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

      Cross-talk between mutant p53 and p62/SQSTM1 augments cancer cell migration by promoting the degradation of cell adhesion proteins

      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

          Missense mutations in the TP53 gene, encoding the p53 tumor suppressor, are very frequent in human cancer. Some of those mutations, particularly the more common (“hotspot”) ones, not only abrogate p53’s tumor suppressor activities but also endow the mutant protein with oncogenic gain of function (GOF). We report that p53 R273H, the most common p53 mutant in pancreatic cancer, interacts with the SQSTM1/p62 protein to accelerate the degradation of cell adhesion proteins. This enables pancreatic cancer cells to detach from the epithelial sheet and engage in individualized cell migration, probably augmenting metastatic spread. By providing insights into mechanisms that underpin mutant p53 GOF, this study may suggest ways to interfere with the progression of cancers bearing particular p53 mutants.

          Abstract

          Missense mutations in the p53 tumor suppressor abound in human cancer. Common (“hotspot”) mutations endow mutant p53 (mutp53) proteins with oncogenic gain of function (GOF), including enhanced cell migration and invasiveness, favoring cancer progression. GOF is usually attributed to transcriptional effects of mutp53. To elucidate transcription-independent effects of mutp53, we characterized the protein interactome of the p53 R273H mutant in cells derived from pancreatic ductal adenocarcinoma (PDAC), where p53 R273H is the most frequent p53 mutant. We now report that p53 R273H, but not the p53 R175H hotspot mutant, interacts with SQSTM1/p62 and promotes cancer cell migration and invasion in a p62-dependent manner. Mechanistically, the p53 R273H-p62 axis drives the proteasomal degradation of several cell junction–associated proteins, including the gap junction protein Connexin 43, facilitating scattered cell migration. Concordantly, down-regulation of Connexin 43 augments PDAC cell migration, while its forced overexpression blunts the promigratory effect of the p53 R273H-p62 axis. These findings define a mechanism of mutp53 GOF.

          Related collections

          Most cited references83

          • Record: found
          • Abstract: found
          • Article: found

          Hallmarks of Cancer: The Next Generation

          The hallmarks of cancer comprise six biological capabilities acquired during the multistep development of human tumors. The hallmarks constitute an organizing principle for rationalizing the complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. Underlying these hallmarks are genome instability, which generates the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hallmark functions. Conceptual progress in the last decade has added two emerging hallmarks of potential generality to this list-reprogramming of energy metabolism and evading immune destruction. In addition to cancer cells, tumors exhibit another dimension of complexity: they contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hallmark traits by creating the "tumor microenvironment." Recognition of the widespread applicability of these concepts will increasingly affect the development of new means to treat human cancer. Copyright © 2011 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Mutant p53 in Cancer: New Functions and Therapeutic Opportunities

            Many different types of cancer show a high incidence of TP53 mutations, leading to the expression of mutant p53 proteins. There is growing evidence that these mutant p53s have both lost wild-type p53 tumor suppressor activity and gained functions that help to contribute to malignant progression. Understanding the functions of mutant p53 will help in the development of new therapeutic approaches that may be useful in a broad range of cancer types.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mutant p53: one name, many proteins.

              There is now strong evidence that mutation not only abrogates p53 tumor-suppressive functions, but in some instances can also endow mutant proteins with novel activities. Such neomorphic p53 proteins are capable of dramatically altering tumor cell behavior, primarily through their interactions with other cellular proteins and regulation of cancer cell transcriptional programs. Different missense mutations in p53 may confer unique activities and thereby offer insight into the mutagenic events that drive tumor progression. Here we review mechanisms by which mutant p53 exerts its cellular effects, with a particular focus on the burgeoning mutant p53 transcriptome, and discuss the biological and clinical consequences of mutant p53 gain of function.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc Natl Acad Sci U S A
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                19 April 2022
                26 April 2022
                19 October 2022
                : 119
                : 17
                : e2119644119
                Affiliations
                [1] aDepartment of Molecular Cell Biology, Weizmann Institute of Science , 7610001 Rehovot, Israel;
                [2] bCentre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital , Toronto, ON M5S 1A8, Canada;
                [3] cDepartment of Molecular Genetics, University of Toronto , Toronto, ON M5S 1A8, Canada;
                [4] dDepartment of Biological Regulation, Weizmann Institute of Science , 7610001 Rehovot, Israel;
                [5] eDepartment of Biomolecular Science, Weizmann Institute of Science , 7610001 Rehovot, Israel;
                [6] fTechnion Integrated Cancer Center, The Rappaport Faculty of Medicine and Research Institute, Technion–Israel Institute of Technology , 3109601 Haifa, Israel;
                [7] gSmoler Proteomic Center, Faculty of Biology, Technion–Israel Institute of Technology , 3200003 Haifa, Israel;
                [8] hAdvanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre , 410210 Kharghar, Navi Mumbai, India
                Author notes
                2To whom correspondence may be addressed. Email: moshe.oren@ 123456weizmann.ac.il .

                Contributed by Moshe Oren; received October 27, 2021; accepted March 11, 2022; reviewed by Carol Prives and Arnold Levine

                Author contributions: S. Mukherjee and M.O. designed research; S. Mukherjee, M.M., Y.L., S. Martinez, N.B.N., A.N., S.S., V.C.-K., T.Z., S.A., O.H., R.C., A.-C.P., and A.G. performed research; Y.A., Z.E., Y.Y., and D.S. supervised research; S. Mukherjee, Y.L., V.C.-K., Y.A., Z.E., Y.Y., and D.S. contributed new reagents/analytic tools; S. Martinez, K.T., T.Z., and M.S.-S. analyzed data; and S. Mukherjee and M.O. wrote the paper.

                1M.M. and Y.L. contributed equally to this work.

                Author information
                https://orcid.org/0000-0003-2970-6739
                https://orcid.org/0000-0001-7535-4143
                https://orcid.org/0000-0003-4168-7884
                https://orcid.org/0000-0003-4311-7172
                Article
                202119644
                10.1073/pnas.2119644119
                9173583
                35439056
                9750fd6a-4c2a-49b7-918c-b1fc076d1a11
                Copyright © 2022 the Author(s). Published by PNAS.

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

                History
                : 11 March 2022
                Page count
                Pages: 12
                Funding
                Funded by: Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (AMRF) 100005984
                Award ID: 713588
                Award Recipient : Moshe Oren
                Funded by: Israel Science Foundation (ISF) 501100003977
                Award ID: 2609/17
                Award Recipient : Moshe Oren
                Categories
                409
                Biological Sciences
                Cell Biology

                mutant p53,p62,protein–protein interaction,migration,cell adhesion

                Comments

                Comment on this article