27
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      lncRNA SNHG6 regulates EZH2 expression by sponging miR-26a/b and miR-214 in colorectal cancer

      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

          Background

          Abnormal expression of long non-coding RNAs (lncRNAs) has been found in almost all human tumors, providing numerous potential diagnostic biomarkers, prognostic biomarkers, and therapeutic targets.

          Methods

          We analyzed RNA sequencing data to explore abnormally expressed lncRNAs in colorectal cancer (CRC). The functions of small nucleolar RNA host gene 6 (SNHG6) were investigated through in vitro and in vivo assays (CCK-8 assay, colony formation assay, flow cytometry assay, EdU assay, wound healing assay, transwell assay, and xenograft model). The mechanism of action of SNHG6 was explored through bioinformatics, RNA fluorescence in situ hybridization, luciferase reporter assay, RNA pull-down assay, chromatin immunoprecipitation assay, and RNA immunoprecipitation assay.

          Results

          We identified aberrantly expressed lncRNAs in CRC. We found that elevated SNHG6 expression was associated with poor prognosis and CRC progression. We also demonstrated that the high SNHG6 expression was partly due to DNA copy number gains and SP1 induction. Functional studies showed that SNHG6 promoted CRC cell growth, migration, and invasion both in vitro and in vivo . Mechanistically, we found that SNHG6 expressed predominantly in the cytoplasm. SNHG6 could interact with miR-26a, miR-26b, and miR-214 and regulate their common target EZH2.

          Conclusions

          Our study elucidated that SNHG6 acted as an oncogene in CRC, which might serve as a novel target for CRC diagnosis and therapy.

          Electronic supplementary material

          The online version of this article (10.1186/s13045-018-0690-5) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references24

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

          Targeting EZH2 in cancer.

          Recent genomic studies have resulted in an emerging understanding of the role of chromatin regulators in the development of cancer. EZH2, a histone methyl transferase subunit of a Polycomb repressor complex, is recurrently mutated in several forms of cancer and is highly expressed in numerous others. Notably, both gain-of-function and loss-of-function mutations occur in cancers but are associated with distinct cancer types. Here we review the spectrum of EZH2-associated mutations, discuss the mechanisms underlying EZH2 function, and synthesize a unifying perspective that the promotion of cancer arises from disruption of the role of EZH2 as a master regulator of transcription. We further discuss EZH2 inhibitors that are now showing early signs of promise in clinical trials and also additional strategies to combat roles of EZH2 in cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Roles of the EZH2 histone methyltransferase in cancer epigenetics.

            EZH2 is the catalytic subunit of Polycomb repressive complex 2 (PRC2), which is a highly conserved histone methyltransferase that targets lysine-27 of histone H3. This methylated H3-K27 chromatin mark is commonly associated with silencing of differentiation genes in organisms ranging from plants to flies to humans. Studies on human tumors show that EZH2 is frequently over-expressed in a wide variety of cancerous tissue types, including prostate and breast. Although the mechanistic contributions of EZH2 to cancer progression are not yet determined, functional links between EZH2-mediated histone methylation and DNA methylation suggest partnership with the gene silencing machinery implicated in tumor suppressor loss. Here we review the basic molecular biology of EZH2 and the findings that implicate EZH2 in different cancers. We also discuss EZH2 connections to other silencing enzymes, such as DNA methyltransferases and histone deacetylases, and we consider progress on deciphering mechanistic consequences of EZH2 overabundance and its potential roles in tumorigenesis. Finally, we review recent findings that link EZH2 roles in stem cells and cancer, and we consider prospects for integrating EZH2 blockade into strategies for developing epigenetic therapies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Regulation of the INK4b-ARF-INK4a tumour suppressor locus: all for one or one for all.

              The INK4b-ARF-INK4a locus encodes two members of the INK4 family of cyclin-dependent kinase inhibitors, p15(INK4b) and p16(INK4a), and a completely unrelated protein, known as ARF. All three products participate in major tumour suppressor networks that are disabled in human cancer and influence key physiological processes such as replicative senescence, apoptosis and stem-cell self-renewal. Transcription from the locus is therefore kept under strict control. Mounting evidence suggests that although the individual genes can respond independently to positive and negative signals in different contexts, the entire locus might be coordinately suppressed by a cis-acting regulatory domain or by the action of Polycomb group repressor complexes.
                Bookmark

                Author and article information

                Contributors
                xumu123456@126.com
                chenxiaoxiang1987@126.com
                linkang345@126.com
                kaixuanzeng@163.com
                15720802961@163.com
                18795867526@163.com
                panbei9656@163.com
                18360861755@163.com
                540585362@qq.com
                hebangshun@163.com
                panyuqin01@163.com
                sunhuiling1988@yeah.net
                +86 025 52271000 , sk_wang@njmu.edu.cn
                Journal
                J Hematol Oncol
                J Hematol Oncol
                Journal of Hematology & Oncology
                BioMed Central (London )
                1756-8722
                9 January 2019
                9 January 2019
                2019
                : 12
                : 3
                Affiliations
                [1 ]General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, Nanjing, 210006 China
                [2 ]ISNI 0000 0004 1761 0489, GRID grid.263826.b, School of Medicine, , Southeast University, ; Nanjing, 210009 China
                [3 ]ISNI 0000 0000 9255 8984, GRID grid.89957.3a, Department of Laboratory Medicine, Nanjing First Hospital, , Nanjing Medical University, ; Nanjing, 210006 China
                [4 ]GRID grid.452511.6, Department of Laboratory Medicine, , The Second Affiliated Hospital of Nanjing Medical University, ; Nanjing, 210011 China
                Article
                690
                10.1186/s13045-018-0690-5
                6327409
                30626446
                eeac537e-6d12-4172-9c63-db67e51345fb
                © The Author(s). 2019

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 6 August 2018
                : 25 December 2018
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100001809, National Natural Science Foundation of China;
                Award ID: No. 81472027
                Award Recipient :
                Funded by: Key Project of Science and Technology Development of Nanjing Medicine
                Award ID: ZDX16001
                Award Recipient :
                Funded by: Innovation team of Jiangsu provincial health-strengthening engineering by science and education
                Award ID: CXTDB2017008
                Award Recipient :
                Funded by: Jiangsu Youth Medical Talents Training Project
                Award ID: QNRC2016066
                Award ID: QNRC2016074
                Award Recipient :
                Funded by: Nanjing Medical Science and Technique Development Foundation
                Award ID: No. JQX13003
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2019

                Oncology & Radiotherapy
                snhg6,ezh2,microrna,colorectal cancer,cerna
                Oncology & Radiotherapy
                snhg6, ezh2, microrna, colorectal cancer, cerna

                Comments

                Comment on this article