2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Tumor-suppressor function of Beclin 1 in breast cancer cells requires E-cadherin

      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.

          Significance

          Beclin 1, an essential autophagy protein, is important for tumor suppression in mice, as well as in human breast and ovarian cancers. However, it is not well understood how Beclin 1 acts as a tumor suppressor. By performing a genetic screen to identify genes whose loss blocks the ability of Beclin 1 to inhibit the growth of breast cancer cells and follow-up biological analyses, we have identified a mechanism by which Beclin 1 prevents breast cancer growth. We found that Beclin 1 promotes the plasma membrane localization of E-cadherin, a breast tumor-suppressor molecule that restricts tumor growth and metastases only when present at the cell surface. These findings have important implications for understanding the cell biology of human breast cancer.

          Abstract

          Beclin 1, an autophagy and haploinsufficient tumor-suppressor protein, is frequently monoallelically deleted in breast and ovarian cancers. However, the precise mechanisms by which Beclin 1 inhibits tumor growth remain largely unknown. To address this question, we performed a genome-wide CRISPR/Cas9 screen in MCF7 breast cancer cells to identify genes whose loss of function reverse Beclin 1-dependent inhibition of cellular proliferation. Small guide RNAs targeting CDH1 and CTNNA1, tumor-suppressor genes that encode cadherin/catenin complex members E-cadherin and alpha-catenin, respectively, were highly enriched in the screen. CRISPR/Cas9-mediated knockout of CDH1 or CTNNA1 reversed Beclin 1-dependent suppression of breast cancer cell proliferation and anchorage-independent growth. Moreover, deletion of CDH1 or CTNNA1 inhibited the tumor-suppressor effects of Beclin 1 in breast cancer xenografts. Enforced Beclin 1 expression in MCF7 cells and tumor xenografts increased cell surface localization of E-cadherin and decreased expression of mesenchymal markers and beta-catenin/Wnt target genes. Furthermore, CRISPR/Cas9-mediated knockout of BECN1 and the autophagy class III phosphatidylinositol kinase complex 2 (PI3KC3-C2) gene, UVRAG, but not PI3KC3-C1–specific ATG14 or other autophagy genes ATG13, ATG5, or ATG7, resulted in decreased E-cadherin plasma membrane and increased cytoplasmic E-cadherin localization. Taken together, these data reveal previously unrecognized cooperation between Beclin 1 and E-cadherin–mediated tumor suppression in breast cancer cells.

          Related collections

          Most cited references48

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

          Improved vectors and genome-wide libraries for CRISPR screening.

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

            The basics of epithelial-mesenchymal transition.

            The origins of the mesenchymal cells participating in tissue repair and pathological processes, notably tissue fibrosis, tumor invasiveness, and metastasis, are poorly understood. However, emerging evidence suggests that epithelial-mesenchymal transitions (EMTs) represent one important source of these cells. As we discuss here, processes similar to the EMTs associated with embryo implantation, embryogenesis, and organ development are appropriated and subverted by chronically inflamed tissues and neoplasias. The identification of the signaling pathways that lead to activation of EMT programs during these disease processes is providing new insights into the plasticity of cellular phenotypes and possible therapeutic interventions.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9

              CRISPR-Cas9-based genetic screens are a powerful new tool in biology. By simply altering the sequence of the single-guide RNA (sgRNA), Cas9 can be reprogrammed to target different sites in the genome with relative ease, but the on-target activity and off-target effects of individual sgRNAs can vary widely. Here, we use recently-devised sgRNA design rules to create human and mouse genome-wide libraries, perform positive and negative selection screens and observe that the use of these rules produced improved results. Additionally, we profile the off-target activity of thousands of sgRNAs and develop a metric to predict off-target sites. We incorporate these findings from large-scale, empirical data to improve our computational design rules and create optimized sgRNA libraries that maximize on-target activity and minimize off-target effects to enable more effective and efficient genetic screens and genome engineering.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc Natl Acad Sci U S A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                02 February 2021
                25 January 2021
                25 January 2021
                : 118
                : 5
                : e2020478118
                Affiliations
                [1] aCenter for Autophagy Research, University of Texas Southwestern Medical Center , Dallas, TX 75390;
                [2] bDepartment of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, TX 75390;
                [3] cHoward Hughes Medical Institute , University of Texas Southwestern Medical Center , Dallas, TX 75390;
                [4] dQuantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center , Dallas, TX 75390;
                [5] eBroad Institute of MIT and Harvard , Cambridge, MA 02142;
                [6] fDepartment of Microbiology, University of Texas Southwestern Medical Center , Dallas, TX 75390
                Author notes
                1To whom correspondence may be addressed. Email: lynda.bennett@ 123456utsouthwestern.edu .

                Edited by Tak W. Mak, University of Toronto, Toronto, Canada, and approved November 11, 2020 (received for review October 7, 2020)

                Author contributions: T.W., L.B., and B.L. designed research; T.W., Z.Z., and Y.X. performed research; Y.X. and J.G.D. contributed new reagents/analytic tools; T.W., Z.Z., B.C., L.Z., G.X., L.B., and B.L. analyzed data; and T.W., L.B., and B.L. wrote the paper.

                Author information
                https://orcid.org/0000-0003-1784-2100
                https://orcid.org/0000-0003-3937-5902
                https://orcid.org/0000-0002-3707-9889
                Article
                202020478
                10.1073/pnas.2020478118
                7865132
                33495338
                52209af6-75d1-4d0a-bc0a-5bdb32983ff2
                Copyright © 2021 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 9
                Categories
                409
                Biological Sciences
                Cell Biology

                beclin 1,e-cadherin,breast cancer
                beclin 1, e-cadherin, breast cancer

                Comments

                Comment on this article