5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Cinobufagin Suppresses Melanoma Cell Growth by Inhibiting LEF1

      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.

          Abstract

          Constitutive activation of the β-catenin dependent canonical Wnt signaling pathway, which enhances tumor growth and progression in multiple types of cancer, is commonly observed in melanoma. LEF1 activates β-catenin/TCF4 transcriptional activity, promoting tumor growth and progression. Although several reports have shown that LEF1 is highly expressed in melanoma, the functional role of LEF1 in melanoma growth is not fully understood. While A375, A2058, and G361 melanoma cells exhibit abnormally high LEF1 expression, lung cancer cells express lower LEF1 levels. A luciferase assay-based high throughput screening (HTS) with a natural compound library showed that cinobufagin suppressed β-catenin/TCF4 transcriptional activity by inhibiting LEF1 expression. Cinobufagin decreases LEF1 expression in a dose-dependent manner and Wnt/β-catenin target genes such as Axin-2, cyclin D1, and c-Myc in melanoma cell lines. Cinobufagin sensitively attenuates cell viability and induces apoptosis in LEF1 expressing melanoma cells compared to LEF1-low expressing lung cancer cells. In addition, ectopic LEF1 expression is sufficient to attenuate cinobufagin-induced apoptosis and cell growth retardation in melanoma cells. Thus, we suggest that cinobufagin is a potential anti-melanoma drug that suppresses tumor-promoting Wnt/β-catenin signaling via LEF1 inhibition.

          Related collections

          Most cited references44

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

          Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma.

          The adenomatous polyposis coli (APC) tumor suppressor protein binds to beta-catenin, a protein recently shown to interact with Tcf and Lef transcription factors. The gene encoding hTcf-4, a Tcf family member that is expressed in colonic epithelium, was cloned and characterized. hTcf-4 transactivates transcription only when associated with beta-catenin. Nuclei of APC-/- colon carcinoma cells were found to contain a stable beta-catenin-hTcf-4 complex that was constitutively active, as measured by transcription of a Tcf reporter gene. Reintroduction of APC removed beta-catenin from hTcf-4 and abrogated the transcriptional transactivation. Constitutive transcription of Tcf target genes, caused by loss of APC function, may be a crucial event in the early transformation of colonic epithelium.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis.

            Metastasis from lung adenocarcinoma can occur swiftly to multiple organs within months of diagnosis. The mechanisms that confer this rapid metastatic capacity to lung tumors are unknown. Activation of the canonical WNT/TCF pathway is identified here as a determinant of metastasis to brain and bone during lung adenocarcinoma progression. Gene expression signatures denoting WNT/TCF activation are associated with relapse to multiple organs in primary lung adenocarcinoma. Metastatic subpopulations isolated from independent lymph node-derived lung adenocarcinoma cell lines harbor a hyperactive WNT/TCF pathway. Reduction of TCF activity in these cells attenuates their ability to form brain and bone metastases in mice, independently of effects on tumor growth in the lungs. The WNT/TCF target genes HOXB9 and LEF1 are identified as mediators of chemotactic invasion and colony outgrowth. Thus, a distinct WNT/TCF signaling program through LEF1 and HOXB9 enhances the competence of lung adenocarcinoma cells to colonize the bones and the brain. For a video summary of this article, see the PaperFlick file available with the online Supplemental Data.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Dysregulation of Wnt/β-catenin signaling in gastrointestinal cancers.

              Aberrant Wnt/β-catenin signaling is widely implicated in numerous malignancies, including cancers of the gastrointestinal tract. Dysregulation of signaling is traditionally attributed to mutations in Axin, adenomatous polyposis coli, and β-catenin that lead to constitutive hyperactivation of the pathway. However, Wnt/β-catenin signaling is also modulated through various other mechanisms in cancer, including cross talk with other altered signaling pathways. A more complex view of Wnt/β-catenin signaling and its role in gastrointestinal cancers is now emerging as divergent phenotypic outcomes are found to be dictated by temporospatial context and relative levels of pathway activation. This review summarizes the dysregulation of Wnt/β-catenin signaling in colorectal carcinoma, hepatocellular carcinoma, and pancreatic ductal adenocarcinoma, with particular emphasis on the latter two. We conclude by addressing some of the major challenges faced in attempting to target the pathway in the clinic. Copyright © 2012 AGA Institute. Published by Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Journal
                Int J Mol Sci
                Int J Mol Sci
                ijms
                International Journal of Molecular Sciences
                MDPI
                1422-0067
                13 September 2020
                September 2020
                : 21
                : 18
                : 6706
                Affiliations
                [1 ]Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Korea; rlarjsgml4@ 123456kku.ac.kr (G.-H.K.); gkrrnjs654852@ 123456kku.ac.kr (X.-Q.F.); lwj0908@ 123456kku.ac.kr (W.-J.L.); pkjhdn@ 123456kku.ac.kr (J.P.)
                [2 ]Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Korea
                [3 ]Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Korea; kangtbko@ 123456kku.ac.kr
                [4 ]Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
                Author notes
                [* ]Correspondence: jay_kim@ 123456korea.ac.kr (J.H.K.); jhlim@ 123456kku.ac.kr (J.-H.L.)
                Author information
                https://orcid.org/0000-0003-1441-9470
                Article
                ijms-21-06706
                10.3390/ijms21186706
                7554883
                32933177
                65495702-e9e7-492d-91cd-8f8317c8f959
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 11 August 2020
                : 11 September 2020
                Categories
                Article

                Molecular biology
                lef1,tcf4,cinobufagin,melanoma
                Molecular biology
                lef1, tcf4, cinobufagin, melanoma

                Comments

                Comment on this article