10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Loss of tumor suppressor IGFBP4 drives epigenetic reprogramming in hepatic carcinogenesis

      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

          Genomic sequencing of hepatocellular carcinoma (HCC) uncovers a paucity of actionable mutations, underscoring the necessity to exploit epigenetic vulnerabilities for therapeutics. In HCC, EZH2-mediated H3K27me3 represents a major oncogenic chromatin modification, but how it modulates the therapeutic vulnerability of signaling pathways remains unknown. Here, we show EZH2 acts antagonistically to AKT signaling in maintaining H3K27 methylome through epigenetic silencing of IGFBP4. ChIP-seq revealed enrichment of Ezh2/H3K27me3 at silenced loci in HBx-transgenic mouse-derived HCCs, including Igfbp4 whose down-regulation significantly correlated with EZH2 overexpression and poor survivals of HCC patients. Functional characterizations demonstrated potent growth- and invasion-suppressive functions of IGFBP4, which was associated with transcriptomic alterations leading to deregulation of multiple signaling pathways. Mechanistically, IGFBP4 stimulated AKT/EZH2 phosphorylation to abrogate H3K27me3-mediated silencing, forming a reciprocal feedback loop that suppressed core transcription factor networks ( FOXA1/HNF1A/HNF4A/KLF9/NR1H4) for normal liver homeostasis. Consequently, the in vivo tumorigenicity of IGFBP4-silenced HCC cells was vulnerable to pharmacological inhibition of EZH2, but not AKT. Our study unveils chromatin regulation of a novel liver tumor suppressor IGFBP4, which constitutes an AKT-EZH2 reciprocal loop in driving H3K27me3-mediated epigenetic reprogramming. Defining the aberrant chromatin landscape of HCC sheds light into the mechanistic basis of effective EZH2-targeted inhibition.

          Related collections

          Most cited references46

          • Record: found
          • Abstract: found
          • Article: found

          Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma

          (2017)
          Liver cancer has the second highest worldwide cancer mortality rate and has limited therapeutic options. We analyzed 363 hepatocellular carcinoma (HCC) cases by whole exome sequencing and DNA copy number analyses, and 196 HCC also by DNA methylation, RNA, miRNA, and proteomic expression. DNA sequencing and mutation analysis identified significantly mutated genes including LZTR1 , EEF1A1 , SF3B1 , and SMARCA4 . Significant alterations by mutation or down-regulation by hypermethylation in genes likely to result in HCC metabolic reprogramming ( ALB , APOB , and CPS1 ) were observed. Integrative molecular HCC subtyping incorporating unsupervised clustering of five data platforms identified three subtypes, one of which was associated with poorer prognosis in three HCC cohorts. Integrated analyses enabled development of a p53 target gene expression signature correlating with poor survival. Potential therapeutic targets for which inhibitors exist include WNT signaling, MDM4, MET, VEGFA, MCL1, IDH1, TERT, and immune checkpoint proteins CTLA-4, PD-1, and PD-L1. Multiplex molecular profiling of human hepatocellular carcinoma patients provides insight into subtype characteristics and points toward key pathways to target therapeutically.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found

            Histological Subtypes of Hepatocellular Carcinoma Are Related To Gene Mutations and Molecular Tumour Classification.

            Our increasing understanding of hepatocellular carcinoma (HCC) biology holds promise for personalized care, however its translation into clinical practice requires a precise knowledge of its relationship to tumour phenotype.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells.

              EZH2 catalyzes trimethylation of histone H3 lysine 27 (H3K27). Point mutations of EZH2 at Tyr641 and Ala677 occur in subpopulations of non-Hodgkin's lymphoma, where they drive H3K27 hypertrimethylation. Here we report the discovery of EPZ005687, a potent inhibitor of EZH2 (K(i) of 24 nM). EPZ005687 has greater than 500-fold selectivity against 15 other protein methyltransferases and has 50-fold selectivity against the closely related enzyme EZH1. The compound reduces H3K27 methylation in various lymphoma cells; this translates into apoptotic cell killing in heterozygous Tyr641 or Ala677 mutant cells, with minimal effects on the proliferation of wild-type cells. These data suggest that genetic alteration of EZH2 (for example, mutations at Tyr641 or Ala677) results in a critical dependency on enzymatic activity for proliferation (that is, the equivalent of oncogene addiction), thus portending the clinical use of EZH2 inhibitors for cancers in which EZH2 is genetically altered.
                Bookmark

                Author and article information

                Journal
                Nucleic Acids Res
                Nucleic Acids Res
                nar
                Nucleic Acids Research
                Oxford University Press
                0305-1048
                1362-4962
                28 September 2018
                10 July 2018
                10 July 2018
                : 46
                : 17
                : 8832-8847
                Affiliations
                [1 ]School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
                [2 ]Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
                [3 ]Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
                [4 ]Department of Liver Disease, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
                [5 ]Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
                [6 ]Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
                [7 ]Disease Model Research Laboratory, Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
                [8 ]Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
                [9 ]Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, State Key Laboratory in Oncology in South China, The Chinese University of Hong Kong, Hong Kong, China
                [10 ]State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
                Author notes
                To whom correspondence should be addressed. Tel: +852 39439842; Fax: +852 26035123; Email: alfredcheng@ 123456cuhk.edu.hk

                The authors wish it to be known that, in their opinion, the first two authors should be regarded as joint First Authors.

                Article
                gky589
                10.1093/nar/gky589
                6158508
                29992318
                94a3449c-aa69-45c9-b4a0-1fdc79fca518
                © The Author(s) 2018. Published by Oxford University Press on behalf of Nucleic Acids Research.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@ 123456oup.com

                History
                : 19 June 2018
                : 01 June 2018
                : 29 August 2017
                Page count
                Pages: 16
                Funding
                Funded by: University Grants Committee 10.13039/501100001839
                Award ID: C4017-14G
                Award ID: C4014-14G
                Funded by: General Research Fund
                Award ID: 14102914
                Award ID: 14120816
                Funded by: Focused Innovations Scheme-Scheme B
                Award ID: 1907309
                Funded by: National Natural Science Foundation of China 10.13039/501100001809
                Award ID: 81403272
                Award ID: 81774240
                Funded by: Shanghai Chenguang Program
                Award ID: 15CG48
                Funded by: Shanghai Rising-Star Program
                Award ID: 17QA1403900
                Funded by: Shanghai training plan of outstanding young medical talents
                Award ID: 2017YQ021
                Categories
                Gene regulation, Chromatin and Epigenetics

                Genetics
                Genetics

                Comments

                Comment on this article