Characterizing and Selectively Targeting RNF20 Defects Within Colorectal Cancer Cells by Brent John Guppy a Thesis Submitted To

Total Page:16

File Type:pdf, Size:1020Kb

Characterizing and Selectively Targeting RNF20 Defects Within Colorectal Cancer Cells by Brent John Guppy a Thesis Submitted To Characterizing and Selectively Targeting RNF20 Defects Within Colorectal Cancer Cells by Brent John Guppy A Thesis submitted to the Faculty of Graduate Studies of The University of Manitoba In partial fulfillment of the requirements of the degree of Doctor of Philosophy Department of Biochemistry and Medical Genetics University of Manitoba Winnipeg Copyright © 2016 by Brent John Guppy ABSTRACT By 2030, the global colorectal cancer burden is projected to approximately double. This highlights the immediate need to expand our understanding of the etiological origins of colorectal cancer, so that novel therapeutic strategies can be identified and validated. The putative tumor suppressor gene RNF20 encodes a histone H2B mono-ubiquitin ligase and has been found altered/mutated in colorectal and numerous other cancer types. Several studies suggest that RNF20, and by extension mono-ubiquitinated histone H2B (H2Bub1), play important roles in maintaining genome stability in human cells. Indeed, hypomorphic RNF20 expression and/or function have been shown to underlie several phenotypes consistent with genome instability, making aberrant RNF20 biology a potential driver in oncogenesis. Through an evolutionarily conserved trans-histone pathway, RNF20 and H2Bub1 have been shown to modulate downstream di-methylation events at lysines 4 (H3K4me2) and 79 (H3K79me2) of histone H3. Accordingly, understanding the biology associated with RNF20, H2Bub1, H3K4me2, and H3K79me2 is an essential preliminary step towards understanding the etiological origins of cancer-associated RNF20 alterations and identifying a novel therapeutic strategy to selectively kill RNF20-deficient cancers. In this thesis, I employ single-cell imaging, and multiple biochemical techniques to investigate the spatial and temporal patterning and characterize the biology of RNF20, H2Bub1, H3K4me2 and H3K79me2 throughout the cell cycle. In addition, I employ the CRISPR-Cas9 genome editing system to generate RNF20-deficient HCT116 cells. Finally, I employ synthetic lethal strategies to selectively kill RNF20-depleted cells. In conclusion, the research chapters contained within this thesis have characterized putative drivers in cancer (Chapter 3), generated a valuable research reagent for CRISPR-Cas9 ii genome editing experiments (Chapter 4), and identified a novel therapeutic strategy to selectively kill certain cancer cells (Chapter 5). This thesis has increased our understanding of the etiological origins of cancer and generated novel reagents and treatments strategies that after further validation and clinical studies, could be employed to reduce morbidity and mortality rates associated with cancer. iii ACKNOWLEDGEMENTS First and foremost I would like to thank my supervisor, Dr. Kirk McManus, for providing me with guidance, support, and knowledge throughout the years. I am truly fortunate to have been a team member within the McManus lab. Kirk you are truly a science-mentor rockstar. I have enjoyed watching the lab grow over the past 6 years and look forward to the future. I would also like to thank my research committee, Drs. Mai, Davie, and Kung for providing me with valuable input and direction during my studies at the University of Manitoba. I would also like to thank the additional members of my Candidacy Examination Committee, Drs. Wigle, Ogilvie- Werbowetski, Kung, Gietz, and Murphy. I would like to acknowledge additional individuals for technical assistance during my PhD program including Zelda Lichtensztejn, Babu Sajesh, Laura Thompson, Shannon Healy, Amy Cisyk, Aaron MacAulay, Jeff Schacter, Signe Penner-Goeke, Raghvendra Vishwakarma, Alexandra Kuzyk, Nicole Wilkinson, Laryssa Sawchuk and Nermin Moujani. I would like to acknowledge the various agencies that have provided me funding in the form of studentships and travel awards during my tenure as a PhD student; the Mindel and Tom Olenick Research Studentship in Medicine, the Sheu L. Lee Family Scholarship in Oncology, the Manitoba Health Research Council PhD Studentship, the Dieter R. Hettig Scholarship in Cancer Biology, the Rt. Hon. Don Mazankowski Award for Excellence in Oncology Research, the Faculty of Graduate Studies Travel Award, the Canadian Cancer Research Alliance Conference Travel Award, and the Canadian Society for Molecular Biosciences Travel Award. I would like to also thank those who I consider family and who have always supported my dreams; Mom, Dad, Ted, Aaron, Ryan, Christine, Craig, Camille, Mark, Jill, Sajesh, and Zelda. Thank you very much for all your support. iv TABLE OF CONTENTS ABSTRACT………………………………………………………………………………... ii ACKNOWLEDGEMENTS………………………………………………………………... iv TABLE OF CONTENTS…………………………………………………………………... v LIST OF ABBREVIATIONS……………………………………………………………… ix LIST OF TABLES…………………………………………………………………………. xii LIST OF FIGURES………………………………………………………………………... xiv USED WITH PERMISSION AND CONTRIBUTION OF AUTHOR……………………. xvi Chapter 1 - INTRODUCTION 1.0.0 The Future Cancer Burden in Developed Nations…………………………………… 1 1.0.1 The Future Cancer Burden in Canada……………………………………… 2 1.0.2 A Focus on Colorectal Cancer in Canada………………………………….. 3 1.0.3 Colorectal Cancer Diagnosis, Clinical Pathogenesis, and Treatment……… 4 1.1.0 The Chromosome Instability Phenotype……………………………………………... 5 1.1.1 Numerical and Structural Chromosome Instability………………………… 7 1.1.2 Mechanisms of CIN………………………………………………………… 7 1.1.3 The Biological Impact of Chromosome Instability………………………… 9 1.1.4 Therapeutic Interventions Targeting Chromosome Instability…………….. 10 1.2.0 Histone Post-Translational Modifications - Structure and Function………………… 11 1.2.1 The Regulation and Abundance of Histone PTMs Impact Genome Stability…………………………………………………………… 13 1.2.2 Regulation of Chromatin Structure via Histone PTMs and the Link to Oncogenesis………………………………………………………... 15 1.3.0 Histone H2B Ubiquitination…………………………………………………………. 16 1.3.1 H2Bub1, RNF20, RNF40 and their Roles in Oncogenesis………………… 17 1.3.2 H2Bub1 Promotes Tumor Suppressing Transcriptional Profiles…………... 20 1.3.3 H2Bub1 Plays a Role in DNA Damage Repair…………………………….. 22 1.3.4 Rnf20/Rnf40 are Suppressors of CIN……………………………………… 23 1.3.5 The H2Bub1 Trans-Histone Pathway………………………………………. 24 1.4.0 Histone H3 Methylation……………………………………………………………… 25 1.4.1 H3K79 Methylation………………………………………………………… 27 1.4.2 H3K79 Methylation, DOT1L (KMT4) and their Roles in Oncogenesis…… 29 1.5.0 Entering the Era of Highly-Efficient Human Genome Editing………………………. 31 1.5.1 Exploiting HRR and NHEJ for CRISPR-Cas9 Genome Editing…………... 32 1.5.2 CRISPR-Cas9 - A Historical Prospective………………………………….. 34 1.5.3 Employing CRISPR-Cas9 to Target Mammalian Cells……………………. 35 1.6.0 Synthetic Genetic Targeting in Cancer………………………………………………. 37 1.6.1 Synthetic Lethality…………………………………………………………. 38 1.6.2 Therapeutically Exploiting Genome Instability……………………………. 42 1.6.3 Knowledge-Based Direct Tests to Identify Synthetic Lethal Interactors…... 43 1.6.4 The Prototypic Human Synthetic Lethal Interaction Between BRCA1/2 and PARP1……………................................................................. 44 1.6.5 Employing Knowledge-Based Direct Tests to Identify Novel Synthetic Lethal Interactors……………........................................................ 46 v 1.7.0 Rationale, Hypotheses, and Aims……………………………………………………. 47 1.7.1 Chapter 3 - Mitotic Accumulation of Dimethylated Lysine 79 of Histone H3 is Important for Maintaining Genome Integrity During Mitosis in Human Cells………….................................................................. 47 1.7.2 Chapter 4 - Generation and Phenotypic Characterization of an HCT116-Cas9 Expressing Cell Line…………….......................................... 48 1.7.3 Chapter 5 - RNF20 and PARP1 are Synthetic Lethal Interactors in Human Cells……........................................................................................... 49 Chapter 2 - MATERIALS AND METHODS…………………………………………… 50 2.1 Reagents ……………………………………………………………………………… 50 2.2 Cell Lines and Tissue Culture…………………………………………………………. 50 2.3 Cell Synchronization and Mitotic Enrichment………………………………………… 52 2.4 Flow Cytometry………………………………………………………………………... 52 2.4.1 Ethanol Fixation……………………………………………………………. 52 2.4.2 Propidium Iodide Staining………………………………………………….. 53 2.4.3 Flow Cytometric Analysis…………………………………………………. 53 2.5 Immunoblotting………………………………………………………………………… 54 2.5.1 Whole Cell Lysate Preparation…………………………………………….. 56 2.5.2 Acid-Extraction of Nuclear Proteins……………………………………….. 56 2.5.3 Protein Quantification via Bicinchoninic Acid Assay……………………... 57 2.5.4 Protein Resolution via Electrophoresis and Immunoblotting………………. 57 2.5.5 Semi-Quantitative Immunoblot Analysis…………………………………... 59 2.6 siRNA-Based Silencing………………………………………………………………... 59 2.7 DO1L Inhibition………………………………………………………………………... 60 2.8 Mitotic Chromosome Spreads………………………………………………………….. 60 2.9 Mitotic Defect Analysis Based on Chromatin Morphology…………………………… 61 2.10 Lentiviral and Stable Cell Line Production…………………………………………… 62 2.10.1 pGIPZ Lentiviral shRNA Plasmid Preparations…………………………... 62 2.10.2 Lentiviral Production……………………………………………………… 62 2.10.3 Lentiviral Transduction……………………………………………………. 63 2.11 Immunofluorescent Labeling…………………………………………………………. 64 2.11.1 Indirect Immunofluorescent Labeling…………………………………….. 64 2.12 Antibody Validation - Peptide Competition and Dot Blot Assays…………………… 65 2.13 Image Acquisition……………………………………………………………………. 66 2.14 Image Analyses……………………………………………………………………….. 66 2.14.1 Semi-Quantitative Signal Intensity Normalization and Analyses………… 67 2.14.2 Line Scans………………………………………………………………….
Recommended publications
  • Cytogenetics in Oncology and Haematology
    Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Scope The Atlas of Genetics and Cytogenetics in Oncology and Haematology is a peer reviewed on-line journal in open access, devoted to genes, cytogenetics, and clinical entities in cancer, and cancer-prone diseases. It presents structured review articles ("cards") on genes, leukaemias, solid tumours, cancer-prone diseases, more traditional review articles on these and also on surrounding topics ("deep insights"), case reports in hematology, and educational items in the various related topics for students in Medicine and in Sciences. Editorial correspondance Jean-Loup Huret Genetics, Department of Medical Information, University Hospital F-86021 Poitiers, France tel +33 5 49 44 45 46 or +33 5 49 45 47 67 [email protected] or [email protected] Staff Mohammad Ahmad, Mélanie Arsaban, Mikael Cordon, Isabelle Dabin, Marie-Christine Jacquemot-Perbal, Maureen Labarussias, Anne Malo, Catherine Morel-Pair, Laurent Rassinoux, Sylvie Yau Chun Wan - Senon, Alain Zasadzinski. Philippe Dessen is the Database Director, and Alain Bernheim the Chairman of the on-line version (Gustave Roussy Institute – Villejuif – France). The Atlas of Genetics and Cytogenetics in Oncology and Haematology (ISSN 1768-3262) is published 12 times a year by ARMGHM, a non profit organisation, and by the INstitute for Scientific and Technical Information of the French National Center for Scientific Research (INIST-CNRS) since 2008. The Atlas is hosted by INIST-CNRS (http://www.inist.fr) http://AtlasGeneticsOncology.org The PDF version of the Atlas of Genetics and Cytogenetics in Oncology and Haematology is a reissue of the original articles published in collaboration with the Institute for Scientific and Technical Information (INstitut de l’Information Scientifique et Technique - INIST) of the French National Center for Scientific Research (CNRS) on its electronic publishing platform I-Revues.
    [Show full text]
  • Dynamic Chromatin Associated Ubiquitination with Cell Cycle
    Dynamic chromatin associated ubiquitination with cell cycle progression in human cancer cells DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Mansi Shyam Arora Graduate Program in Molecular, Cellular and Developmental Biology The Ohio State University 2014 Dissertation Committee: Dr. Jeffrey Parvin, Advisor Dr. Anita Hopper Dr. Mark Parthun Dr. Robin Wharton Copyright by Mansi Shyam Arora 2014 Abstract In this dissertation work, we have analyzed the pattern of ubiquitin conjugates on human chromatin and its changes with the progression of cell cycle. Our work shows that during interphase, ubiquitination marks the transcribed regions of the genome. This ubiquitination correlates with the ubiquitination of H2B, is dependent on active transcription and is removed during mitosis. We had anticipated that all the ubiquitin associated with the transcribed regions would be removed from chromatin during mitosis, but contrary to our expectation, we found that at the promoters of active genes chromatin ubiquitination levels actually increase thus implying this modification as a possible mitotic bookmark. In the second half of this project, we set out to identify the substrate modified by this post translational modification at these promoters and the enzymes involved in its deposition and removal before and after mitosis respectively. Our results show the surprising involvement of the SAGA associated deubiquitinase USP22 and the polycomb complex proteins BMI1 and RING1A in the regulation of this bookmark during mitosis. The polycomb complex proteins are thought to primarily regulate gene expression by transcriptional repression. Although some previous studies have implied the involvement ii of the polycomb proteins in the regulation of active genes, their association with the transcriptional regulation of active genes during the mitosis to G1 transition has not been described before this work.
    [Show full text]
  • Deep Insights Into the Response of Human Cervical Carcinoma Cells to a New Cyano Enone-Bearing Triterpenoid Soloxolone Methyl: a Transcriptome Analysis
    www.oncotarget.com Oncotarget, 2019, Vol. 10, (No. 51), pp: 5267-5297 Research Paper Deep insights into the response of human cervical carcinoma cells to a new cyano enone-bearing triterpenoid soloxolone methyl: a transcriptome analysis Andrey V. Markov1, Alexander E. Kel1,2, Oksana V. Salomatina1,3, Nariman F. Salakhutdinov3, Marina A. Zenkova1 and Evgeniya B. Logashenko1 1Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation 2geneXplain GmbH, Wolfenbüttel 38302, Germany 3N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russian Federation Correspondence to: Andrey V. Markov, email: [email protected] Keywords: soloxolone methyl; cervical carcinoma; CDDO-Me; cytoscape; mechanism of action Received: April 18, 2019 Accepted: June 19, 2019 Published: September 03, 2019 Copyright: Markov et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Semisynthetic triterpenoids, bearing cyano enone functionality in ring A, are considered now as novel promising anti-tumor agents. However, despite the large- scale studies, their effects on cervical carcinoma cells and, moreover, mechanisms underlying cell death activation by such compounds in this cell type have not been fully elucidated. In this work, we attempted to reconstitute the key pathways and master regulators involved in the response of human cervical carcinoma KB-3-1 cells to the novel glycyrrhetinic acid derivative soloxolone methyl (SM) by a transcriptomic approach.
    [Show full text]
  • Transcriptional Alterations During Mammary Tumor Progression in Mice
    TRANSCRIPTIONAL ALTERATIONS DURING MAMMARY TUMOR PROGRESSION IN MICE AND HUMANS By Karen Fancher B.S. Hartwick College, 1995 A THESIS Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy (Interdisciplinary in Functional Genomics and in Biomedical Sciences) The Graduate School The University of Maine December, 2008 Advisory Committee: Barbara B. Knowles Ph.D., Professor, The Jackson Laboratory, Co-Advisor Gary A. Churchill Ph.D., Professor, The Jackson Laboratory, Co-Advisor Keith W. Hutchison Ph.D., Professor of Biochemistry and Molecular Biology, The University of Maine, Thesis Committee Chair M. Kate Beard-Tisdale Ph.D., Professor of Spatial Information Science and Engineering, The University of Maine Shaoguang Li M.D., Ph.D., Associate Professor, The Jackson Laboratory LIBRARY RIGHTS STATEMENT In presenting this thesis in partial fulfillment of the requirements for an advanced degree at The University of Maine, I agree that the Library shall make it freely available for inspection. I further agree that permission for "fair use" copying of this thesis for scholarly purposes may be granted by the Librarian. It is understood that any copying or publication of this thesis for financial gain shall not be allowed without my written permission. Signature: Date: TRANSCRIPTIONAL ALTERATIONS DURING MAMMARY TUMOR PROGRESSION IN MICE AND HUMANS By Karen Fancher Thesis Co-Advisors: Dr. Barbara B. Knowles and Dr. Gary A. Churchill An Abstract of the Thesis Presented in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy (Interdisciplinary in Functional Genomics and in Biomedical Sciences) December, 2008 Family history, reproductive factors, hormonal exposures, and subjective immunohisto- chemical evaluations of in situ lesions, and to a lesser extent age, remain the best clinical predictors of an individual’s risk of developing breast cancer.
    [Show full text]
  • Molecular Mechanisms and Outcomes of Arsenic-Induced Histone Acetylation and Microrna Regulation in Cellular Transformation
    Molecular Mechanisms and Outcomes of Arsenic-Induced Histone Acetylation and MicroRNA Regulation in Cellular Transformation. By Sunniyat Rahman - A thesis submitted for the degree of Doctor of Philosophy (PhD). - Imperial College London Centre for Pharmacology and Therapeutics Department of Medicine - 2015 - 1 Abstract Chronic exposure to arsenic causes negative health outcomes, particularly malignant neoplasms of the skin, lung and bladder. Although epidemiological data has associated arsenic exposure to cancer, a clear molecular mechanism has remained elusive. This thesis studied the impact of As2O3 exposure on histone acetylation and microRNA expression at both tolerated and toxic levels in vitro to determine an epigenetic-based mechanism of carcinogenesis. This thesis outlines a framework for identifying tolerated and toxic As2O3 exposures, as a prerequisite to epigenetic characterisation. Tolerated As2O3 exposure increased cellular survival, anchorage-independent colony formation, cell-cycle progression and proliferation in HEK293T cells. HEK293T and UROtsa cells treated with tolerated As2O3 exhibited global H3K9 hyperacetylation at 3 hours and global H3K9 hypoacetylation at 72 hours. This was mediated by an imbalance in the intracellular HDAC2 to PCAF mRNA expression ratio. Global H3K9 hypoacetylation occurred for both tolerated and toxic exposures, giving poor mechanistic differentiation between these separated cellular outcomes. Chromatin immunoprecipitation identified PCAF recruitment, E2F1 binding and H3K9 acetylation at the FOS proto-oncogenic promoter leading to an elevation in FOS mRNA levels at the tolerated concentration only. This thesis also reports As2O3 induced chromatin relaxation in HEK293T cells followed by a return to nominal levels for the tolerated concentration. This is in contrast to the toxic exposure, which leads to clear chromatin condensation and apoptosis.
    [Show full text]
  • A Novel Expression Signature from the Perspective of Mesenchymal
    Hindawi Journal of Oncology Volume 2021, Article ID 5033416, 14 pages https://doi.org/10.1155/2021/5033416 Research Article A Novel Expression Signature from the Perspective of Mesenchymal-Epithelial Transition for Hepatocellular Carcinoma with Regard to Prognosis, Clinicopathological Features, Immune Cell Infiltration, Chemotherapeutic Efficacy, and Immunosuppressive Molecules Lijun Xu and Qing Zheng Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine Shanghai Jiao Tong University, 160# Pu Jian Ave, Shanghai 200127, China Correspondence should be addressed to Qing Zheng; [email protected] Received 6 May 2021; Revised 30 June 2021; Accepted 14 July 2021; Published 29 July 2021 Academic Editor: Alessandro Granito Copyright © 2021 Lijun Xu and Qing Zheng. -is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Purpose. Mesenchymal-epithelial transition (MET), a reverse biological process to epithelial-mesenchymal transition (EMT), is involved in tumor metastasis and invasion. However, the role of MET-related genes (MRGs) in hepatocellular carcinoma (HCC) prognosis remains unclear. Methods. In this research, we obtained MRGs data and clinical information from public databases. In the TCGA dataset, a prognostic signature for HCC was constructed by the least absolute shrinkage and selection operator (LASSO) method and externally verified using the ICGC dataset. Results. -ere were 148 differentially expressed MRGs (DEMRGs), out of which 37 MRGs were found associated with overall survival (OS) in the univariate Cox analysis.
    [Show full text]
  • WO 2014/172470 A2 23 October 2014 (23.10.2014) P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2014/172470 A2 23 October 2014 (23.10.2014) P O P C T (51) International Patent Classification: (81) Designated States (unless otherwise indicated, for every C12N 15/85 (2006.01) kind of national protection available): AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, (21) International Application Number: BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, PCT/US2014/034387 DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, (22) International Filing Date: HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KN, KP, KR, 16 April 2014 (16.04.2014) KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, (25) Filing Language: English OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, (26) Publication Language: English SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, (30) Priority Data: ZW. 61/812,720 16 April 2013 (16.04.2013) US 61/824,920 17 May 2013 (17.05.2013) US (84) Designated States (unless otherwise indicated, for every 61/858,437 25 July 2013 (25.07.2013) US kind of regional protection available): ARIPO (BW, GH, 61/865,888 14 August 2013 (14.08.2013) US GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, SZ, TZ, UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, (71) Applicant: WHITEHEAD INSTITUTE FOR BIOMED¬ TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, ICAL RESEARCH [US/US]; Nine Cambridge Center, EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, ΓΓ, LT, LU, LV, Cambridge, MA 02142 (US).
    [Show full text]
  • Genetic Interactions of G-Quadruplexes in Humans
    TOOLS AND RESOURCES Genetic interactions of G-quadruplexes in humans Katherine G Zyner1†, Darcie S Mulhearn1†, Santosh Adhikari2, Sergio Martı´nezCuesta1, Marco Di Antonio2, Nicolas Erard1, Gregory J Hannon1, David Tannahill1, Shankar Balasubramanian1,2,3* 1Cancer Research United Kingdom Cambridge Institute, Cambridge, United Kingdom; 2Department of Chemistry, University of Cambridge, Cambridge, United Kingdom; 3School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom Abstract G-quadruplexes (G4) are alternative nucleic acid structures involved in transcription, translation and replication. Aberrant G4 formation and stabilisation is linked to genome instability and cancer. G4 ligand treatment disrupts key biological processes leading to cell death. To discover genes and pathways involved with G4s and gain mechanistic insights into G4 biology, we present the first unbiased genome-wide study to systematically identify human genes that promote cell death when silenced by shRNA in the presence of G4-stabilising small molecules. Many novel genetic vulnerabilities were revealed opening up new therapeutic possibilities in cancer, which we exemplified by an orthogonal pharmacological inhibition approach that phenocopies gene silencing. We find that targeting the WEE1 cell cycle kinase or USP1 deubiquitinase in combination with G4 ligand treatment enhances cell killing. We also identify new genes and pathways regulating or interacting with G4s and demonstrate that the DDX42 DEAD-box helicase is a newly discovered G4-binding protein. DOI: https://doi.org/10.7554/eLife.46793.001 *For correspondence: [email protected] †These authors contributed Introduction equally to this work G-quadruplex secondary structures (G4s) form in nucleic acids through the self-association of gua- nines (G) in G-rich sequences to form stacked tetrad structures (reviewed in Bochman et al., 2012; Competing interest: See Rhodes and Lipps, 2015).
    [Show full text]
  • RNF40 Regulates Gene Expression in an Epigenetic Context-Dependent Manner Wanhua Xie1, Sankari Nagarajan1, Simon J
    Xie et al. Genome Biology (2017) 18:32 DOI 10.1186/s13059-017-1159-5 RESEARCH Open Access RNF40 regulates gene expression in an epigenetic context-dependent manner Wanhua Xie1, Sankari Nagarajan1, Simon J. Baumgart1, Robyn Laura Kosinsky1, Zeynab Najafova1, Vijayalakshmi Kari1, Magali Hennion2, Daniela Indenbirken3, Stefan Bonn2, Adam Grundhoff3, Florian Wegwitz1, Ahmed Mansouri4,5 and Steven A. Johnsen1* Abstract Background: Monoubiquitination of H2B (H2Bub1) is a largely enigmatic histone modification that has been linked to transcriptional elongation. Because of this association, it has been commonly assumed that H2Bub1 is an exclusively positively acting histone modification and that increased H2Bub1 occupancy correlates with increased gene expression. In contrast, depletion of the H2B ubiquitin ligases RNF20 or RNF40 alters the expression of only a subset of genes. Results: Using conditional Rnf40 knockout mouse embryo fibroblasts, we show that genes occupied by low to moderate amounts of H2Bub1 are selectively regulated in response to Rnf40 deletion, whereas genes marked by high levels of H2Bub1 are mostly unaffected by Rnf40 loss. Furthermore, we find that decreased expression of RNF40- dependent genes is highly associated with widespread narrowing of H3K4me3 peaks. H2Bub1 promotes the broadening of H3K4me3 to increase transcriptional elongation, which together lead to increased tissue-specific gene transcription. Notably, genes upregulated following Rnf40 deletion, including Foxl2, are enriched for H3K27me3, which is decreased following Rnf40 deletion due to decreased expression of the Ezh2 gene. As a consequence, increased expression of some RNF40-“suppressed” genes is associated with enhancer activation via FOXL2. Conclusion: Together these findings reveal the complexity and context-dependency whereby one histone modification can have divergent effects on gene transcription.
    [Show full text]
  • The Gene Ontology Handbook
    The Gene Ontology Handbook Editors. Christophe Dessimoz, Nives Škunca Methods in Molecular Biology 1446 Christophe Dessimoz Nives Škunca Editors The Gene Ontology Handbook M ETHODS IN MOLECULAR BIOLOGY Series Editor John M. Walker School of Life and Medical Sciences University of Hertfordshire Hatfield, Hertfordshire, AL10 9AB, UK For further volumes: http://www.springer.com/series/7651 The Gene Ontology Handbook Edited by Christophe Dessimoz Department of Genetics, Evolution & Environment, University College London, London, UK; Swiss Institute of Bioinformatics, Lausanne, Switzerland; Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland; Center of Integrative Genomics, University of Lausanne, Lausanne, Switzerland; Department of Computer Science, University College London, London, UK Nives Škunca Department of Computer Science, ETH Zurich, Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Zurich, Switzerland; University College London, London, UK Editors Christophe Dessimoz Nives Škunca Department of Genetics Department of Computer Science Evolution and Environment ETH Zurich University College London Zurich , Switzerland London , UK SIB Swiss Institute of Bioinformatics Swiss Institute of Bioinformatics Zurich, Switzerland Lausanne, Switzerland University College London Department of Ecology and Evolution London, UK University of Lausanne Lausanne, Switzerland Center of Integrative Genomics University of Lausanne Lausanne, Switzerland Department of Computer Science University College London London,
    [Show full text]