β-diol (100 nmol/L) for 72 hours. Cells were scraped from flasks and whole-cell lysates prepared in TE buffer (10 nmol/L Tris-HCl and 1 mmol/L EDTA, pH 7.6). Protein concentration was determined by Bradford assay (Bio-Rad), according to the manufacturer's protocol. The testosterone glucuronidation assay was conducted as previously reported (17).

Luciferase reporter assays

MCF7 cells were plated into 96-well dishes in 100 μL of phenol red-free RPMI1640 supplemented with 5% DCC–FBS and cultured for 24 to 48 hours prior to transfection. Cells were transfected with wild-type or mutated UGT2B15 or UGT2B17 promoter constructs, treated with hormones as indicated, and assessed for luciferase activity as described previously (11, 15). Promoter construct details are provided in Supplementary Methods. Transfections were performed in triplicate and experiments repeated at least twice.

Electrophoretic mobility shift and supershift assays

MCF7 cells were cultured in T75 flasks for 6 days in phenol red-free RPMI1640 supplemented with 5% DCC–FBS, then treated overnight (∼16 hours) with 0.1% ethanol (vehicle), E2 (10 nmol/L), DHT (10 nmol/L), 3α-diol (100 nmol/L), or 3β-diol (100 nmol/L). Nuclear extracts were prepared and electrophoretic mobility shift and supershift assays (EMSA) performed as previously reported (11, 18). Probe and antibody information is provided in Supplementary Methods.

siRNA knockdown experiments

ON-TARGETplus SMARTpool siRNAs were purchased from Dharmacon RNA Technologies: (i) FOXA1 (NM_004496); (ii) ERα (NM_000125); (iii) AR (NM_001011645); and (iv) ON-TARGETplus nontargeting pool siRNA (scramble siRNA). Cells were cultured for 3 days in phenol red-free RPMI1640 supplemented with 5% DCC–FBS, then seeded into 6-well plates at 5 × 105 cells per well in 2 mL of fresh medium containing 8 μL of Lipofectamine 2000 (Invitrogen) and 100 nmol/L siRNA. After 24 hours, transfection media were replaced by fresh media supplemented with 5% DCC–FBS. After 48 hours, cells were treated overnight as indicated. Cells were harvested for RNA isolation and transcriptional analyses as above.

Chromatin immunoprecipitation experiments

Standard methodologies were employed as described previously for chromatin immunoprecipitation (ChIP), followed by qRT-PCR (11) and for ChIP followed by exonuclease digestion and deep sequencing (ChIP-exo; ref. 19). Immunoprecipitations were performed with Santa Cruz Biotechnology, Inc. antibodies to ERα (HC-20), FOXA1 (H-120), AR (N-20), or rabbit preimmune IgG control (sc-2027).

Statistical analyses

Log-transformed data met the assumption of equal variance at a significance level of less than 0.01 (Levene test). Analyses of transformed data were conducted by one- or two-way ANOVA with the Tukey post hoc test or independent t test using SPSS software (IBM, version 22). P < 0.05 was considered statistically significant. Recursive partitioning was used to dichotomize the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) transcriptional data to assess the influence of relatively high versus low expression of each gene on patient survival using an R statistical language program as previously implemented (20). This was performed for all probes representing a gene.

Results

Expression of UGT2B15 and UGT2B17 in clinical breast cancers

The METABRIC investigated 2,000 cases of breast cancer and redefined this heterogeneous disease into 10 molecular subgroups with distinct survival outcomes (20). The subgroups, called iClusters, were created by an integration of gene copy number variations and cis-acting transcriptomes (20, 21). Analysis of the microarray probes representing UGT2B15 and UGT2B17 in the METABRIC cohort revealed cases that displayed very high expression of these genes (Fig. 1A). Their expression was positively correlated in this (Supplementary Fig. S1A) as well as an independent breast cancer cohort (n = 825 cases) from The Cancer Genome Atlas (Supplementary Fig. S1B; refs. 22, 23). In the METABRIC cohort, ERα+ cases had significantly higher expression of these genes compared to ERα− cases (Fig. 1B), and highest expression was observed in integrative cluster 1 (iCluster 1), predominantly composed of ERα+, luminal B breast cancers (Fig. 1C). Integrative cluster 10, largely composed of aggressive, triple-negative breast cancers, had the lowest expression (Fig. 1C). Transcript levels of both enzymes significantly decreased with increasing tumor grade (Fig. 1D). Higher levels of UGT2B17 were significantly associated with increased survival outcomes in cases classified into iCluster 5, largely composed of high-grade breast cancers with HER2 amplification and very poor survival outcomes, as well as all cases in the cohort classified as being HER2 amplified by SNP6 array analysis (Fig. 1E). Note that these groups contain both ERα+ and ERα− breast cancers. Higher levels of both UGT2B15 and UGT2B17 were associated with increased survival in iCluster 6 (Fig. 1F), dominated by ERα+, HER2− breast cancers with amplification of the 8p12 gene locus. Conversely, higher levels of UGT2B15 were associated with poor survival in iCluster 9, composed of ERα+ cancers with a luminal B phenotype and a high rate of TP53 mutations (Fig. 1G). We also analyzed the relationship between expression of these genes and patient outcome in another publicly available resource (KM plotter) containing normalized microarray data from multiple studies that collectively represent 3,455 cases of breast cancer (24). At present, this cohort can only be interrogated via the older PAM50 gene classifier. A high level of UGT2B15 expression was associated with improved relapse-free survival in the entire cohort and in all PAM50 subgroups (Supplementary Fig. S2, top row). Although high UGT2B17 expression was also associated with improved relapse-free survival in the entire cohort, this association was only sustained in the luminal B and HER2+ subtypes (Supplementary Fig. S2, bottom row), the latter supporting results in Fig. 1E. Collectively, these data suggest that expression of UGT2B15 or UGT2B17 impacts breast cancer survival in a gene- and disease subtype-specific manner.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

UGT2B15 and UGT2B17 expression in clinical breast cancers. A, heatmap showing relative expression of UGT2B15 (2 probes) and UGT2B17 (3 probes) in the METABRIC cohort of 2,000 breast cancers. The degree of red or blue color indicates high and low expression, respectively. The 10 integrative clusters (iClusters) defined by the METABRIC study are indicated by colors that correspond to the boxes in C. B, UGT2B15 and UGT2B17 gene expression is significantly higher in ERα+ compared with ERα− breast cancers. C, levels of UGT2B15 and UGT2B17 in the iClusters. D, UGT2B15 and UGT2B17 expression significantly decreases with increasing tumor grade. E, high expression of UGT2B17 is associated with increased disease-specific survival in iCluster 5, representing cases in which HER2 amplification is the key genomic feature (left) and in all cases with HER2 amplification as determined by SNP6 array (right). F, high expression of UGT2B15 (left) and UGT2B17 (right) is associated with better disease-specific survival in iCluster 6. G, high expression of UGT2B15 is associated with decreased disease-specific survival in iCluster 9. Box and whisker plots (B–D), median and interquartile range (25th–75th percentiles; ***, P < 0.0001).

ERα and AR transcriptionally regulate UGT2B15 and UGT2B17 in an estrogen-sensitive context

As expected, E2 significantly increased transcripts of UGT2B15 and UGT2B17 (Fig. 2A and B) and an ERα-regulated positive control gene (GREB1; Supplementary Fig. S3A), effects that were abolished by cotreatment with fulvestrant (ICI), an ERα inhibitor. Treatment with DHT or its metabolites, 3α-diol or 3β-diol, dose dependently increased UGT2B15 and UGT2B17 transcripts (Fig. 2A and B) and the control gene (Supplementary Fig. S3A) to varying degrees. These stimulatory effects were all significantly attenuated by coincubation with fulvestrant, supporting the hypothesis that DHT indirectly stimulates ERα activity via conversion to diols. Individual hormone treatments also significantly increased levels of glucuronidated testosterone (Fig. 2C), indicative of enhanced enzyme activity. Surprisingly, fulvestrant also inhibited the activity of R1881 (Fig. 2A and B), a synthetic androgen that is not metabolized to estrogenic compounds, although direct binding of R1881 to ERα at high doses has been reported (25). While these data collectively suggested lack of direct regulation by AR, subsequent analyses revealed that the stimulatory effects of DHT were also significantly attenuated by treatment with flutamide, an AR antagonist (Supplementary Fig. S3B), and treatment with the ERα and AR inhibitors combined resulted in greater inhibition of steroid-mediated upregulation of UGT2B15 and UGT2B17 than either inhibitor alone (Fig. 2D). These complementary effects were not evident for regulation of GREB1 by DHT, which was completely dependent on ERα (Supplementary Fig. S3C). Consistent with a direct role for AR in upregulating the UGT genes, siRNA-mediated ablation of AR in MCF7 cells significantly reduced both E2- and DHT-mediated transcriptional effects (Fig. 2E). The same patterns of hormone regulation and the influence of AR loss were evident for the UGT2B15 gene in ZR75-1 cells, another model of ERα+AR+ breast cancer (Supplementary Fig. S3D). The UGT2B17 gene was not detected under basal or hormone stimulation in ZR75-1 cells. Collectively, these findings indicate that AR directly regulates UGT2B15 and UGT2B17 in ERα+AR+ breast cancer cells, acting cooperatively with ERα.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Regulation of UGT2B15 and UGT2B17 by estrogenic and androgenic steroids. A and B, relative mRNA levels of UGT2B15 (A) and UGT2B17 (B) in MCF7 cells treated for 24 hours with estrogen (E2), androgen (DHT), metabolites of DHT (3α-diol, 3β-diol), or a synthetic androgen (R1881) alone or in combination with the ERα inhibitor fulvestrant (ICI) at concentrations as indicated. C, levels of glucuronidated testosterone, indicative of UGT2B15/17 enzyme activity, in MCF7 cells treated with hormones as indicated. D, influence of ERα inhibition (ICI) and/or AR inhibition with flutamide (Flut) on upregulation of UGT2B15 (top) and UGT2B17 (bottom) by DHT in MCF7 cells. E, influence of siRNA-mediated silencing of AR (siAR) versus a nontarget siRNA (siCtrl) on E2- or DHT-mediated induction of UGT2B15 (top) or UGT2B17 (bottom) in MCF7 cells. All data shown are means ± SEM of three independent experiments. Asterisks above columns, comparisons with the vehicle control, set at 1. Asterisks above a line, comparisons between selected columns (one-way ANOVA followed by Tukey post hoc multiple comparison test; *, P < 0.05; **, P < 0.01; ***, P < 0.001).

Steroid-induced transactivation of the proximal promoters of UGT2B15 and UGT2B17

Highly conserved estrogen response units (ERU) reside in the proximal promoters of the UGT2B15 and UGT2B17 genes, comprised of three estrogen response element (ERE) half-sites and two AP-1–binding sites, all required for E2-mediated transcriptional regulation (11). A FOXA1-binding site has recently been identified in the region between two of the ERE half-sites (11, 16), but its importance in breast cancer cells is unknown. The prototypical ERU, including the FOXA1-binding site, is depicted in Fig. 3A. As expected, E2 significantly enhanced basal promoter activity in an ERα-dependent manner (Fig. 3B). All other steroids, including DHT, 3α-diol, 3β-diol, and R1881, stimulated UGT2B15 promoter activity at levels similar to or greater than that induced by E2, and these stimulatory effects were all significantly repressed to some degree by the ERα inhibitor. However, the AR inhibitor also significantly reduced DHT-induced promoter activity (Fig. 3C), supporting direct regulation by both sex steroid receptors.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Estrogen and androgen regulation of the UGT2B15 and UGT2B17 promoters. A, schematic depiction of highly conserved ERUs in the UGT2B15 and UGT2B17 proximal promoters, positioned between −454 and −172 nucleotides in the UGT2B15 promoter and between nucleotides −396 and −112 in the UGT2B17 promoter relative to their translation start sites (ATG). From 5′ to 3′, the six key binding motifs are 5′-AP-1 and 3′-AP-1 sites, an imperfect ERE, a 5′-ERE half-site, a FOXA1 site, and a 3′-ERE half-site. MCF7 cells were transfected with wild-type (WT) promoter constructs or mutated constructs as indicated by a backslash over the relevant motif. B, stimulation of the WT UGT2B15 promoter by estrogen (E2), androgen (DHT), metabolites of DHT (3α-diol, 3β-diol), or a synthetic androgen (R1881), alone or together with the ERα inhibitor fulvestrant (ICI). C, inhibition of DHT-stimulated WT UGT2B15 promoter activity by ICI and an AR inhibitor, flutamide (Flut). D, influence of UGT2B15 promoter mutations on steroid-mediated transactivation. E, activity of WT and FOXA1-mutant (FOXA1mt) UGT2B17 promoters in response to steroids alone or in combination with ICI. Relative luciferase activities of promoter constructs are expressed as fold induction over that of the empty vector (set to 1) after normalizing transfection efficiency with R. reniformis luciferase. Data shown are means ± SD of three independent experiments performed in triplicate (one-way ANOVA followed by Tukey post hoc multiple comparison test. *, P < 0.05; **, P < 0.01; ***, P < 0.001).

An ERU-mutant UGT2B15 promoter construct dramatically reduced transactivation by E2 and obliterated transactivation by other tested steroids (Fig. 3D; left), and mutation of the FOXA1 site abolished all steroid-induced promoter activity (Fig. 3D; right). Transactivation of the wild-type (Fig. 3E; left) and FOXA1-mutated (Fig. 3E; right) UGT2B17 promoter constructs by steroids mirrored that of the UGT2B15 constructs, but with more attenuated stimulatory effects.

To confirm the direct involvement of FOXA1, EMSA assays were performed. A major protein/DNA complex formed on a DNA probe containing the UGT2B15 promoter FOXA1 site following incubation with nuclear extracts from vehicle and steroid-treated MCF7 cells (Fig. 4A). Addition of a FOXA1 antibody reduced the intensity of the complex and also induced formation of super-shifted complexes. Furthermore, mutation of the FOXA1 site abolished the formation of FOXA1 protein/DNA complexes. Although the UGT2B15 and UGT2B17 gene promoters are highly conserved, a unique but prevalent −155A/G SNP occurs within the FOXA1-binding site in the UGT2B17 promoter (15). Therefore, a UGT2B17 probe with an A-containing FOXA1 site was tested (Fig. 4B). A major complex was formed on the A-containing UGT2B17 FOXA1 probe, which was supershifted by addition of FOXA1 antibody and abolished by mutating the FOXA1 site or adding a 100-fold molar excess of unlabeled probe. Therefore, endogenous FOXA1 in MCF7 cells can bind the “A-containing” FOXA1-binding site SNP in the UGT2B17 promoter. These results confirm a critical role for FOXA1 in steroid-mediated transactivation of the UGT2B15 and UGT2B17 promoters, even in the context of a common SNP variant in UGT2B17.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

FOXA1, ERα and AR enrichment at UGT2B15 and UGT2B17 promoters. A and B, EMSA blot to demonstrate FOXA1 binding to DNA fragments representing the UGT2B15 (A) and UGT2B17 (B) promoters. MCF7 cells were treated with vehicle (lanes 1–8), estrogen (E2; lanes 9–12), androgen (DHT; lanes 13–14), DHT metabolites 3α-diol (lanes 15–16), or 3β-diol (lanes 17–18). EMSAs were performed with 50,000 cpm (∼1 ng) of indicated 32P-labeled probes. FOXA1-bound DNA complexes are labeled A for the PSA control probe, B for UGT2B15 or UGT2B17 probes, C for nonspecific complexes, and SS for antibody-induced super-shifted complexes. For supershift assays, FOXA1 antibody was added (lanes 4, 7, 12, 14, 16, and 18). For competition assays, unlabeled probes were added at a 10-fold (lane 11) or 100-fold (lane 10) molar excess. In lane 8, a mutated FOXA1 probe was added. C–E, ChIP–qPCR data showing enrichment of ERα, FOXA1, and AR chromatin binding at UGT2B15 (C), UGT2B17 (D), or pS1/TFF (E) promoters in MCF7 cells following stimulation with indicated steroids. Data are presented as relative fold enrichment over the normal rabbit immunoglobulin (IgG) controls (set to 1). Bars, means ± SEM of three independent experiments (one-way ANOVA followed by Tukey post hoc multiple comparison test. *, P < 0.05; **, P < 0.01;***, P < 0.001 compared with the relevant IgG control).

Recruitment of ERα, FOXA1, and AR to the UGT2B15 and UGT2B17 promoters

ERα occupancy at the UGT2B15 and UGT2B17 proximal promoters and a positive control locus (pS2/TIF1) was significantly stimulated by E2 in MCF7 cells (Fig. 4C–E). Of the other steroids tested, 3β-diol stimulated the highest level of ERα enrichment, a compound with significantly less affinity for ERα than E2 (26). FOXA1 bound to the UGT2B15 and UGT2B17 loci before ligand stimulation, consistent with its role as a pioneer factor (27). Interestingly, FOXA1 binding was significantly increased by all hormone treatments (Fig. 4C–E), contrary to observations at other genomic loci (28). This steroid-induced increase in FOXA1 occupancy is consistent with a recent study showing that FOXA1 binding can be altered by activation of steroid receptors, including ERα, in breast cancer cells (29). The AR did not display a strong enrichment over IgG controls under any treatment condition; maximal AR enrichment (2.6-fold) occurred at the positive control locus for ERα binding (pS2/TIFF1) when cells were stimulated with R1881. Low enrichment of AR may be a technical issue due to low AR protein levels in MCF7 cells. However, the requirement for ERα, FOXA1, and AR in the regulation of UGT2B15 and UGT2B17 by steroids was confirmed by siRNA-mediated ablation (Supplementary Fig. S4). Analysis of genome-wide ERα, FOXA1, and AR chromatin binding in MCF7 and ZR75-1 cells generated by ChIP-exo (19) revealed the presence of all three factors in the proximal promoters of the UGT genes (Fig. 5A and B; magnified views in Supplementary Fig. S5). ChIP-exo generates sharper peaks than ChIP sequencing (ChIP-seq) due to the inclusion of an exonuclease step following immunoprecipitation of protein–DNA complexes, allowing better discernment of relative binding positions. These data suggest that AR binds to or overlaps the ERE half-site located 3′ to the FOXA1 and ERα peaks in the promoters of UGT2B15 and UGT2B17. The tandem pattern of chromatin binding strongly supports the concept that ERα, AR, and FOXA1 interact to regulate transcription of these two androgen-inactivating enzymes in an estrogen-sensitive breast cancer context.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

AR regulation of UGT2B15 and UGT2B17 promoters in ERα+ breast cancer cells. A and B, representative genome browser images showing enrichment of DNA sequences (peaks) pulled down by immunoprecipitation of FOXA1, ERα, or AR by the process of ChIP-exo, indicative of chromatin binding at the proximal promoters of UGT2B15 (A) and UGT2B17 (B) in two models of ERα+, AR+ breast cancer (MCF7, ZR75-1). C, wild-type (WT) UGT2B15 and UGT2B17 promoter constructs were transfected into MCF7 cells, followed by treatment with the selective AR modulator, enobosarm (Eno), as indicated. D, influence of site-specific mutations (MT) in the UGT2B15 promoter constructs transfected into MCF7 cells followed by treatment with enobosarm (100 nmol/L) alone or in combination with the AR inhibitor, bicalutamide (Bic; 1 μmol/L). NS, not significant. Data in C and D are means ± SD of two independent experiments performed in triplicate. Asterisks above columns, comparisons with the appropriate vehicle (Veh) control, set to 1. Asterisks above lines, comparisons between selected columns (one-way ANOVA, followed by Tukey post hoc multiple comparison test. *, P < 0.05; **, P < 0.01).

Regulation of UGT2B15 and UGT2B17 by enobosarm, a new selective AR modulator

Enobosarm (GTx024) is a selective AR modulator (30) currently being tested for clinical efficacy in women with ERα+AR+ breast cancer (NCT02463032). This compound dose dependently activated an AR reporter construct transfected into MCF7 cells (Supplementary Fig. S6A), upregulated UGT2B15 and UGT2B17 in an AR-dependent manner (Supplementary Fig. S6B), and dose dependently increased activation of the wild-type UGT promoter constructs (Fig. 5C). Using selective ERE-mutant constructs of the UGT2B15 promoter, we show that the previously defined “3′ ERE half-site” is critical for AR-mediated upregulation of promoter activity (Fig. 5D).

AR does not regulate UGT2B15 or UGT2B17 in an estrogen-insensitive context

AR activity sustains a luminal phenotype in MDA-MB-453 cells, a model of AR+ERα− breast cancer, by mimicking the action of ERα in MCF7 cells (31). This type of breast cancer also has similarities to prostate cancer (3). Interrogation of our previously published ChIP-seq data (31) shows that AR and FOXA1 occupy the proximal promoter regions of UGT2B15 and UGT2B17 in MDA-MB-453 cells, similar to ERα and FOXA1 occupancy in MCF7 cells and AR and FOXA1 occupancy in LNCaP prostate cancer cells (Fig. 6A and B). However, treatment with DHT did not upregulate expression of these genes in the MDA-MB-453 or MFM223 breast cancer cell lines (Fig. 6C), which both have a “luminal AR” phenotype (32). UGT2B17 expression was undetectable under any treatment conditions in these AR+ ERα− cell lines. In contrast, DHT potently downregulates both genes in LNCaP cells (33). Thus, although ERα and AR positively coregulate transcription of the two major androgen-metabolizing enzymes in an estrogen-sensitive breast cancer context, AR alone does not appear to have this capacity in an estrogen-insensitive context.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Androgen regulation of UGT2B15 and UGT2B17 promoters in ERα− breast cancer cells. A and B, representative genome browser images showing enrichment (peaks) of DNA sequences pulled down by immunoprecipitation of FOXA1 or AR by the process of ChIP-seq, indicative of chromatin binding at the proximal promoters of UGT2B15 (A) and UGT2B17 (B) in MDA-MB-453 breast cancer cells, an AR+, ERα− model of breast cancer. For comparison, ERα and FOXA1 peaks are shown for the same loci in MCF7 breast cancer cells and AR and FOXA1 peaks in prostate cancer cells (LNCaP). C, qRT-PCR data corresponding to indicated cell lines treated with estrogen (E2) or androgen (DHT). Data shown are means ± SEM of three independent experiments. One-way ANOVA followed by Tukey post hoc multiple comparison test (***, P < 0.0001). NS, not significant; Veh, vehicle.

Estrogen and androgen regulation of UGT2B15 and UGT2B17 in clinical tissues

Normal breast tissue from reduction mammoplasties (n = 8) were cultured ex vivo on gelatin sponges to sustain tissue architecture, cellular heterogeneity, and hormone receptor expression as described previously (14). In 3 of 8 cases, all three treatments (E2, DHT, and enobosarm) upregulated transcript levels of UGT2B15 or UGT2B17 compared with the vehicle control (Supplementary Fig. S6C). In other cases, one or two agents demonstrated stimulatory action, and in a few cases, there were no effects, demonstrating the expected heterogeneity of primary tissue responses. This methodology also provides a unique preclinical model for testing drug responses (13, 34). In primary ERα+ breast tumors (n = 13) cultured ex vivo under estrogenic conditions, treatment with enobosarm upregulated UGT2B15 in a total of 7 of 13 cases and UGT2B17 in 8 of 13 cases (Fig. 7A). Two cases demonstrated no stimulatory effect on either gene above baseline, four demonstrated stimulatory effects on both genes, three demonstrated upregulation of UGT2B15 alone, and four demonstrated upregulation of UGT2B17 alone. Collectively, these data indicate that therapeutic activation of AR can significantly upregulate expression of these UGT genes in primary ERα+ breast cancers.

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

Sex hormone regulation of UGT2B15 and UGT2B17 in clinical samples. A, relative mRNA levels of UGT2B15 and UGT2B17 in ex vivo–cultured pieces of ERα+AR+ primary breast tumors (n = 13 independent cases) following treatment with the selective AR modulator, enobosarm (Eno; 100 nmol/L). B and C, representative genome browser images of ERα occupancy at the UGT2B15 (B) and UGT2B17 (C) proximal promoters in clinical samples, representing normal breast tissue (n = 2), good outcome ERα+, PR+, HER2− tumors (n = 3), poor outcome ERα+, PR±, HER2± tumors (n = 3), and ERα+ distant metastases (n = 3).

A previous study investigated genome-wide ERα chromatin binding in primary breast tumors (35). The cohort consisted of ERα+, progesterone receptor (PR)-positive and HER2− tumors associated with good disease outcomes (n = 8); ERα+, PR−, and/or HER2+ tumors associated with poor outcomes (n = 7) and ERα+ distant metastases (n = 3). Interrogation of those data revealed no evidence for ERα occupancy at the promoters of UGT2B15 or UGT2B17 in normal breast tissues (0/2 cases) or ERα+ tumors associated with a good outcome (0/8 cases). However, ERα occupancy was evident at the UGT2B15 (2/7 cases) and UGT2B17 (1/7 cases) promoters in tumors associated with poor outcomes and in 2 of 3 metastases (Fig. 7B and C). Although the study represented a small number of cases, it was the first study to investigate genome-wide ERα–chromatin interactions in clinical breast tissues.

Discussion

Herein, the potential clinical significance and hormone-mediated regulation of two steroid glucuronidation enzymes, UGT2B15 and UGT2B17, was investigated in relation to breast cancer. These enzymes can functionally impact both ERα and AR activity by directly or indirectly reducing levels of their activating ligands in the tissue microenvironment. Although positive regulation of these genes via ERα activity at their proximal promoters has been previously described in breast cancer cell lines (10, 11), we provide the first evidence that this activity is exquisitely dependent on the pioneer factor FOXA1, is facilitated by AR binding to an adjacent ERE half-site, and is potently stimulated by metabolites of DHT normally considered “weak” estrogens. These data are consistent with the fact that UGT2B15 and UGT2B17 levels were highest in clinical ERα+ breast cancers and in low-grade tumors, which are known to be characterized by high levels of both ERα and AR. Importantly, higher expression of one or both of the UGT enzymes was associated with better survival outcomes in a number of molecular subgroups of ERα+ disease, which may have clinical implications. For example, therapeutic activation of AR with enobosarm significantly increased expression of one or both genes in ex vivo–cultured normal human breast tissues and ERα+ breast tumors. Whether this plays a mechanistic role in attenuating the estrogenic signal or determining clinical responses of women treated with this compound for estrogen-sensitive breast cancer (NCT02463032) remains to be determined.

In breast malignancies, the levels of estrogenic or androgenic hormones can greatly exceed levels measured in plasma or in adjacent nonmalignant breast tissues (36, 37). The altered intracrinology involves deregulation of steroid-metabolizing genes, such as 17β-HSDs, sulfotransferases, 5α-reductases, and estrogen-metabolizing UGTs (38–40). Our data indicate that androgen-metabolizing UGTs are also involved, particularly in the context of ERα+ and/or HER2+ clinical breast cancers, in which relative expression of UGT2B15 and/or UGT2B17 could discern cases with distinct survival outcomes. Although it is tempting to speculate on how relative levels of either enzyme influence steroid intracrinology and hence disease outcome in various breast cancer contexts, it is important to note that UGT2B15 and UGT2B17 have different potencies in the inactivation of androgen hormones and that one or both have been shown to inactivate xenobiotics and drugs relevant to breast cancer, including bisphenol AF (41), exemestane (42), the histone deacetylase inhibitor vorinostat (43), and the major active tamoxifen metabolite, 4-OH-tamoxifen (42, 44). Differential activity of the two enzymes in the glucuronidation of androgens or other compounds may explain gene-specific influences on breast cancer outcomes. We recently found that tamoxifen and its metabolites upregulate UGT2B15 via a mechanism involving ERα (45), which may have implications for resistance to this drug. Collectively, these studies indicate that the influence of UGT2B15 and UGT2B17 expression in breast cancer includes, but also has implications that extend beyond, their ability to metabolize androgen hormones.

Both metabolites of DHT (3α- and 3β-diol) could recruit ERα to the proximal promoters of UGT2B15 and UGT2B17 to stimulate transcription, which in part explains the ERα-dependent stimulatory effects of this androgen. Indeed, rapid conversion of DHT into 3α- and 3β-diol has been previously reported in MCF7 cells (46, 47) and all 4 human 3α-hydroxysteroid dehydrogenases capable of this conversion are expressed in breast tissues (48). However, the action of DHT also stimulated AR binding to the promoters at a site 3′ to the ERα-binding site, and ablation or chemical inhibition of AR reduced estrogen- and androgen-mediated transcriptional regulation of these genes. The cooperative activity of AR and ERα at these loci is suggestive of an “assisted loading” mechanism described for the glucocorticoid receptor, a steroid receptor closely related to AR (49). Cooperative regulation of UGT2B15 by ERα and AR was also evident in ZR75-1 cells, another luminal breast cancer model, indicating that the findings are not limited to one cell line.

There has been increasing interest in the capacity of AR signaling to drive breast cancer, particularly in ERα-negative tumors. The MDA-MB-453 cell line has emerged as the prototypical and most studied model of this type of breast cancer, in which AR is expressed at high level, and treatment with DHT can increase proliferation. Indeed, AR signaling in this cell line shares many similarities to AR signaling in LNCaP cells (2, 31). However, we show that AR and FOXA1 occupy the promoter regions of UGT2B15 and UGT2B17 in MDA-MB-453 cells in a similar manner to their occupancy in LNCaP cells, but androgen treatment did not influence their transcription in two models of AR+, ERα− breast cancer. As AR potently downregulates these genes in LNCaP cells (14) and we show that AR upregulates them in ERα+ breast cancer cells, regulation of these genes by AR is highly context dependent.

In summary, the UGT2B15 and UGT2B17 enzymes are transcriptionally regulated by sex hormone signaling in ERα+ breast cancer cells and are highly expressed in a subset of primary breast cancers, which has significance for survival outcomes in distinct genetic subgroups of this disease. Apart from their influence on the level of active steroids in the microenvironment, these enzymes may also determine the level of active xenobiotics and drugs relevant to breast cancer, which could have implications for response to a range of adjuvant hormone or other therapies. Collectively, these observations add new knowledge about the complexity of breast cancer intracrinology that has implications for other hormone-dependent cancers. The findings provide a basis for further exploration of the role of UGTs in moderating the steroid milieu and modulating drug responses in breast and other cancers.

Disclosure of Potential Conflicts of Interest

C. Caldas reports receiving commercial research grants from AstraZeneca, Genentech, and Roche and is a consultant/advisory board member for AstraZeneca. No potential conflicts of interest were disclosed by the other authors.

Authors' Contributions

Conception and design: D.G. Hu, L.A. Selth, R. Meech, W.D. Tilley, P.I. Mackenzie, T.E. Hickey

Development of methodology: D.G. Hu, L.A. Selth, P.I. Mackenzie, T.E. Hickey

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D.G. Hu, L.A. Selth, D. Wijayakumara, A. Chanawong, C. Caldas, J.L.L. Robinson, J.S. Carroll, W.D. Tilley, T.E. Hickey

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D.G. Hu, L.A. Selth, R. Russell, C. Caldas, J.L.L. Robinson, W.D. Tilley, T.E. Hickey

Writing, review, and/or revision of the manuscript: D.G. Hu, L.A. Selth, G.A. Tarulli, R. Meech, J.L.L. Robinson, J.S. Carroll, W.D. Tilley, T.E. Hickey

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L.A. Selth, G.A. Tarulli, C. Caldas, W.D. Tilley, P.I. Mackenzie, T.E. Hickey

Study supervision: D.G. Hu, L.A. Selth, T.E. Hickey

Grant Support

This work was supported by funding from the National Health and Medical Research Council of Australia (ID1020931 to P.I. Mackenzie; ID1085410 to R. Meech and P.I. Mackenzie; ID 1008349 to W.D. Tilley; ID 1084416 to W.D. Tilley and T.E. Hickey), Flinders Medical Centre Foundation (D.G. Hu and P.I. Mackenzie), Cancer Australia (ID 1043497 to W. D. Tilley and T. E. Hickey), and a Prostate Cancer Research Programs Transformative Impact Award from the U.S. Department of Defense (W81XWH-13-2-0093 to W. D. Tilley and L. A. Selth). L. A. Selth was supported by a Young Investigator Award from the Prostate Cancer Foundation (Foundation 14 award). T.E. Hickey held a Postdoctoral Fellowship Award from the U.S. Department of Defense Breast Cancer Research Program (BCRP; W81XWH-11-1-0592).

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 in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Acknowledgments

We acknowledge Geraldine Laven-Law for technical assistance and the clinical personnel (particularly Drs. Stephen Birrell and Clive Hoffman) and patients from Adelaide clinics who supported the study.

Footnotes

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

  • Received December 10, 2015.
  • Revision received June 20, 2016.
  • Accepted July 14, 2016.
  • ©2016 American Association for Cancer Research.

References

  1. 1.↵
    1. Risbridger GP,
    2. Davis ID,
    3. Birrell SN,
    4. Tilley WD
    . Breast and prostate cancer: more similar than different. Nat Rev Cancer 2010;10:205–12.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Doane AS,
    2. Danso M,
    3. Lal P,
    4. Donaton M,
    5. Zhang L,
    6. Hudis C,
    7. et al.
    An estrogen receptor-negative breast cancer subset characterized by a hormonally regulated transcriptional program and response to androgen. Oncogene 2006;25:3994–4008.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Farmer P,
    2. Bonnefoi H,
    3. Becette V,
    4. Tubiana-Hulin M,
    5. Fumoleau P,
    6. Larsimont D,
    7. et al.
    Identification of molecular apocrine breast tumours by microarray analysis. Oncogene 2005;24:4660–71.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Hickey TE,
    2. Robinson JL,
    3. Carroll JS,
    4. Tilley WD
    . Minireview: the androgen receptor in breast tissues: growth inhibitor, tumor suppressor, oncogene? Mol Endocrinol 2012;26:1252–67.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. Pasqualini JR,
    2. Chetrite GS
    . Recent insight on the control of enzymes involved in estrogen formation and transformation in human breast cancer. J Steroid Biochem Mol Biol 2005;93:221–36.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Guillemette C,
    2. Belanger A,
    3. Lepine J
    . Metabolic inactivation of estrogens in breast tissue by UDP-glucuronosyltransferase enzymes: an overview. Breast Cancer Res 2004;6:246–54.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Turgeon D,
    2. Carrier JS,
    3. Levesque E,
    4. Hum DW,
    5. Belanger A
    . Relative enzymatic activity, protein stability, and tissue distribution of human steroid-metabolizing UGT2B subfamily members. Endocrinology 2001;142:778–87.
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. Lepine J,
    2. Bernard O,
    3. Plante M,
    4. Tetu B,
    5. Pelletier G,
    6. Labrie F,
    7. et al.
    Specificity and regioselectivity of the conjugation of estradiol, estrone, and their catecholestrogen and methoxyestrogen metabolites by human uridine diphospho-glucuronosyltransferases expressed in endometrium. J Clin Endocrinol Metab 2004;89:5222–32.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Nakamura A,
    2. Nakajima M,
    3. Yamanaka H,
    4. Fujiwara R,
    5. Yokoi T
    . Expression of UGT1A and UGT2B mRNA in human normal tissues and various cell lines. Drug Metab Dispos 2008;36:1461–4.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    1. Harrington WR,
    2. Sengupta S,
    3. Katzenellenbogen BS
    . Estrogen regulation of the glucuronidation enzyme UGT2B15 in estrogen receptor-positive breast cancer cells. Endocrinology 2006;147:3843–50.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Hu DG,
    2. Mackenzie PI
    . Estrogen receptor alpha, fos-related antigen-2, and c-Jun coordinately regulate human UDP glucuronosyltransferase 2B15 and 2B17 expression in response to 17beta-estradiol in MCF-7 cells. Mol Pharmacol 2009;76:425–39.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Sikora MJ,
    2. Cordero KE,
    3. Larios JM,
    4. Johnson MD,
    5. Lippman ME,
    6. Rae JM
    . The androgen metabolite 5alpha-androstane-3beta,17beta-diol (3betaAdiol) induces breast cancer growth via estrogen receptor: implications for aromatase inhibitor resistance. Breast Cancer Res Treat 2009;115:289–96.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Dean JL,
    2. McClendon AK,
    3. Hickey TE,
    4. Butler LM,
    5. Tilley WD,
    6. Witkiewicz AK,
    7. et al.
    Therapeutic response to CDK4/6 inhibition in breast cancer defined by ex vivo analyses of human tumors. Cell Cycle 2012;11:2756–61.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Ochnik AM,
    2. Moore NL,
    3. Jankovic-Karasoulos T,
    4. Bianco-Miotto T,
    5. Ryan NK,
    6. Thomas MR,
    7. et al.
    Antiandrogenic actions of medroxyprogesterone acetate on epithelial cells within normal human breast tissues cultured ex vivo. Menopause 2014;21:79–88.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Hu DG,
    2. Gardner-Stephen D,
    3. Severi G,
    4. Gregory PA,
    5. Treloar J,
    6. Giles GG,
    7. et al.
    A novel polymorphism in a forkhead box A1 (FOXA1) binding site of the human UDP glucuronosyltransferase 2B17 gene modulates promoter activity and is associated with altered levels of circulating androstane-3alpha,17beta-diol glucuronide. Mol Pharmacol 2010;78:714–22.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Hu DG,
    2. Mackenzie PI
    . Forkhead box protein A1 regulates UDP-glucuronosyltransferase 2B15 gene transcription in LNCaP prostate cancer cells. Drug Metab Dispos 2010;38:2105–9.
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    1. Wijayakumara DD,
    2. Hu DG,
    3. Meech R,
    4. McKinnon RA,
    5. Mackenzie PI
    . Regulation of human UGT2B15 and UGT2B17 by miR-376c in prostate cancer cell lines. J Pharmacol Exp Ther 2015;354:417–25.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Gardner-Stephen DA,
    2. Gregory PA,
    3. Mackenzie PI
    . Identification and characterization of functional hepatocyte nuclear factor 1-binding sites in UDP-glucuronosyltransferase genes. Methods Enzymol 2005;400:22–46.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Serandour AA,
    2. Brown GD,
    3. Cohen JD,
    4. Carroll JS
    . Development of an Illumina-based ChIP-exonuclease method provides insight into FoxA1-DNA binding properties. Genome Biol 2013;14:R147.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Curtis C,
    2. Shah SP,
    3. Chin SF,
    4. Turashvili G,
    5. Rueda OM,
    6. Dunning MJ,
    7. et al.
    The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 2012;486:346–52.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Dawson SJ,
    2. Rueda OM,
    3. Aparicio S,
    4. Caldas C
    . A new genome-driven integrated classification of breast cancer and its implications. EMBO J 2013;32:617–28.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    1. Gao J,
    2. Aksoy BA,
    3. Dogrusoz U,
    4. Dresdner G,
    5. Gross B,
    6. Sumer SO,
    7. et al.
    Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Cerami E,
    2. Gao J,
    3. Dogrusoz U,
    4. Gross BE,
    5. Sumer SO,
    6. Aksoy BA,
    7. et al.
    The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401–4.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    1. Gyorffy B,
    2. Lanczky A,
    3. Eklund AC,
    4. Denkert C,
    5. Budczies J,
    6. Li Q,
    7. et al.
    An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res Treat 2010;123:725–31.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Akahira JI,
    2. Suzuki T,
    3. Ito K,
    4. Darnel AD,
    5. Moriya T,
    6. Sato S,
    7. et al.
    Expression of 5alpha-reductases in human epithelial ovarian cancer: its correlation with androgen receptor status. Jpn J Cancer Res 2001;92:926–32.
    OpenUrl
  26. 26.↵
    1. Kuiper GG,
    2. Carlsson B,
    3. Grandien K,
    4. Enmark E,
    5. Haggblad J,
    6. Nilsson S,
    7. et al.
    Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 1997;138:863–70.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. Augello MA,
    2. Hickey TE,
    3. Knudsen KE
    . FOXA1: master of steroid receptor function in cancer. EMBO J 2011;30:3885–94.
    OpenUrlAbstract/FREE Full Text
  28. 28.↵
    1. Carroll JS,
    2. Liu XS,
    3. Brodsky AS,
    4. Li W,
    5. Meyer CA,
    6. Szary AJ,
    7. et al.
    Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell 2005;122:33–43.
    OpenUrlCrossRefPubMed
  29. 29.↵
    1. Swinstead EE,
    2. Miranda TB,
    3. Paakinaho V,
    4. Baek S,
    5. Goldstein I,
    6. Hawkins M,
    7. et al.
    Steroid receptors reprogram FoxA1 occupancy through dynamic chromatin transitions. Cell 2016;165:593–605.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Coss CC,
    2. Jones A,
    3. Dalton JT
    . Selective androgen receptor modulators as improved androgen therapy for advanced breast cancer. Steroids 2014;90:94–100.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Robinson JL,
    2. Macarthur S,
    3. Ross-Innes CS,
    4. Tilley WD,
    5. Neal DE,
    6. Mills IG,
    7. et al.
    Androgen receptor driven transcription in molecular apocrine breast cancer is mediated by FoxA1. EMBO J 2011;30:3019–27.
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    1. Lehmann BD,
    2. Bauer JA,
    3. Chen X,
    4. Sanders ME,
    5. Chakravarthy AB,
    6. Shyr Y,
    7. et al.
    Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 2011;121:2750–67.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Chouinard S,
    2. Barbier O,
    3. Belanger A
    . UDP-glucuronosyltransferase 2B15 (UGT2B15) and UGT2B17 enzymes are major determinants of the androgen response in prostate cancer LNCaP cells. J Biol Chem 2007;282:33466–74.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    1. Mohammed H,
    2. Russell IA,
    3. Stark R,
    4. Rueda OM,
    5. Hickey TE,
    6. Tarulli GA,
    7. et al.
    Progesterone receptor modulates ERalpha action in breast cancer. Nature 2015;523:313–7.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Ross-Innes CS,
    2. Stark R,
    3. Teschendorff AE,
    4. Holmes KA,
    5. Ali HR,
    6. Dunning MJ,
    7. et al.
    Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 2012;481:389–93.
    OpenUrlPubMed
  36. 36.↵
    1. Shibuya R,
    2. Suzuki T,
    3. Miki Y,
    4. Yoshida K,
    5. Moriya T,
    6. Ono K,
    7. et al.
    Intratumoral concentration of sex steroids and expression of sex steroid-producing enzymes in ductal carcinoma in situ of human breast. Endocr Relat Cancer 2008;15:113–24.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    1. Suzuki T,
    2. Miki Y,
    3. Nakamura Y,
    4. Moriya T,
    5. Ito K,
    6. Ohuchi N,
    7. et al.
    Sex steroid-producing enzymes in human breast cancer. Endocr Relat Cancer 2005;12:701–20.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Sasano H,
    2. Nagasaki S,
    3. Miki Y,
    4. Suzuki T
    . New developments in intracrinology of human breast cancer: estrogen sulfatase and sulfotransferase. Ann N Y Acad Sci 2009;1155:76–9.
    OpenUrlCrossRefPubMed
  39. 39.↵
    1. Sasano H,
    2. Suzuki T,
    3. Miki Y,
    4. Moriya T
    . Intracrinology of estrogens and androgens in breast carcinoma. J Steroid Biochem Mol Biol 2008;108:181–5.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Suzuki T,
    2. Miki Y,
    3. Moriya T,
    4. Akahira J,
    5. Ishida T,
    6. Hirakawa H,
    7. et al.
    5Alpha-reductase type 1 and aromatase in breast carcinoma as regulators of in situ androgen production. Int J Cancer 2007;120:285–91.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Li M,
    2. Yang Y,
    3. Yang Y,
    4. Yin J,
    5. Zhang J,
    6. Feng Y,
    7. et al.
    Biotransformation of bisphenol AF to its major glucuronide metabolite reduces estrogenic activity. PLoS One 2013;8:e83170.
    OpenUrlCrossRef
  42. 42.↵
    1. Sun D,
    2. Chen G,
    3. Dellinger RW,
    4. Sharma AK,
    5. Lazarus P
    . Characterization of 17-dihydroexemestane glucuronidation: potential role of the UGT2B17 deletion in exemestane pharmacogenetics. Pharmacogenet Genomics 2010;20:575–85.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. Wong NS,
    2. Seah E,
    3. Wang LZ,
    4. Yeo WL,
    5. Yap HL,
    6. Chuah B,
    7. et al.
    Impact of UDP-gluconoryltransferase 2B17 genotype on vorinostat metabolism and clinical outcomes in Asian women with breast cancer. Pharmacogenet Genomics 2011;21:760–8.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Ogura K,
    2. Ishikawa Y,
    3. Kaku T,
    4. Nishiyama T,
    5. Ohnuma T,
    6. Muro K,
    7. et al.
    Quaternary ammonium-linked glucuronidation of trans-4-hydroxytamoxifen, an active metabolite of tamoxifen, by human liver microsomes and UDP-glucuronosyltransferase 1A4. Biochem Pharmacol 2006;71:1358–69.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Chanawong A,
    2. Hu DG,
    3. Meech R,
    4. Mackenzie PI,
    5. McKinnon RA
    . Induction of UDP-glucuronosyltransferase 2B15 gene expression by the major active metabolites of tamoxifen, 4-hydroxytamoxifen and endoxifen, in breast cancer cells. Drug Metab Dispos 2015;43:889–97.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    1. Jorgensen L,
    2. Brunner N,
    3. Spang-Thomsen M,
    4. James MR,
    5. Clarke R,
    6. Dombernowsky P,
    7. et al.
    Steroid metabolism in the hormone dependent MCF-7 human breast carcinoma cell line and its two hormone resistant subpopulations MCF-7/LCC1 and MCF-7/LCC2. J Steroid Biochem Mol Biol 1997;63:275–81.
    OpenUrlCrossRefPubMed
  47. 47.↵
    1. Roy R,
    2. Dauvois S,
    3. Labrie F,
    4. Belanger A
    . Estrogen-stimulated glucuronidation of dihydrotestosterone in MCF-7 human breast cancer cells. J Steroid Biochem Mol Biol 1992;41:579–82.
    OpenUrlCrossRefPubMed
  48. 48.↵
    1. Penning TM,
    2. Jin Y,
    3. Steckelbroeck S,
    4. Lanisnik Rizner T,
    5. Lewis M
    . Structure-function of human 3 alpha-hydroxysteroid dehydrogenases: genes and proteins. Mol Cell Endocrinol 2004;215:63–72.
    OpenUrlCrossRefPubMed
  49. 49.↵
    1. Voss TC,
    2. Schiltz RL,
    3. Sung MH,
    4. Yen PM,
    5. Stamatoyannopoulos JA,
    6. Biddie SC,
    7. et al.
    Dynamic exchange at regulatory elements during chromatin remodeling underlies assisted loading mechanism. Cell 2011;146:544–54.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Cancer Research: 76 (19)
October 2016
Volume 76, Issue 19
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Androgen and Estrogen Receptors in Breast Cancer Coregulate Human UDP-Glucuronosyltransferases 2B15 and 2B17
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Androgen and Estrogen Receptors in Breast Cancer Coregulate Human UDP-Glucuronosyltransferases 2B15 and 2B17
Dong G. Hu, Luke A. Selth, Gerard A. Tarulli, Robyn Meech, Dhilushi Wijayakumara, Apichaya Chanawong, Roslin Russell, Carlos Caldas, Jessica L.L. Robinson, Jason S. Carroll, Wayne D. Tilley, Peter I. Mackenzie and Theresa E. Hickey
Cancer Res October 1 2016 (76) (19) 5881-5893; DOI: 10.1158/0008-5472.CAN-15-3372

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Androgen and Estrogen Receptors in Breast Cancer Coregulate Human UDP-Glucuronosyltransferases 2B15 and 2B17
Dong G. Hu, Luke A. Selth, Gerard A. Tarulli, Robyn Meech, Dhilushi Wijayakumara, Apichaya Chanawong, Roslin Russell, Carlos Caldas, Jessica L.L. Robinson, Jason S. Carroll, Wayne D. Tilley, Peter I. Mackenzie and Theresa E. Hickey
Cancer Res October 1 2016 (76) (19) 5881-5893; DOI: 10.1158/0008-5472.CAN-15-3372
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Authors' Contributions
    • Grant Support
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • mtDNA-Oncogene Cross-Talk
  • Alternative NFκB Regulates ALDH+ Ovarian Cancer Cells
  • JAM-C Identifies Leukemia-Initiating Cells in AML
Show more Tumor and Stem Cell Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement