7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Increased Efficacy of Histone Methyltransferase G9a Inhibitors Against MYCN-Amplified Neuroblastoma

      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

          Targeted inhibition of proteins modulating epigenetic changes is an increasingly important priority in cancer therapeutics, and many small molecule inhibitors are currently being developed. In the case of neuroblastoma (NB), a pediatric solid tumor with a paucity of intragenic mutations, epigenetic deregulation may be especially important. In this study we validate the histone methyltransferase G9a/EHMT2 as being associated with indicators of poor prognosis in NB. Immunological analysis of G9a protein shows it to be more highly expressed in NB cell-lines with MYCN amplification, which is a primary determinant of dismal outcome in NB patients. Furthermore, G9a protein in primary tumors is expressed at higher levels in poorly differentiated/undifferentiated NB, and correlates with high EZH2 expression, a known co-operative oncoprotein in NB. Our functional analyses demonstrate that siRNA-mediated G9a depletion inhibits cell growth in all NB cell lines, but, strikingly, only triggers apoptosis in NB cells with MYCN amplification, suggesting a synthetic lethal relationship between G9a and MYCN. This pattern of sensitivity is also evident when using small molecule inhibitors of G9a, UNC0638, and UNC0642. The increased efficacy of G9a inhibition in the presence of MYCN-overexpression is also demonstrated in the SHEP-21N isogenic model with tet-regulatable MYCN. Finally, using RNA sequencing, we identify several potential tumor suppressor genes that are reactivated by G9a inhibition in NB, including the CLU, FLCN, AMHR2, and AKR1C1-3. Together, our study underlines the under-appreciated role of G9a in NB, especially in MYCN-amplified tumors.

          Related collections

          Most cited references35

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

          Set domain-containing protein, G9a, is a novel lysine-preferring mammalian histone methyltransferase with hyperactivity and specific selectivity to lysines 9 and 27 of histone H3.

          The covalent modification of histone tails has regulatory roles in various nuclear processes, such as control of transcription and mitotic chromosome condensation. Among the different groups of enzymes known to catalyze the covalent modification, the most recent additions are the histone methyltransferases (HMTases), whose functions are now being characterized. Here we show that a SET domain-containing protein, G9a, is a novel mammalian lysine-preferring HMTase. Like Suv39 h1, the first identified lysine-preferring mammalian HMTase, G9a transfers methyl groups to the lysine residues of histone H3, but with a 10-20-fold higher activity. It was reported that lysines 4, 9, and 27 in H3 are methylated in mammalian cells. G9a was able to add methyl groups to lysine 27 as well as 9 in H3, compared with Suv39 h1, which was only able to methylate lysine 9. Our data clearly demonstrated that G9a has an enzymatic nature distinct from Suv39 h1 and its homologue h2. Finally, fluorescent protein-labeled G9a was shown to be localized in the nucleus but not in the repressive chromatin domains of centromeric loci, in which Suv39 h1 family proteins were localized. This finding indicates that G9a may contribute to the organization of the higher order chromatin structure of non-centromeric loci.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Marked for death: targeting epigenetic changes in cancer

            Human cancers commonly have mutations in epigenetic regulatory genes, and several small molecules that target epigenetic regulators are in clinical trials. Here, Pfister and Ashworth discuss the biological complexity of epigenetic regulation in cancer and provide an overview of inhibitors that target gain-of-function mutations, as well as synthetic lethal approaches to target loss-of-function mutations in epigenetic regulators.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Functional MYCN signature predicts outcome of neuroblastoma irrespective of MYCN amplification.

              Neuroblastoma is a pediatric tumor of the sympathetic nervous system. MYCN (V-myc myelocytomatosis viral-related oncogene, neuroblastoma derived [avian]) is amplified in 20% of neuroblastomas, and these tumors carry a poor prognosis. However, tumors without MYCN amplification also may have a poor outcome. Here, we identified downstream targets of MYCN by shRNA-mediated silencing MYCN in neuroblastoma cells. From these targets, 157 genes showed an expression profile correlating with MYCN mRNA levels in NB88, a series of 88 neuroblastoma tumors, and therefore represent in vivo relevant MYCN pathway genes. This 157-gene signature identified very poor prognosis tumors in NB88 and independent neuroblastoma cohorts and was more powerful than MYCN amplification or MYCN expression alone. Remarkably, this signature also identified poor outcome of a group of tumors without MYCN amplification. Most of these tumors have low MYCN mRNA levels but high nuclear MYCN protein levels, suggesting stabilization of MYCN at the protein level. One tumor has an MYC amplification and high MYC expression. Chip-on-chip analyses showed that most genes in this signature are directly regulated by MYCN. MYCN induces genes functioning in cell cycle and DNA repair while repressing neuronal differentiation genes. The functional MYCN-157 signature recognizes classical neuroblastoma with MYCN amplification, as well as a newly identified group marked by MYCN protein stabilization.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                27 May 2020
                2020
                : 10
                : 818
                Affiliations
                [1] 1Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol , Bristol, United Kingdom
                [2] 2Department of Cellular Pathology, Southmead Hospital , Bristol, United Kingdom
                [3] 3The Kids Research Institute, The Children's Hospital at Westmead , Westmead, NSW, Australia
                Author notes

                Edited by: Elizabeth R. Lawlor, Michigan Medicine University of Michigan, United States

                Reviewed by: Jaume Mora, Hospital Sant Joan de Déu Barcelona, Spain; Joseph Louis Lasky, Cure 4 the Kids, United States

                *Correspondence: Karim Malik K.T.A.Malik@ 123456bristol.ac.uk

                This article was submitted to Pediatric Oncology, a section of the journal Frontiers in Oncology

                †These authors share first authorship

                Article
                10.3389/fonc.2020.00818
                7269128
                32537432
                da692514-4354-4c40-bbba-f4eba50bffcd
                Copyright © 2020 Bellamy, Szemes, Melegh, Dallosso, Kollareddy, Catchpoole and Malik.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 18 November 2019
                : 27 April 2020
                Page count
                Figures: 8, Tables: 1, Equations: 0, References: 53, Pages: 15, Words: 8166
                Categories
                Oncology
                Original Research

                Oncology & Radiotherapy
                g9a inhibitors,neuroblastoma,mycn,apoptosis,epigenetic therapy
                Oncology & Radiotherapy
                g9a inhibitors, neuroblastoma, mycn, apoptosis, epigenetic therapy

                Comments

                Comment on this article