21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      p53 and MicroRNA-34 Are Suppressors of Canonical Wnt Signaling

      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

          The tumor suppressor p53 activates miRNA-34 to inhibit Wnt signaling and colorectal cancer cell invasiveness.

          p53 Activates MicroRNA-34 to Inhibit Wnt Signaling

          The tumor suppressor p53 is missing or nonfunctional in many cancers, whereas the canonical Wnt signaling pathway is frequently activated. Here, Kim et al . show that p53 restrained Wnt signaling during Xenopus development, whereas loss of p53 function led to aberrant activation of the canonical Wnt signaling pathway, with microRNA-34 (miR-34) providing the connection between the two. They found that p53 stimulated production of miR-34, which, in turn, targeted key genes in the Wnt signaling pathway. Analyses of gene expression data sets indicated that loss of p53 or miR-34 function was associated with activation of Wnt signaling in human cancers; moreover, loss of p53 function increased Wnt signaling in colon cancer cells in vitro. In p53-mutant colon cancer cells, miR-34 attenuated Wnt signaling and decreased the invasiveness of these cells in vitro. Thus, the p53–miR-34–Wnt pathway appears to be crucial not only during development but also for p53’s tumor suppressor function.

          Abstract

          Although loss of p53 function and activation of canonical Wnt signaling cascades are frequently coupled in cancer, the links between these two pathways remain unclear. We report that p53 transactivated microRNA-34 (miR-34), which consequently suppressed the transcriptional activity of β-catenin–T cell factor and lymphoid enhancer factor (TCF/LEF) complexes by targeting the untranslated regions (UTRs) of a set of conserved targets in a network of genes encoding elements of the Wnt pathway. Loss of p53 function increased canonical Wnt signaling by alleviating miR-34–specific interactions with target UTRs, and miR-34 depletion relieved p53-mediated Wnt repression. Gene expression signatures reflecting the status of β-catenin–TCF/LEF transcriptional activity in breast cancer and pediatric neuroblastoma patients were correlated with p53 and miR-34 functional status. Loss of p53 or miR-34 contributed to neoplastic progression by triggering the Wnt-dependent, tissue-invasive activity of colorectal cancer cells. Further, during development, miR-34 interactions with the β-catenin UTR affected Xenopus body axis polarity and the expression of Wnt-dependent patterning genes. These data provide insight into the mechanisms by which a p53–miR-34 network restrains canonical Wnt signaling cascades in developing organisms and human cancer.

          Related collections

          Most cited references69

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

          MicroRNAs

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

            MicroRNA biogenesis: coordinated cropping and dicing.

            V Kim (2005)
            The recent discovery of microRNAs (miRNAs) took many by surprise because of their unorthodox features and widespread functions. These tiny, approximately 22-nucleotide, RNAs control several pathways including developmental timing, haematopoiesis, organogenesis, apoptosis, cell proliferation and possibly even tumorigenesis. Among the most pressing questions regarding this unusual class of regulatory miRNA-encoding genes is how miRNAs are produced in cells and how the genes themselves are controlled by various regulatory networks.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours.

              Mutations in the p53 tumour-suppressor gene are the most frequently observed genetic lesions in human cancers. To investigate the role of the p53 gene in mammalian development and tumorigenesis, a null mutation was introduced into the gene by homologous recombination in murine embryonic stem cells. Mice homozygous for the null allele appear normal but are prone to the spontaneous development of a variety of neoplasms by 6 months of age. These observations indicate that a normal p53 gene is dispensable for embryonic development, that its absence predisposes the animal to neoplastic disease, and that an oncogenic mutant form of p53 is not obligatory for the genesis of many types of tumours.
                Bookmark

                Author and article information

                Journal
                Science Signaling
                Sci. Signal.
                American Association for the Advancement of Science (AAAS)
                1945-0877
                1937-9145
                November 2011
                November 2011
                : 4
                : 197
                Affiliations
                [1 ]Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul 120-752, Korea.
                [2 ]Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA.
                [3 ]miRcore, 2929 Plymouth Road, Ann Arbor, MI 48105, USA.
                [4 ]Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea.
                [5 ]Division of Molecular Medicine and Genetics, Department of Internal Medicine and Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
                [6 ]Department of Biochemistry and Molecular Biology, Brain Korea 21 Project for Medical Science of Yonsei University, Center for Chronic Metabolic Disease Research, School of Medicine, Yonsei University, Seoul 120-752, Korea.
                [7 ]Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea.
                Article
                10.1126/scisignal.2001744
                3447368
                22045851
                b5477166-febc-4fde-9ae2-52be9d3aedf5
                © 2011
                History

                Comments

                Comment on this article