VHL Deficiency Drives Enhancer Activation of Oncogenes in Clear Cell Renal Cell Carcinoma

Total Page:16

File Type:pdf, Size:1020Kb

VHL Deficiency Drives Enhancer Activation of Oncogenes in Clear Cell Renal Cell Carcinoma Published OnlineFirst September 11, 2017; DOI: 10.1158/2159-8290.CD-17-0375 RESEARCH ARTICLE VHL Deficiency Drives Enhancer Activation of Oncogenes in Clear Cell Renal Cell Carcinoma Xiaosai Yao1,2, Jing Tan3,4, Kevin Junliang Lim5, Joanna Koh1, Wen Fong Ooi1, Zhimei Li3, Dachuan Huang3, Manjie Xing1,5,6, Yang Sun Chan1, James Zhengzhong Qu1, Su Ting Tay5, Giovani Wijaya3, Yue Ning Lam1, Jing Han Hong5, Ai Ping Lee-Lim1, Peiyong Guan3, Michelle Shu Wen Ng2, Cassandra Zhengxuan He1, Joyce Suling Lin1, Tannistha Nandi1, Aditi Qamra1,7, Chang Xu5,8, Swe Swe Myint3, James O. J. Davies9, Jian Yuan Goh1, Gary Loh1, Bryan C. Tan10, Steven G. Rozen5, Qiang Yu1, Iain Bee Huat Tan1,11, Christopher Wai Sam Cheng12, Shang Li5, Kenneth Tou En Chang13, Puay Hoon Tan14, David Lawrence Silver9, Alexander Lezhava15, Gertrud Steger16, Jim R. Hughes9, Bin Tean Teh2,3,5,8,17, and Patrick Tan1,5,8,17 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2017 American Association for Cancer Research. 15-CD-17-0375_p1284-1305.indd 1284 10/23/17 2:18 PM Published OnlineFirst September 11, 2017; DOI: 10.1158/2159-8290.CD-17-0375 ABSTRACT Protein-coding mutations in clear cell renal cell carcinoma (ccRCC) have been exten- sively characterized, frequently involving inactivation of the von Hippel– Lindau (VHL ) tumor suppressor. Roles for noncoding cis -regulatory aberrations in ccRCC tumorigenesis, however, remain unclear. Analyzing 10 primary tumor/normal pairs and 9 cell lines across 79 chromatin profi les, we observed pervasive enhancer malfunction in ccRCC, with cognate enhancer-target genes associated with tissue-specifi c aspects of malignancy. Superenhancer profi ling identifi edZNF395 as a ccRCC- specifi c and VHL-regulated master regulator whose depletion causes near-complete tumor elimination in vitro and in vivo . VHL loss predominantly drives enhancer/superenhancer deregulation more so than promoters, with acquisition of active enhancer marks (H3K27ac, H3K4me1) near ccRCC hallmark genes. Mechanistically, VHL loss stabilizes HIF2α–HIF1β heterodimer binding at enhancers, subsequently recruiting histone acetyltransferase p300 without overtly affecting preexisting promoter–enhancer interactions. Subtype-specifi c driver mutations such asVHL may thus propagate unique pathogenic dependencies in ccRCC by modulating epigenomic landscapes and cancer gene expression. SIGNIFICANCE: Comprehensive epigenomic profi ling of ccRCC establishes a compendium of somati- cally altered cis -regulatory elements, uncovering new potential targets including ZNF395, a ccRCC master regulator. Loss of VHL , a ccRCC signature event, causes pervasive enhancer malfunction, with binding of enhancer-centric HIF2α and recruitment of histone acetyltransferase p300 at preexisting lineage-specifi c promoter–enhancer complexes.Cancer Discov; 7(11); 1284–305. ©2017 AACR. See related commentary by Ricketts and Linehan, p. 1221. INTRODUCTION (Myc ), Vhl loss drives spontaneous ccRCC formation in mouse models ( 7–11 ). VHL encodes an E3 ubiquitin ligase ( 12, 13 ) Clear cell renal cell carcinoma (ccRCC) is the most com- that targets HIF1α (HIF1A ) and HIF2α (EPAS1 ) for degrada- mon subtype of kidney cancer, with 338,000 new cases in 2012 tion ( 14, 15 ). VHL loss in ccRCC results in constitutive activa- worldwide ( 1 ). With most ccRCCs being radiochemoresistant, tion of HIF1/2α and subsequent transactivation ( 16, 17 ) of patients with metastatic ccRCC exhibit dismal 8% fi ve-year downstream genes regulating angiogenesis, glycolysis ( 18 ), overall survival ( 2 ). Although targeted therapies inhibiting lipogenesis ( 19, 20 ), cell cycle ( 21 ), and antiapoptosis ( 22 ). angiogenesis and mTOR pathways can lead to initial tumor Most reports studying VHL/HIF transcriptional activation control, most patients develop resistance in less than a year have focused on HIF-bound promoters ( 23–28 ). However, ( 3, 4 ). A better understanding of ccRCC molecular dependen- recent evidence suggests an emerging role for distal enhancer cies and vulnerabilities is thus needed to develop new therapeu- elements in VHL/HIF transcriptional control ( 29, 30 ). For tic strategies for patients who fail standard-of-care treatment. example, HIF2α-bound distal enhancers activate the proto- Loss of the von Hippel–Lindau (VHL ) tumor suppressor oncogenes MYC ( 31 ) and CCND1 ( 21 ) and coincide with is a defi ning feature of ccRCC ( 5, 6 ). When partnered with ccRCC genetic risk alleles. Nevertheless, such studies focused inactivation of additional tumor suppressors (Pbrm1, Bap1, on individual enhancers, and the majority of distal elements Trp53, Rb1, and/or Cdkn2a ) and/or activation of oncogenes in ccRCC remain largely unexplored. 1 Cancer Therapeutics and Stratifi ed Oncology, Genome Institute of Singapore. 15 Translational Research, Genome Institute of Singapore, Singapore, Singapore. 2 Institute of Molecular and Cell Biology, Singapore. Singapore. 16 Institute of Virology, University of Cologne, Fuerst-Pueckler- 3 Laboratory of Cancer Epigenome, Department of Medical Sciences, Strasse, Cologne, Germany. 17 SingHealth/Duke-NUS Institute of Precision National Cancer Centre, Singapore. 4 State Key Laboratory of Oncology Medicine, National Heart Centre Singapore, Singapore . in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China. Note: Supplementary data for this article are available at Cancer Discovery 5 Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Sin- Online (http://cancerdiscovery.aacrjournals.org/). gapore. 6 NUS Graduate School for Integrative Sciences and Engineering, X. Yao, J. Tan, and K.J. Lim contributed equally to this article. National University of Singapore, Singapore. 7 Department of Physiol- ogy, Yong Loo Lin School of Medicine, National University of Singapore, Corresponding Authors: Patrick Tan, Duke-NUS Medical School, 8 Col- Singapore. 8 Cancer Science Institute of Singapore, National University lege Road, Singapore 169857, Singapore. Phone: 65-6516-1783; Fax: of Singapore, Singapore. 9 Medical Research Council (MRC) Molecular 65-6221-2402; E-mail: [email protected] ; and Bin Tean Teh, Duke- Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford NUS Medical School , 8 College Road, Singapore 169857, Singapore. Phone: University, United Kingdom. 10 Cardiovascular and Metabolic Disorders 65-6601-1324; Fax: 65-6221-2402; E-mail: teh.bin.tean@ singhealth. Programme, Duke-NUS Medical School, Singapore. 11 Division of Medi- com.sg 12 cal Oncology, National Cancer Centre Singapore, Singapore. Depart- doi: 10.1158/2159-8290.CD-17-0375 ment of Urology, Singapore General Hospital, Singapore. 13 Department 2017 American Association for Cancer Research. of Pathology and Laboratory Medicine, KK Women’s and Children’s Hos- © pital, Singapore. 14 Department of Pathology, Singapore General Hospital, NOvember 2017 CANCER DISCOVERY | 1285 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2017 American Association for Cancer Research. 15-CD-17-0375_p1284-1305.indd 1285 10/23/17 2:18 PM Published OnlineFirst September 11, 2017; DOI: 10.1158/2159-8290.CD-17-0375 RESEARCH ARTICLE Yao et al. Delineating the global ccRCC cis-regulatory landscape may Fig. S1A). Among the 10 primary ccRCCs, 9 harbored VHL also identify master regulators involved in tissue-specific mutations, detected by targeted sequencing and confirmed by disease processes. Compared with promoters that are largely Sanger sequencing (Supplementary Table S3). Cell lines 786-O cell-type invariant, distal enhancers integrate multiple line- and A-498 also harbor VHL truncating mutations (Supplemen- age- and context-dependent signals, catering to the special- tary Table S3). The VHL mutations co-occurred with somatic ized needs of diverse cell types and diseases (32, 33). In cancer, mutations of other chromatin modifiers commonly found in such master regulators are frequently located near “super- ccRCC, including PBRM1 (7/10), SETD2 (1/10), KDM5A (1/10), enhancers” or “stretch-enhancers” marked by long stretches KDM5C (1/10), ARID1A (1/10), and KMT2C (1/10). of H3K27ac signals (34, 35). For example, subtype-specific Specific histone modifications can distinguish different genomic alterations such as EGFRvIII in glioblastoma (36) categories of functional regulatory elements—H3K4me3 is and EWS–FLI in Ewing sarcoma (37) induce de novo enhanc- generally associated with promoters, H3K4me1 with enhanc- ers, causing reactivation of developmental master regula- ers, and H3K27ac with active elements (33, 43). Integrating tors required for self-renewal and lineage specification (36). signals from three histone marks and GENCODE v19 anno- Although VHL inactivation has been shown to modulate tated transcription start sites (TSS), we defined active promot- protein levels of different histone modifiers (e.g., KDM5C/ ers as H3K27ac+/H3K4me3+/±2.0 kb TSS regions, and distal JARID1C, ref. 38; HDAC1, ref. 39; JMJD1A, ref. 40; JMJD2B, enhancers as H3K27ac+/H3K4me1+ regions not overlapping ref. 40; JMJD2C, ref. 41), the impact of these protein altera- with promoters. Focusing on epigenomic events specific to tions at specific epigenomic loci remains unclear. Moreover, somatic cancer cells, we derived cell lines from five primary previous studies profiling histone modifications in ccRCCs tumors and, combined with the commercial lines, excluded have also been limited by small sample sizes (2 cases; ref. 42), peaks not found in any of the cell lines to reduce confounding
Recommended publications
  • Hypoxia-Driven Effects in Cancer: Characterization, Mechanisms, and Therapeutic Implications
    cells Review Hypoxia-Driven Effects in Cancer: Characterization, Mechanisms, and Therapeutic Implications Rachel Shi, Chengheng Liao and Qing Zhang * Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; [email protected] (R.S.); [email protected] (C.L.) * Correspondence: [email protected]; Tel.: +1-214-645-4671 Abstract: Hypoxia, a common feature of solid tumors, greatly hinders the efficacy of conventional cancer treatments such as chemo-, radio-, and immunotherapy. The depletion of oxygen in proliferat- ing and advanced tumors causes an array of genetic, transcriptional, and metabolic adaptations that promote survival, metastasis, and a clinically malignant phenotype. At the nexus of these intercon- nected pathways are hypoxia-inducible factors (HIFs) which orchestrate transcriptional responses under hypoxia. The following review summarizes current literature regarding effects of hypoxia on DNA repair, metastasis, epithelial-to-mesenchymal transition, the cancer stem cell phenotype, and therapy resistance. We also discuss mechanisms and pathways, such as HIF signaling, mitochon- drial dynamics, exosomes, and the unfolded protein response, that contribute to hypoxia-induced phenotypic changes. Finally, novel therapeutics that target the hypoxic tumor microenvironment or interfere with hypoxia-induced pathways are reviewed. Keywords: hypoxia; metastasis; hypoxia-inducible factors; chemoresistance Citation: Shi, R.; Liao, C.; Zhang, Q. Hypoxia-Driven Effects in Cancer: 1. Introduction Characterization, Mechanisms, and Understanding the mechanisms by which cells sense oxygen and maintain oxygen Therapeutic Implications. Cells 2021, homeostasis is of pivotal importance for science and medicine. Only in recent decades have 10, 678. https://doi.org/10.3390/ breakthrough discoveries of mechanisms for eukaryotic oxygen sensing been made.
    [Show full text]
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [Show full text]
  • Mutant IDH, (R)-2-Hydroxyglutarate, and Cancer
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer Julie-Aurore Losman1 and William G. Kaelin Jr.1,2,3 1Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA; 2Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA Mutations in metabolic enzymes, including isocitrate whether altered cellular metabolism is a cause of cancer dehydrogenase 1 (IDH1) and IDH2, in cancer strongly or merely an adaptive response of cancer cells in the face implicate altered metabolism in tumorigenesis. IDH1 of accelerated cell proliferation is still a topic of some and IDH2 catalyze the interconversion of isocitrate and debate. 2-oxoglutarate (2OG). 2OG is a TCA cycle intermediate The recent identification of cancer-associated muta- and an essential cofactor for many enzymes, including tions in three metabolic enzymes suggests that altered JmjC domain-containing histone demethylases, TET cellular metabolism can indeed be a cause of some 5-methylcytosine hydroxylases, and EglN prolyl-4-hydrox- cancers (Pollard et al. 2003; King et al. 2006; Raimundo ylases. Cancer-associated IDH mutations alter the enzymes et al. 2011). Two of these enzymes, fumarate hydratase such that they reduce 2OG to the structurally similar (FH) and succinate dehydrogenase (SDH), are bone fide metabolite (R)-2-hydroxyglutarate [(R)-2HG]. Here we tumor suppressors, and loss-of-function mutations in FH review what is known about the molecular mechanisms and SDH have been identified in various cancers, in- of transformation by mutant IDH and discuss their im- cluding renal cell carcinomas and paragangliomas.
    [Show full text]
  • An Animal Model with a Cardiomyocyte-Specific Deletion of Estrogen Receptor Alpha: Functional, Metabolic, and Differential Netwo
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2014 An animal model with a cardiomyocyte-specific deletion of estrogen receptor alpha: Functional, metabolic, and differential network analysis Sriram Devanathan Washington University School of Medicine in St. Louis Timothy Whitehead Washington University School of Medicine in St. Louis George G. Schweitzer Washington University School of Medicine in St. Louis Nicole Fettig Washington University School of Medicine in St. Louis Attila Kovacs Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Devanathan, Sriram; Whitehead, Timothy; Schweitzer, George G.; Fettig, Nicole; Kovacs, Attila; Korach, Kenneth S.; Finck, Brian N.; and Shoghi, Kooresh I., ,"An animal model with a cardiomyocyte-specific deletion of estrogen receptor alpha: Functional, metabolic, and differential network analysis." PLoS One.9,7. e101900. (2014). https://digitalcommons.wustl.edu/open_access_pubs/3326 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Sriram Devanathan, Timothy Whitehead, George G. Schweitzer, Nicole Fettig, Attila Kovacs, Kenneth S. Korach, Brian N. Finck, and Kooresh I. Shoghi This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/3326 An Animal Model with a Cardiomyocyte-Specific Deletion of Estrogen Receptor Alpha: Functional, Metabolic, and Differential Network Analysis Sriram Devanathan1, Timothy Whitehead1, George G. Schweitzer2, Nicole Fettig1, Attila Kovacs3, Kenneth S.
    [Show full text]
  • ARTICLE Doi:10.1038/Nature10523
    ARTICLE doi:10.1038/nature10523 Spatio-temporal transcriptome of the human brain Hyo Jung Kang1*, Yuka Imamura Kawasawa1*, Feng Cheng1*, Ying Zhu1*, Xuming Xu1*, Mingfeng Li1*, Andre´ M. M. Sousa1,2, Mihovil Pletikos1,3, Kyle A. Meyer1, Goran Sedmak1,3, Tobias Guennel4, Yurae Shin1, Matthew B. Johnson1,Zˇeljka Krsnik1, Simone Mayer1,5, Sofia Fertuzinhos1, Sheila Umlauf6, Steven N. Lisgo7, Alexander Vortmeyer8, Daniel R. Weinberger9, Shrikant Mane6, Thomas M. Hyde9,10, Anita Huttner8, Mark Reimers4, Joel E. Kleinman9 & Nenad Sˇestan1 Brain development and function depend on the precise regulation of gene expression. However, our understanding of the complexity and dynamics of the transcriptome of the human brain is incomplete. Here we report the generation and analysis of exon-level transcriptome and associated genotyping data, representing males and females of different ethnicities, from multiple brain regions and neocortical areas of developing and adult post-mortem human brains. We found that 86 per cent of the genes analysed were expressed, and that 90 per cent of these were differentially regulated at the whole-transcript or exon level across brain regions and/or time. The majority of these spatio-temporal differences were detected before birth, with subsequent increases in the similarity among regional transcriptomes. The transcriptome is organized into distinct co-expression networks, and shows sex-biased gene expression and exon usage. We also profiled trajectories of genes associated with neurobiological categories and diseases, and identified associations between single nucleotide polymorphisms and gene expression. This study provides a comprehensive data set on the human brain transcriptome and insights into the transcriptional foundations of human neurodevelopment.
    [Show full text]
  • Epigenetic Regulation of Endothelial-Cell-Mediated Vascular Repair Sylvain Fraineau1,2,3, Carmen G
    REVIEW ARTICLE Epigenetic regulation of endothelial-cell-mediated vascular repair Sylvain Fraineau1,2,3, Carmen G. Palii1,3, David S. Allan1 and Marjorie Brand1,2,3 1 Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada 2 Department of Cellular and Molecular Medicine, University of Ottawa, Canada 3 Ottawa Institute of Systems Biology, Canada Keywords Maintenance of vascular integrity is essential for the prevention of vascular DNA methylation; endothelial progenitors; disease and for recovery following cardiovascular, cerebrovascular and epigenetics; epigenetic drugs; histone peripheral vascular events including limb ischemia, heart attack and stroke. acetylation; histone methylation; non-coding Endothelial stem/progenitor cells have recently gained considerable interest RNAs; stem cell therapy; transcription factors; vascular ischemic disease due to their potential use in stem cell therapies to mediate revascularization after ischemic injury. Therefore, there is an urgent need to understand fun- Correspondence damental mechanisms regulating vascular repair in specific cell types to M. Brand, Sprott Center for Stem Cell develop new beneficial therapeutic interventions. In this review, we high- Research, Regenerative Medicine Program, light recent studies demonstrating that epigenetic mechanisms (including Ottawa Hospital Research Institute, Ottawa post-translational modifications of DNA and histones as well as non-cod- ON K1H8L6, Canada ing RNA-mediated processes) play essential roles in the regulation of endo- Fax: +1 613 739 6294 Tel: +1 613 737 7700 ext. 70336 thelial stem/progenitor cell functions through modifying chromatin E-mail: [email protected] structure. Furthermore, we discuss the potential of using small molecules that modulate the activities of epigenetic enzymes to enhance the vascular (Received 21 October 2014, revised 17 repair function of endothelial cells and offer insight on potential strategies December 2014, accepted 19 December that may accelerate clinical applications.
    [Show full text]
  • UTX Inhibition As Selective Epigenetic Therapy Against TAL1-Driven T-Cell Acute Lymphoblastic Leukemia
    Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press UTX inhibition as selective epigenetic therapy against TAL1-driven T-cell acute lymphoblastic leukemia Aissa Benyoucef,1,2,3 Carmen G. Palii,1,3 Chaochen Wang,4 Christopher J. Porter,5 Alphonse Chu,1 Fengtao Dai,1 Véronique Tremblay,3,6 Patricia Rakopoulos,1 Kulwant Singh,1,3 Suming Huang,7 Francoise Pflumio,8,9,10 Josée Hébert,11,12 Jean-Francois Couture,3,6 Theodore J. Perkins,1,5,6 Kai Ge,4 F. Jeffrey Dilworth,1,2,3 and Marjorie Brand1,2,3 1The Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada; 2Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada; 3Ottawa Institute for Systems Biology, Ottawa, Ontario K1H 8L6, Canada; 4National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA; 5Ottawa Bioinformatics Core Facility, The Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada; 6Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; 7Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, Florida 32610, USA; 8Commissariat á l’Energie Atomique et aux Energies Alternatives, Direction des Sciences du Vivant (DSV)-Institut de Recherche en Radiobiologie Cellulaire et Moléculaire
    [Show full text]
  • Role of CREB on Heme Oxygenase-1 Induction in Adrenal Cells: Involvement of the PI3K Pathway
    57 2 F ASTORT, E M REPETTO and CREB mediates adrenal HO-1 57: 2 113–124 Research others induction by cAMP Role of CREB on heme oxygenase-1 induction in adrenal cells: involvement of the PI3K pathway F Astort1,*, E M Repetto1,*, L Rocha-Viegas2, M E Mercau1, S Sanchez Puch1, C V Finkielstein3, A Pecci2 and C B Cymeryng1 1Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, CEFYBO-CONICET, Buenos Aires, Argentina 2Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Correspondence Aires, IFIBYNE-CONICET, Buenos Aires, Argentina should be addressed 3Integrated Cellular Responses Laboratory, Department of Biological Sciences, Polytechnic Institute and to C B Cymeryng State University, Blacksburg, Virginia, USA Email *(F Astort and E M Repetto contributed equally to this work) [email protected] Abstract In addition to the well-known function of ACTH as the main regulator of adrenal Key Words steroidogenesis, we have previously demonstrated its effect on the transcriptional f heme oxygenase stimulation of HO-1 expression, a component of the cellular antioxidant defense system. f cAMP/PKA In agreement, we hereby demonstrate that, in adrenocortical Y1 cells, HO-1 induction f adrenocortical cells correlates with a significant prevention of the generation of reactive oxygen species f CREB 2+ induced by H2O2/Fe . ACTH/cAMP-dependent activation of redox-imbalanced related f PI3K factors such as NRF2 or NFκB and the participation of MAPKs in this mechanism was, however, discarded based on results with specific inhibitors and reporter plasmids. We Journal of Molecular Endocrinology suggest the involvement of CREB in HO-1 induction by ACTH/cAMP, as transfection of cells with a dominant-negative isoform of CREB (DN-CREB-M1) decreased, while overexpression of CREB increased HO-1 protein levels.
    [Show full text]
  • Cell-Specific Proteomic Analysis in Caenorhabditis Elegans
    Supporting Information Appendix (265 Pages) for Cell-Specific Proteomic Analysis in Caenorhabditis elegans Authors: Kai P. Yueta, Meenakshi K. Domab, c, John T. Ngoa, 2, Michael J. Sweredoskid, Robert L. J. Grahamd, 3, Annie Moradiand, Sonja Hessd, Erin M. Schumane, Paul W. Sternbergb,c and David A. Tirrella,1 Author Affiliations: aDivision of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America bDivision of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America cHoward Hughes Medical Institute, California Institute of Technology, Pasadena, California, United States of America dProteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America eMax Planck Institute for Brain Research, Frankfurt am Main, Germany 1To whom correspondence may be addressed. 2Current Address: Department of Pharmacology, University of California, San Diego, La Jolla, California, United States of America 3Current Address: Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom Supporting Information Materials and Methods - Adenosine Triphosphate (ATP)-Pyrophosphate (PPi) Exchange Assay - Chloroform/Methanol Precipitation - Enrichment of p-Azido-L-Phenylalanine-Labeled Proteins - Fluorescence Microscopy of Live C. elegans - Fluorescence Microscopy of p-Azido-L-Phenylalanine-Labeled Proteins in Fixed C. elegans - In-Gel Fluorescence
    [Show full text]
  • Loss of UTX/KDM6A and the Activation of FGFR3 Converge to Regulate Differentiation Gene-Expression Programs in Bladder Cancer
    Loss of UTX/KDM6A and the activation of FGFR3 converge to regulate differentiation gene-expression programs in bladder cancer Douglas Barrowsa, Lijuan Fenga, Thomas S. Carrollb, and C. David Allisa,1 aLaboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065; and bBioinformatics Resource Center, The Rockefeller University, New York, NY 10065 Contributed by C. David Allis, August 18, 2020 (sent for review April 27, 2020; reviewed by Stephen B. Baylin, Kai Ge, and Yang Shi) Bladder cancer prognosis is closely linked to the underlying Bladder tumors have a relatively high mutational burden, un- differentiation state of the tumor, ranging from the less aggres- derlying the importance of understanding the contexts in which sive and most-differentiated luminal tumors to the more aggres- certain alterations have functional roles in tumorigenesis (6). sive and least-differentiated basal tumors. Sequencing of bladder The Cancer Genome Atlas (TCGA) contains both sequencing cancer has revealed that loss-of-function mutations in chromatin and gene-expression data for over 400 muscle invasive bladder regulators and mutations that activate receptor tyrosine kinase tumors, providing the opportunity to link transcriptional sub- (RTK) signaling frequently occur in bladder cancer. However, little types with mutational patterns (4). TCGA, in addition to many is known as to whether and how these two types of mutations other sequencing studies, revealed that loss-of-function muta- functionally interact or cooperate to regulate tumor growth and tions in chromatin-modifying enzymes are common across all differentiation state. Here, we focus on loss of the histone de- subtypes of bladder cancer, including those proteins known to methylase UTX (also known as KDM6A) and activation of the posttranslationally modify histones (e.g., UTX/KDM6A, MLL3/ RTK FGFR3, two events that commonly cooccur in muscle invasive KMT2C, MLL4/KMT2D, and EP300) and those that remodel bladder tumors.
    [Show full text]
  • Histone Methylation Regulation in Neurodegenerative Disorders
    International Journal of Molecular Sciences Review Histone Methylation Regulation in Neurodegenerative Disorders Balapal S. Basavarajappa 1,2,3,4,* and Shivakumar Subbanna 1 1 Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; [email protected] 2 New York State Psychiatric Institute, New York, NY 10032, USA 3 Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA 4 New York University Langone Medical Center, Department of Psychiatry, New York, NY 10016, USA * Correspondence: [email protected]; Tel.: +1-845-398-3234; Fax: +1-845-398-5451 Abstract: Advances achieved with molecular biology and genomics technologies have permitted investigators to discover epigenetic mechanisms, such as DNA methylation and histone posttransla- tional modifications, which are critical for gene expression in almost all tissues and in brain health and disease. These advances have influenced much interest in understanding the dysregulation of epigenetic mechanisms in neurodegenerative disorders. Although these disorders diverge in their fundamental causes and pathophysiology, several involve the dysregulation of histone methylation- mediated gene expression. Interestingly, epigenetic remodeling via histone methylation in specific brain regions has been suggested to play a critical function in the neurobiology of psychiatric disor- ders, including that related to neurodegenerative diseases. Prominently, epigenetic dysregulation currently brings considerable interest as an essential player in neurodegenerative disorders, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), Amyotrophic lateral sclerosis (ALS) and drugs of abuse, including alcohol abuse disorder, where it may facilitate connections between genetic and environmental risk factors or directly influence disease-specific Citation: Basavarajappa, B.S.; Subbanna, S.
    [Show full text]
  • Supplemental Digital Content 2 List of Down-Regulated Transcripts In
    Supplemental Digital Content 2 List of down-regulated transcripts in colonic mucosal UC-associated dysplasia and pancolitis compared to left-sided colitis Probe Set ID Gene Title Gene Symbol 225655_at ubiquitin-like, containing PHD and RING finger domains, 1 UHRF1 216640_s_at protein disulfide isomerase family A, member 6 PDIA6 1555358_a_at ectonucleoside triphosphate diphosphohydrolase 4 ENTPD4 201340_s_at ectodermal-neural cortex (with BTB-like domain) ENC1 203843_at ribosomal protein S6 kinase, 90kDa, polypeptide 3 RPS6KA3 206668_s_at secretory carrier membrane protein 1 SCAMP1 207791_s_at RAB1A, member RAS oncogene family RAB1A 228996_at roquin KIAA2025 Transcribed locus, weakly similar to XP_517454.1 PREDICTED: 228623_at similar to hypothetical protein MGC45438 [Pan troglodytes] --- 202817_s_at synovial sarcoma translocation, chromosome 18 SS18 235234_at hypothetical protein FLJ36874 FLJ36874 201514_s_at Ras-GTPase-activating protein SH3-domain-binding protein G3BP 218238_at GTP binding protein 4 GTPBP4 241343_at Ribonuclease H1 RNASEH1 1553033_at synaptotagmin-like 5 SYTL5 203210_s_at replication factor C (activator 1) 5, 36.5kDa RFC5 213875_x_at chromosome 6 open reading frame 62 C6orf62 1553108_at hypothetical protein FLJ37562 FLJ37562 215820_x_at sorting nexin 13 SNX13 230130_at Slit homolog 2 (Drosophila) SLIT2 200607_s_at RAD21 homolog (S. pombe) RAD21 235296_at eukaryotic translation initiation factor 5A2 EIF5A2 1552656_s_at U2AF homology motif (UHM) kinase 1 UHMK1 214155_s_at La ribonucleoprotein domain family, member 4 LARP4
    [Show full text]