14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Fisetin decreases TET1 activity and CCNY/CDK16 promoter 5hmC levels to inhibit the proliferation and invasion of renal cancer stem cell

      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

          As a natural flavonol, fisetin has significant inhibitory effects on many cancers. Although fisetin can inhibit kidney cancer, its effects on kidney renal stem cells (Hu RCSCs) remain unknown. Our study found that renal cancer tissues and CD44+/ CD105+ Hu RCSCs both show high TET1 protein expression. Both in vivo and in vitro experiments showed that fisetin can effectively inhibit Hu RCSC cell division and proliferation, invasion, in vivo tumourigenesis and angiogenesis. Our findings showed that fisetin can significantly decrease TET1 expression levels in Hu RCSCs and overall 5hmC levels in the genomes of these cells. At the same time, Ch IPPCR results showed that fisetin can effectively inhibit 5hmC modification levels at the CpG islands in cyclin Y ( CCNY) and CDK16 and reduce their transcription and activity. Thus, we conclude that fisetin inhibits the epigenetic mechanism in renal cancer stem cells, that is, fisetin inhibits TET1 expression and reduces 5hmC modification in specific loci in the promoters of CCNY/ CDK16 in Hu RSCs. This in turn inhibits transcription of these genes, causing cell cycle arrest and ultimately inhibiting renal cancer stem cell activity.

          Related collections

          Most cited references18

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

          Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain.

          Cytosine methylation is the major covalent modification of mammalian genomic DNA and plays important roles in transcriptional regulation. The molecular mechanism underlying the enzymatic removal of this epigenetic mark, however, remains elusive. Here, we show that 5-methylcytosine (5mC) hydroxylase TET1, by converting 5mCs to 5-hydroxymethylcytosines (5hmCs), promotes DNA demethylation in mammalian cells through a process that requires the base excision repair pathway. Though expression of the 12 known human DNA glycosylases individually did not enhance removal of 5hmCs in mammalian cells, demethylation of both exogenously introduced and endogenous 5hmCs is promoted by the AID (activation-induced deaminase)/APOBEC (apolipoprotein B mRNA-editing enzyme complex) family of cytidine deaminases. Furthermore, Tet1 and Apobec1 are involved in neuronal activity-induced, region-specific, active DNA demethylation and subsequent gene expression in the dentate gyrus of the adult mouse brain in vivo. Our study suggests a TET1-induced oxidation-deamination mechanism for active DNA demethylation in mammals. Copyright © 2011 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Active DNA demethylation: many roads lead to Rome.

            DNA methylation is one of the best-characterized epigenetic modifications and has been implicated in numerous biological processes, including transposable element silencing, genomic imprinting and X chromosome inactivation. Compared with other epigenetic modifications, DNA methylation is thought to be relatively stable. Despite its role in long-term silencing, DNA methylation is more dynamic than originally thought as active DNA demethylation has been observed during specific stages of development. In the past decade, many enzymes have been proposed to carry out active DNA demethylation and growing evidence suggests that, depending on the context, this process may be achieved by multiple mechanisms. Insight into how DNA methylation is dynamically regulated will broaden our understanding of epigenetic regulation and have great implications in somatic cell reprogramming and regenerative medicine.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              MicroRNA-199a targets CD44 to suppress the tumorigenicity and multidrug resistance of ovarian cancer-initiating cells.

              In ovarian cancer, CD44(+) /CD117(+) stem cells, also known as cancer-initiating cells (CICs), are highly proliferative, have a low degree of differentiation, and are resistant to chemotherapeutics. Therefore, the CD44(+) /CD117(+) subpopulation is thought to be an important target for novel therapeutic strategies. In this study, we investigated the role of microRNA-199a (miR-199a) in ovarian cancer stem cells. Luciferase reporter gene assays confirmed that miR-199a targets CD44 via an miR-199a-binding site in the 3'-UTR. CD44(+) /CD117(+) ovarian CICs were enriched from human primary ovarian tumor tissues and confirmed by flow cytometric sorting. miR-199a was cloned and transfected into ovarian CICs. CD44 mRNA and protein expression was significantly decreased in miR-199a-transfected ovarian CICs as compared with miR-199a mutant-transfected and untransfected cells. Cell cycle analysis, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide proliferation assays, the colony formation assay and the transwell migration assay indicated that miR-199a significantly affected cell cycle regulation and suppressed the proliferation and invasive capacity of ovarian CICs in vitro. miR-199a significantly increased the chemosensitivity of ovarian CICs to cisplatin, pacitaxel, and adriamycin, and reduced mRNA expression of the multidrug resistance gene ABCG2 as compared with miR-199a mutant-transfected and untransfected cells. The expression of stemness markers was also significantly reduced in miR-199a-transfected CICs as compared with miR-199a mutant-transfected and untransfected ovarian cells. Furthermore, xenograft experiments confirmed that miR-199a suppressed the growth of xenograft tumors formed by ovarian CICs in vivo. Thus, expression of endogenous mature miR-199a may prevent tumorigenesis in human ovarian cancer by regulating expression of its target gene CD44. © 2012 The Authors Journal compilation © 2012 FEBS.
                Bookmark

                Author and article information

                Contributors
                xuejun@fudan.edu.cn
                te.liu@yale.edu
                Journal
                J Cell Mol Med
                J. Cell. Mol. Med
                10.1111/(ISSN)1582-4934
                JCMM
                Journal of Cellular and Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1582-1838
                1582-4934
                08 November 2018
                February 2019
                : 23
                : 2 ( doiID: 10.1111/jcmm.2019.23.issue-2 )
                : 1095-1105
                Affiliations
                [ 1 ] Division of Nephrology Huashan Hospital Fudan University Shanghai China
                [ 2 ] Nursing Department Huashan Hospital Fudan University Shanghai China
                [ 3 ] Department of Urology Beijing Friendship Hospital Capital Medical University Beijing China
                [ 4 ] Shanghai Topbiox Co. Ltd Shanghai China
                [ 5 ] Shanghai Geriatric Institute of Chinese Medicine Shanghai University of Traditional Chinese Medicine Shanghai China
                [ 6 ] Department of Pathology Yale University School of Medicine New Haven Connecticut
                [ 7 ] Department of Biochemistry College of Basic Medicine Shanghai University of Traditional Chinese Medicine Shanghai China
                Author notes
                [*] [* ] Correspondence

                Jun Xue, Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China.

                Email: xuejun@ 123456fudan.edu.cn

                Te Liu, Department of Pathology, Yale University School of Medicine, New Haven, CT.

                Email: te.liu@ 123456yale.edu

                [†]

                These authors contributed equally to this work and shared the first authorship.

                Author information
                http://orcid.org/0000-0003-1514-9548
                Article
                JCMM14010
                10.1111/jcmm.14010
                6349178
                30411496
                6fcc49e2-18ba-4d34-bd74-2f87f8b79558
                © 2018 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 07 August 2018
                : 12 October 2018
                Page count
                Figures: 6, Tables: 0, Pages: 11, Words: 5999
                Funding
                Funded by: Natural Science Foundation of Shanghai
                Award ID: 16ZR1434000
                Funded by: Natural Science Foundation of China
                Award ID: 81670697
                Award ID: 81774019
                Funded by: Shanghai Natural Science Foundation of China
                Award ID: 16ZR1437400
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                jcmm14010
                February 2019
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.5.6 mode:remove_FC converted:28.01.2019

                Molecular medicine
                5‐hydroxymethylation,cell cyclin‐dependent kinases,fisetin,renal cancer stem cells

                Comments

                Comment on this article