29
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Progesterone receptor-B enhances estrogen responsiveness of breast cancer cells via scaffolding PELP1- and estrogen receptor-containing transcription complexes

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Progesterone and estrogen are important drivers of breast cancer proliferation. Herein, we probed ER-alpha and PR cross-talk in breast cancer models. Stable expression of PR-B in PR-low/ER+ MCF7 cells increased cellular sensitivity to estradiol and IGF1, as measured in growth assays performed in the absence of exogenous progestin; similar results were obtained in PR-null/ER+ T47D cells stably expressing PR-B. Genome-wide microarray analyses revealed that unliganded PR-B induced robust expression of a subset of estradiol-responsive ER-target genes, including CathepsinD ( CTSD). Estradiol-treated MCF7 cells stably expressing PR-B exhibited enhanced ER Ser167 phosphorylation and recruitment of ER, PR, and the proline, glutamate and leucine rich protein 1 (PELP1) to an estrogen response element (ERE) in the CTSD distal promoter; this complex co-immunoprecipitated with IGF1R in whole cell lysates. Importantly, ER/PR/PELP1 complexes were also detected in human breast cancer samples. Inhibition of IGF1R or PI3K blocked PR-B-dependent CTSD mRNA upregulation in response to estradiol. Similarly, inhibition of IGF1R or PR significantly reduced ER recruitment to the CTSD promoter. Stable knockdown of endogenous PR or onapristone treatment of multiple unmodified breast cancer cell lines blocked estradiol-mediated CTSD induction, inhibited growth in soft agar, and partially restored tamoxifen-sensitivity of resistant cells. Further, combination treatment of breast cancer cells with both onapristone and IGF1R tyrosine kinase inhibitor AEW541 was more effective than either agent alone. In summary, unliganded PR-B enhanced proliferative responses to estradiol and IGF1 via scaffolding of ERalpha/PELP1/IGF1R-containing complexes. Our data provide a strong rationale for targeting PR in combination with ER and IGF1R in patients with luminal breast cancer.

          Related collections

          Most cited references44

          • Record: found
          • Abstract: found
          • Article: not found

          Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal.

          The cBioPortal for Cancer Genomics (http://cbioportal.org) provides a Web resource for exploring, visualizing, and analyzing multidimensional cancer genomics data. The portal reduces molecular profiling data from cancer tissues and cell lines into readily understandable genetic, epigenetic, gene expression, and proteomic events. The query interface combined with customized data storage enables researchers to interactively explore genetic alterations across samples, genes, and pathways and, when available in the underlying data, to link these to clinical outcomes. The portal provides graphical summaries of gene-level data from multiple platforms, network visualization and analysis, survival analysis, patient-centric queries, and software programmatic access. The intuitive Web interface of the portal makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries. Here, we provide a practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Biological determinants of endocrine resistance in breast cancer.

            Endocrine therapies targeting oestrogen action (anti-oestrogens, such as tamoxifen, and aromatase inhibitors) decrease mortality from breast cancer, but their efficacy is limited by intrinsic and acquired therapeutic resistance. Candidate molecular biomarkers and gene expression signatures of tamoxifen response emphasize the importance of deregulation of proliferation and survival signalling in endocrine resistance. However, definition of the specific genetic lesions and molecular processes that determine clinical endocrine resistance is incomplete. The development of large-scale computational and genetic approaches offers the promise of identifying the mediators of endocrine resistance that may be exploited as potential therapeutic targets and biomarkers of response in the clinic.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription.

              Many cofactors bind the hormone-activated estrogen receptor (ER), yet the specific regulators of endogenous ER-mediated gene transcription are unknown. Using chromatin immunoprecipitation (ChIP), we find that ER and a number of coactivators rapidly associate with estrogen responsive promoters following estrogen treatment in a cyclic fashion that is not predicted by current models of hormone activation. Cycles of ER complex assembly are followed by transcription. In contrast, the anti-estrogen tamoxifen (TAM) recruits corepressors but not coactivators. Using a genetic approach, we show that recruitment of the p160 class of coactivators is sufficient for gene activation and for the growth stimulatory actions of estrogen in breast cancer supporting a model in which ER cofactors play unique roles in estrogen signaling.
                Bookmark

                Author and article information

                Journal
                8711562
                6325
                Oncogene
                Oncogene
                Oncogene
                0950-9232
                1476-5594
                4 February 2014
                27 January 2014
                22 January 2015
                22 July 2015
                : 34
                : 4
                : 506-515
                Affiliations
                [1 ]Departments of Medicine and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
                [2 ]Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
                [3 ]Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
                [4 ]Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
                [5 ]Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
                Author notes
                [* ]To whom correspondence and reprint requests should be addressed: Carol A. Lange, PhD., University of Minnesota Masonic Cancer Center, 2812 Cancer Cardiology Research Building, 2231 6 th St. SE, Minneapolis, MN 55455, Lange047@ 123456umn.edu , 612-626-0621 (phone), 612-624-3869 (fax)
                Article
                NIHMS550292
                10.1038/onc.2013.579
                4112172
                24469035
                0d8943b2-7bd5-442c-a861-a86f755ae6b1

                Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Categories
                Article

                Oncology & Radiotherapy
                estrogen receptor,progesterone receptor,pelp1,igf1r,cathepsin d,breast cancer
                Oncology & Radiotherapy
                estrogen receptor, progesterone receptor, pelp1, igf1r, cathepsin d, breast cancer

                Comments

                Comment on this article