21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      PRMT5 Is a Critical Regulator of Breast Cancer Stem Cell Function via Histone Methylation and FOXP1 Expression

      research-article

      Read this article at

      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.

          Summary

          Breast cancer progression, treatment resistance, and relapse are thought to originate from a small population of tumor cells, breast cancer stem cells (BCSCs). Identification of factors critical for BCSC function is therefore vital for the development of therapies. Here, we identify the arginine methyltransferase PRMT5 as a key in vitro and in vivo regulator of BCSC proliferation and self-renewal and establish FOXP1, a winged helix/forkhead transcription factor, as a critical effector of PRMT5-induced BCSC function. Mechanistically, PRMT5 recruitment to the FOXP1 promoter facilitates H3R2me2s, SET1 recruitment, H3K4me3, and gene expression. Our findings are clinically significant, as PRMT5 depletion within established tumor xenografts or treatment of patient-derived BCSCs with a pre-clinical PRMT5 inhibitor substantially reduces BCSC numbers. Together, our findings highlight the importance of PRMT5 in BCSC maintenance and suggest that small-molecule inhibitors of PRMT5 or downstream targets could be an effective strategy eliminating this cancer-causing population.

          Graphical Abstract

          Highlights

          • PRMT5 expression is elevated in breast cancer stem cells (BCSCs)

          • PRMT5 is required for BCSC function and self-renewal in vitro and in vivo

          • Targeting PRMT5 in an established tumor reduces stem cell numbers and tumor growth

          • FOXP1 promoter is methylated by PRMT5 and is an effector of PRMT5-driven BCSC function

          Abstract

          Chiang et al. show that the arginine methyltransferase PRMT5 contributes to breast cancer stem cell function, in part through histone methylation regulating FOXP1 expression. Targeting of PRMT5 through depletion or inhibition reduces stem cell frequency in vitro and in vivo, implicating PRMT5 as important in breast cancer pathogenesis.

          Related collections

          Most cited references32

          • 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: found
            Is Open Access

            Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy

            Introduction The phenotypic and functional differences between cells that initiate human breast tumors (cancer stem cells) and those that comprise the tumor bulk are difficult to study using only primary tumor tissue. We embarked on this study hypothesizing that breast cancer cell lines would contain analogous hierarchical differentiation programs to those found in primary breast tumors. Methods Eight human breast cell lines (human mammary epithelial cells, and MCF10A, MCF7, SUM149, SUM159, SUM1315 and MDA.MB.231 cells) were analyzed using flow cytometry for CD44, CD24, and epithelial-specific antigen (ESA) expression. Limiting dilution orthotopic injections were used to evaluate tumor initiation, while serial colony-forming unit, reconstitution and tumorsphere assays were performed to assess self-renewal and differentiation. Pulse-chase bromodeoxyuridine (5-bromo-2-deoxyuridine [BrdU]) labeling was used to examine cell cycle and label-retention of cancer stem cells. Cells were treated with paclitaxol and 5-fluorouracil to test selective resistance to chemotherapy, and gene expression profile after chemotherapy were examined. Results The percentage of CD44+/CD24- cells within cell lines does not correlate with tumorigenicity, but as few as 100 cells can form tumors when sorted for CD44+/CD24-/low/ESA+. Furthermore, CD44+/CD24-/ESA+ cells can self-renew, reconstitute the parental cell line, retain BrdU label, and preferentially survive chemotherapy. Conclusion These data validate the use of cancer cell lines as models for the development and testing of novel therapeutics aimed at eradicating cancer stem cells.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Epigenetic modulators, modifiers and mediators in cancer aetiology and progression.

              This year is the tenth anniversary of the publication in this journal of a model suggesting the existence of 'tumour progenitor genes'. These genes are epigenetically disrupted at the earliest stages of malignancies, even before mutations, and thus cause altered differentiation throughout tumour evolution. The past decade of discovery in cancer epigenetics has revealed a number of similarities between cancer genes and stem cell reprogramming genes, widespread mutations in epigenetic regulators, and the part played by chromatin structure in cellular plasticity in both development and cancer. In the light of these discoveries, we suggest here a framework for cancer epigenetics involving three types of genes: 'epigenetic mediators', corresponding to the tumour progenitor genes suggested earlier; 'epigenetic modifiers' of the mediators, which are frequently mutated in cancer; and 'epigenetic modulators' upstream of the modifiers, which are responsive to changes in the cellular environment and often linked to the nuclear architecture. We suggest that this classification is helpful in framing new diagnostic and therapeutic approaches to cancer.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Rep
                Cell Rep
                Cell Reports
                Cell Press
                2211-1247
                19 December 2017
                19 December 2017
                19 December 2017
                : 21
                : 12
                : 3498-3513
                Affiliations
                [1 ]Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
                [2 ]Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, and Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2GW, UK
                [3 ]Structural Genomics Consortium, Botnar Research Centre, NDORMS, University of Oxford, Oxford OX3 7LD, UK
                [4 ]Centre for Tumour Biology, Barts Cancer Institute, A Cancer Research UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, London EC1M 6BQ, UK
                [5 ]Cancer Epigenetics DPU, GlaxoSmithKline, Collegeville, PA 19426, USA
                [6 ]Institute of Molecular and Cell Biology (IMCB), A STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, 138673 Singapore, Singapore
                [7 ]Department of Oncological Sciences and Pharmacological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
                [8 ]European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
                Author notes
                []Corresponding author c.c.davies@ 123456bham.ac.uk
                [9]

                Deceased

                [10]

                Lead Contact

                Article
                S2211-1247(17)31763-1
                10.1016/j.celrep.2017.11.096
                5746596
                29262329
                b198ba98-614c-4913-ba11-3d8b352afd30
                © 2017 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 8 June 2017
                : 1 November 2017
                : 28 November 2017
                Categories
                Article

                Cell biology
                arginine methylation,prmt5,breast cancer stem cell,histone methylation,h3r2me2s,foxp1,epigenetics,self-renewal,breast cancer,drug resistance

                Comments

                Comment on this article