23
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Enhancer of Polycomb and the Tip60 complex repress hematological tumor initiation by negatively regulating JAK/STAT pathway activity

      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

          Myeloproliferative neoplasms (MPNs) are clonal hematopoietic disorders that cause excessive production of myeloid cells. Most MPN patients have a point mutation in JAK2 ( JAK2 V617F ), which encodes a dominant-active kinase that constitutively triggers JAK/STAT signaling. In Drosophila, this pathway is simplified, with a single JAK, Hopscotch (Hop), and a single STAT transcription factor, Stat92E. The hop Tumorous-lethal [ hop Tum ] allele encodes a dominant-active kinase that induces sustained Stat92E activation. Like MPN patients, hop Tum mutants have significantly more myeloid cells, which form invasive tumors. Through an unbiased genetic screen, we found that heterozygosity for Enhancer of Polycomb [ E(Pc)], a component of the Tip60 lysine acetyltransferase complex (also known as KAT5 in humans), significantly increased tumor burden in hop Tum animals. Hematopoietic depletion of E(Pc) or other Tip60 components in an otherwise wild-type background also induced blood cell tumors. The E(Pc) tumor phenotype was dependent on JAK/STAT activity, as concomitant depletion of hop or Stat92E inhibited tumor formation. Stat92E target genes were significantly upregulated in E(Pc)-mutant myeloid cells, indicating that loss of E(Pc) activates JAK/STAT signaling. Neither the hop nor Stat92E gene was upregulated upon hematopoietic E(Pc) depletion, suggesting that the regulation of the JAK/STAT pathway by E(Pc) is dependent on substrates other than histones. Indeed, E(Pc) depletion significantly increased expression of Hop protein in myeloid cells. This study indicates that E(Pc) works as a tumor suppressor by attenuating Hop protein expression and ultimately JAK/STAT signaling. Since loss-of-function mutations in the human homologs of E(Pc) and Tip60 are frequently observed in cancer, our work could lead to new treatments for MPN patients.

          This article has an associated First Person interview with the first author of the paper.

          Abstract

          Editor's choice: Using Drosophila as a low-complexity model for human myeloproliferative neoplasms, the authors identified a conserved mechanism by which the Tip60 lysine acetyltransferase acts as a tumor suppressor by repressing JAK protein expression in a histone-independent manner.

          Related collections

          Most cited references74

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

          Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal.

          The cBioPortal for Cancer Genomics (http://cbioportal.org) provides a Web resource for exploring, visualizing, and analyzing multidimensional cancer genomics data. The portal reduces molecular profiling data from cancer tissues and cell lines into readily understandable genetic, epigenetic, gene expression, and proteomic events. The query interface combined with customized data storage enables researchers to interactively explore genetic alterations across samples, genes, and pathways and, when available in the underlying data, to link these to clinical outcomes. The portal provides graphical summaries of gene-level data from multiple platforms, network visualization and analysis, survival analysis, patient-centric queries, and software programmatic access. The intuitive Web interface of the portal makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries. Here, we provide a practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Reactive Oxygen Species prime Drosophila haematopoietic progenitors for differentiation

            Reactive Oxygen Species (ROS), produced during various electron transfer reactions in vivo are generally considered to be deleterious to cells1. In the mammalian haematopoietic system, haematopoietic stem cells (HSCs) contain low ROS levels, but unexpectedly, the common myeloid progenitors (CMPs), produce significantly elevated levels of ROS2. The functional significance of this difference in ROS level in the two progenitor types remains unresolved2,3. Here, we show that Drosophila multipotent haematopoietic progenitors which are largely akin to the mammalian myeloid progenitors4 display elevated levels of ROS under in vivo physiological conditions, which is downregulated upon differentiation. Scavenging the ROS from these haematopoietic progenitors using in vivo genetic tools, retards their differentiation into mature blood cells. Conversely, increasing the haematopoietic progenitor ROS beyond their basal level triggers precocious differentiation into all three mature blood cell types found in Drosophila, through a signaling pathway that involves JNK and FoxO activation as well as Polycomb downregulation. We conclude that the developmentally regulated, moderately high ROS level in the progenitor population sensitizes them to differentiation, and establishes a signaling role for ROS in the regulation of haematopoietic cell fate. Our results lead to a model that could be extended to reveal a probable signaling role for ROS in the differentiation of CMPs in mammalian haematopoietic development and oxidative stress response.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors.

              The hydroxamic acid (HAA) analogue pan-histone deacetylase (HDAC) inhibitors (HDIs) LAQ824 and LBH589 have been shown to induce acetylation and inhibit the ATP binding and chaperone function of heat shock protein (HSP) 90. This promotes the polyubiquitylation and degradation of the pro-growth and pro-survival client proteins Bcr-Abl, mutant FLT-3, c-Raf, and AKT in human leukemia cells. HDAC6 is a member of the class IIB HDACs. It is predominantly cytosolic, microtubule-associated alpha-tubulin deacetylase that is also known to promote aggresome inclusion of the misfolded polyubiquitylated proteins. Here we demonstrate that in the Bcr-abl oncogene expressing human leukemia K562 cells, HDAC6 can be co-immunoprecipitated with HSP90, and the knock-down of HDAC6 by its siRNA induced the acetylation of HSP90 and alpha-tubulin. Depletion of HDAC6 levels also inhibited the binding of HSP90 to ATP, reduced the chaperone association of HSP90 with its client proteins, e.g. Bcr-Abl, and induced polyubiquitylation and partial depletion of Bcr-Abl. Conversely, the ectopic overexpression of HDAC6 inhibited LAQ824-induced acetylation of HSP90 and alpha-tubulin and reduced LAQ824-mediated depletion of Bcr-Abl, AKT, and c-Raf. Collectively, these findings indicate that HDAC6 is also an HSP90 deacetylase. Targeted inhibition of HDAC6 leads to acetylation of HSP90 and disruption of its chaperone function, resulting in polyubiquitylation and depletion of pro-growth and pro-survival HSP90 client proteins including Bcr-Abl. Depletion of HDAC6 sensitized human leukemia cells to HAA-HDIs and proteasome inhibitors.
                Bookmark

                Author and article information

                Journal
                Dis Model Mech
                Dis Model Mech
                DMM
                dmm
                Disease Models & Mechanisms
                The Company of Biologists Ltd
                1754-8403
                1754-8411
                1 May 2019
                30 May 2019
                30 May 2019
                : 12
                : 5
                : dmm038679
                Affiliations
                [1 ]Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine , New York, NY 10016, USA
                [2 ]Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine , New York, NY 10016, USA
                Author notes
                [*]

                Present address: Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA.

                []Author for correspondence ( erika.bach@ 123456nyu.edu )
                Author information
                http://orcid.org/0000-0001-7551-4813
                http://orcid.org/0000-0002-5997-4489
                Article
                DMM038679
                10.1242/dmm.038679
                6550037
                31072879
                df6dfa2f-0861-4772-b596-3d80ddd52225
                © 2019. Published by The Company of Biologists Ltd

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

                History
                : 27 December 2018
                : 18 April 2019
                Funding
                Funded by: National Institutes of Health;
                Funded by: National Science Foundation, http://dx.doi.org/10.13039/100000001;
                Funded by: Jack Kent Cooke Foundation, http://dx.doi.org/10.13039/100009240;
                Categories
                304
                Dros
                Research Article

                Molecular medicine
                jak/stat,myeloproliferative neoplasms,drosophila,e(pc),tip60,melanotic tumors,lysine acetyltransferases,tumor suppressor

                Comments

                Comment on this article