Impact of Hypoxia on Hepatitis B Virus Replication

Total Page:16

File Type:pdf, Size:1020Kb

Impact of Hypoxia on Hepatitis B Virus Replication i IMPACT OF HYPOXIA ON HEPATITIS B VIRUS REPLICATION NICHOLAS ROSS BAKER FRAMPTON A thesis submitted to the University of Birmingham for the degree of Doctor of Philosophy Institute of Immunology and Immunotherapy College of Medical and Dental Sciences University of Birmingham September 2017 ii University of Birmingham Research Archive e-theses repository This unpublished thesis/dissertation is copyright of the author and/or third parties. The intellectual property rights of the author or third parties in respect of this work are as defined by The Copyright Designs and Patents Act 1988 or as modified by any successor legislation. Any use made of information contained in this thesis/dissertation must be in accordance with that legislation and must be properly acknowledged. Further distribution or reproduction in any format is prohibited without the permission of the copyright holder. ABSTRACT Hepatitis B virus (HBV) is one of the world’s unconquered diseases, with 370 million chronically infected globally. HBV replicates in hepatocytes within the liver that exist under a range of oxygen tensions from 11% in the peri-portal area to 3% in the peri- central lobules. HBV transgenic mice show a zonal pattern of viral antigen with expression in the peri-central areas supporting a hypothesis that low oxygen regulates HBV replication. We investigated this hypothesis using a recently developed in vitro model system that supports HBV replication. We demonstrated that low oxygen significantly increases covalently closed circular viral DNA (cccDNA), viral promoter activity and pre-genomic RNA (pgRNA) levels, consistent with low oxygen boosting viral transcription. Hypoxia inducible factors (HIFs) regulate cellular responses to low oxygen and we investigated a role for HIF-1α or HIF-2α on viral transcription. A combination of HIF inhibitors and silencing of HIF-1α and HIF-2α ablated the effect of low oxygen on cccDNA and pgRNA, suggesting a role in regulating HBV transcription. This study highlights a new role for hepatic oxygen levels to regulate multiple steps in the HBV life cycle and this may impact on future treatments for viral associated pathologies. iii DEDICATION & ACKNOWLEDGEMENTS Firstly, I’d like to thank Professor Jane McKeating for her supervision throughout my PhD and great help in preparing my thesis, her input was invaluable. I’d also like to thank everyone who has ever helped me in the lab. In particular, Peter Balfe, who taught me how to run and analyse PCRs; and Ke Hu, who helped me learn a great many techniques. I’d like to thank all my colleagues from throughout the PhD, both for the fun times and support in the lab. I’d also like to thank Dan Tennant and members of his lab who helped me during the early stages of my PhD. I’d like to thank Chunkyu Ko, Maarten van de Klundert and the Protzer Laboratory for a productive collaboration and help in developing some techniques. I’d also like to thank the Ratcliffe Laboratory for providing many of the reagents used in this study. Finally, I’d like to thank Jane and Peter for giving me the opportunity to study for this PhD and the European Commission for funding my research. Thank you to my friends and fellow PhD students for supporting me throughout the PhD and providing great memories. Thank you to my parents Helen and Jon for their great advice during my PhD and continued financial, educational and emotional support throughout my life. Lastly, I’d like to thank my wife Rosalind, my constant companion and light of my life. iv TABLE OF CONTENTS Abstract ............................................................................................................. iii Dedication & Acknowledgements ...................................................................... iv Table of Contents ............................................................................................... v List of Figures ................................................................................................... vii List of Tables ...................................................................................................... x List of Common Abbreviations ........................................................................... xi 1. Introduction ................................................................................................ 12 1.1 The Liver as the Sight for Hepatitis B Infection ............................................. 12 1.1.1 Liver Structure ....................................................................................... 12 1.1.2 Liver Differentiation ................................................................................ 16 1.2 Hepatitis B Virus ........................................................................................... 17 1.2.1 HBV Epidemiology and Vaccinology ...................................................... 17 1.2.2 HBV Morphology ................................................................................... 25 1.2.3 HBV Entry and Nuclear Import ............................................................... 30 1.2.4 cccDNA Formation................................................................................. 39 1.2.5 cccDNA Transcription and Translation ................................................... 43 1.2.6 HBV Protein Roles and Functions .......................................................... 55 1.3 Hypoxia Inducible Factors ............................................................................. 68 1.3.1 HIF1α .................................................................................................... 72 1.3.2 HIF2α .................................................................................................... 73 1.3.3 HIF3α .................................................................................................... 74 1.4 HBV and Hypoxia ......................................................................................... 75 1.5 Aims ............................................................................................................. 80 2. Materials and Methods............................................................................... 81 2.1 Tissue Culture .............................................................................................. 81 2.2 Transfections ................................................................................................ 81 2.3 HRE Luciferase Assay .................................................................................. 84 2.4 Generation of HBV ........................................................................................ 84 2.5 De novo Infections ........................................................................................ 85 2.6 HBV E and S antigen ELISAs ....................................................................... 87 2.7 Western Blotting ........................................................................................... 88 2.7.1 Lysate preparation and sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) ................................................................. 88 2.7.2 Immuno-blotting and chemiluminescent detection of proteins ................ 88 2.8 Quantitative RT-PCR .................................................................................... 90 2.9 SYBR Green PCR ........................................................................................ 90 2.10 PCR Array ................................................................................................... 95 v 2.11 Immunofluorescent Staining of HBV S Antigen ............................................ 97 2.12 Chromatin Immunoprecipitation (ChIP) ........................................................ 97 2.13 Statistical Analysis ....................................................................................... 98 3. Examining the hepatoma cell response to low oxygen ............................ 100 3.1 Introduction ................................................................................................. 100 3.2 Huh-7.5 cell response to low oxygen .......................................................... 103 3.3 HepG2 cell response to low oxygen ............................................................ 105 3.4 The effect of HIF targeting drugs on hepatoma cell lines ............................ 108 3.5 HIF over expression in hepatoma cells ....................................................... 115 3.6 HIF target gene activation in hepatoma cells .............................................. 117 3.7 Hepatoma cell lines and differentiation ....................................................... 123 3.8 Cellular differentiation status and response to low oxygen .......................... 125 3.9 Cell density, differentiation and HIF responses ........................................... 127 3.10 Discussion .................................................................................................. 132 3.11 Summary .................................................................................................... 134 4. Studying HBV and HIF Stabilisation ........................................................ 135 4.1 Introduction ................................................................................................. 135 4.2 HBV models and HIF expression ................................................................ 139 4.3 HIF stabilisation of HIF is independent of HBx ............................................ 141 4.4
Recommended publications
  • Independence of Hif1a and Androgen Signaling Pathways in Prostate Cancer
    bioRxiv preprint doi: https://doi.org/10.1101/848424; this version posted November 26, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Independence of HIF1a and androgen signaling pathways in prostate cancer Maxine GB Tran1, 2*, Becky AS Bibby3†*, Lingjian Yang3, Franklin Lo1, Anne Warren1, Deepa Shukla1, Michelle Osborne1, James Hadfield1, Thomas Carroll1, Rory Stark1, Helen Scott1, Antonio Ramos-Montoya1, Charlie Massie1, Patrick Maxwell1, Catharine ML West3, 4, Ian G. Mills5,6** and David E. Neal1** 1Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, United Kingdom 2UCL division of Surgery and Interventional Science, Royal Free Hospital, Pond Street, London NW3 2QG 3Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital NHS Trust, Manchester, M20 4BX, United Kingdom 4Manchester Biomedical Research Centre, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom. 5Centre for Cancer Research and Cell Biology, Queens University Belfast, Belfast, BT9 7AE, United Kingdom 6Nuffield Department of Surgical Sciences, University of Oxford, OX3 9DU, UK *These authors contributed equally to this work **These authors contributed equally to this work †Corresponding author email: Becky Bibby, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital NHS Trust, Manchester, M20 4BX, United Kingdom, [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/848424; this version posted November 26, 2019.
    [Show full text]
  • Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug
    cancers Article Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug Response Genes in Pediatric Acute Myeloid Leukemia David G.J. Cucchi 1 , Costa Bachas 1 , Marry M. van den Heuvel-Eibrink 2,3, Susan T.C.J.M. Arentsen-Peters 3, Zinia J. Kwidama 1, Gerrit J. Schuurhuis 1, Yehuda G. Assaraf 4, Valérie de Haas 3 , Gertjan J.L. Kaspers 3,5 and Jacqueline Cloos 1,* 1 Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; [email protected] (D.G.J.C.); [email protected] (C.B.); [email protected] (Z.J.K.); [email protected] (G.J.S.) 2 Department of Pediatric Oncology/Hematology, Erasmus MC–Sophia Children’s Hospital, 3015 CN Rotterdam, The Netherlands; [email protected] 3 Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; [email protected] (S.T.C.J.M.A.-P.); [email protected] (V.d.H.); [email protected] (G.J.L.K.) 4 The Fred Wyszkowski Cancer Research, Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel; [email protected] 5 Emma’s Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1081 HV Amsterdam, The Netherlands * Correspondence: [email protected] Received: 21 April 2020; Accepted: 12 May 2020; Published: 15 May 2020 Abstract: Novel treatment strategies are of paramount importance to improve clinical outcomes in pediatric AML. Since chemotherapy is likely to remain the cornerstone of curative treatment of AML, insights in the molecular mechanisms that determine its cytotoxic effects could aid further treatment optimization.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [Show full text]
  • The Roles of Long Noncoding Rnas HNF1-AS1 and HNF4-AS1 in Drug
    non-coding RNA Review The Roles of Long Noncoding RNAs HNF1α-AS1 and HNF4α-AS1 in Drug Metabolism and Human Diseases Liming Chen 1, Yifan Bao 1, Suzhen Jiang 1,2 and Xiao-bo Zhong 1,* 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA; [email protected] (L.C.); [email protected] (Y.B.); [email protected] (S.J.) 2 School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 51006, China * Correspondence: [email protected]; Tel.: +01-860-486-3697 Received: 25 May 2020; Accepted: 22 June 2020; Published: 24 June 2020 Abstract: Long noncoding RNAs (lncRNAs) are RNAs with a length of over 200 nucleotides that do not have protein-coding abilities. Recent studies suggest that lncRNAs are highly involved in physiological functions and diseases. lncRNAs HNF1α-AS1 and HNF4α-AS1 are transcripts of lncRNA genes HNF1α-AS1 and HNF4α-AS1, which are antisense lncRNA genes located in the neighborhood regions of the transcription factor (TF) genes HNF1α and HNF4α, respectively. HNF1α-AS1 and HNF4α-AS1 have been reported to be involved in several important functions in human physiological activities and diseases. In the liver, HNF1α-AS1 and HNF4α-AS1 regulate the expression and function of several drug-metabolizing cytochrome P450 (P450) enzymes, which also further impact P450-mediated drug metabolism and drug toxicity. In addition, HNF1α-AS1 and HNF4α-AS1 also play important roles in the tumorigenesis, progression, invasion, and treatment outcome of several cancers. Through interacting with different molecules, including miRNAs and proteins, HNF1α-AS1 and HNF4α-AS1 can regulate their target genes in several different mechanisms including miRNA sponge, decoy, or scaffold.
    [Show full text]
  • Ten Commandments for a Good Scientist
    Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Wageningen 2010 Thesis committee Thesis supervisors Prof. dr. ir. Ivonne M.C.M. Rietjens Professor of Toxicology Wageningen University Prof. dr. Albertinka J. Murk Personal chair at the sub-department of Toxicology Wageningen University Thesis co-supervisor Dr. ir. Jacques J.M. Vervoort Associate professor at the Laboratory of Biochemistry Wageningen University Other members Prof. dr. Michael R. Muller, Wageningen University Prof. dr. ir. Huub F.J. Savelkoul, Wageningen University Prof. dr. Everardus J. van Zoelen, Radboud University Nijmegen Dr. ir. Toine F.H. Bovee, RIKILT, Wageningen This research was conducted under the auspices of the Graduate School VLAG Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Thesis submitted in fulfillment of the requirements for the degree of doctor at Wageningen University by the authority of the Rector Magnificus Prof. dr. M.J. Kropff, in the presence of the Thesis Committee appointed by the Academic Board to be defended in public on Tuesday 14 September 2010 at 4 p.m. in the Aula Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds. Ana María Sotoca Covaleda Thesis Wageningen University, Wageningen, The Netherlands, 2010, With references, and with summary in Dutch. ISBN: 978-90-8585-707-5 “Caminante no hay camino, se hace camino al andar. Al andar se hace camino, y al volver la vista atrás se ve la senda que nunca se ha de volver a pisar” - Antonio Machado – A mi madre.
    [Show full text]
  • Homozygous Hypomorphic HNF1A Alleles Are a Novel Cause of Young-Onset Diabetes and Result in Sulphonylurea-Sensitive Diabetes
    Diabetes Care 1 Shivani Misra,1 Neelam Hassanali,2 Homozygous Hypomorphic Amanda J. Bennett,2 Agata Juszczak,2 Richard Caswell,3 Kevin Colclough,3 HNF1A Alleles Are a Novel Cause Jonathan Valabhji,4 Sian Ellard,3 of Young-Onset Diabetes and Nicholas S. Oliver,1,4 and Anna L. Gloyn2,5,6 Result in Sulphonylurea-Sensitive Diabetes https://doi.org/10.2337/dc19-1843 OBJECTIVE Heterozygous loss-of-function mutations in HNF1A cause maturity-onset diabetes of the young (MODY). Affected individuals can be treated with low-dose sulpho- nylureas. Individuals with homozygous HNF1A mutations causing MODY have not been reported. RESEARCH DESIGN AND METHODS We phenotyped a kindred with young-onset diabetes and performed molecular genetic testing, a mixed meal tolerance test, a sulphonylurea challenge, and in vitro assays to assess variant protein function. RESULTS A homozygous HNF1A variant (p.A251T) was identified in three insulin-treated 1 family members diagnosed with diabetes before 20 years of age. Those with the Diabetes, Endocrinology and Metabolism, Im- NOVEL COMMUNICATIONS IN DIABETES homozygous variant had low hs-CRP levels (0.2–0.8 mg/L), and those tested dem- perial College London, London, U.K. 2Oxford Centre for Diabetes, Endocrinology and onstrated sensitivity to sulphonylurea given at a low dose, completely transitioning off Metabolism, University of Oxford, Oxford, U.K. insulin. In silico modeling predicted a variant of unknown significance; however, in vitro 3Institute of Biomedical and Clinical Science, Uni- studies supported a modest reduction in transactivation potential (79% of that for the versity of Exeter Medical School, Exeter, U.K.
    [Show full text]
  • Methylation Profile of Hepatitis B Virus Is Not Influenced by Interferon Α in Human Liver Cancer Cells
    MOLECULAR MEDICINE REPORTS 24: 715, 2021 Methylation profile of hepatitis B virus is not influenced by interferon α in human liver cancer cells IN YOUNG MOON1 and JIN‑WOOK KIM1,2 1Department of Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620; 2Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea Received December 20, 2020; Accepted July 12, 2021 DOI: 10.3892/mmr.2021.12354 Abstract. Interferon (IFN) α is used for the treatment of Introduction chronic hepatitis B virus (HBV) infection, but the molecular mechanisms underlying its antiviral effect have not been fully Hepatitis B virus (HBV) is one of the commonest causes elucidated. Epigenetic modifications regulate the transcrip‑ of chronic hepatitis worldwide (1). The chronicity of HBV tional activity of covalently closed circular DNA (cccDNA) in infection is related to the fact that HBV produces very cells with chronic HBV infection. IFN‑α has been shown to stable viral genome in the host cells (1). The HBV genome modify cccDNA‑bound histones, but it is not known whether is partially double‑stranded relaxed circular DNA (rcDNA), the anti‑HBV effect of IFN‑α involves methylation of cccDNA. which is converted to complete double‑stranded covalently The present study aimed to determine whether IFN‑α induced closed circular DNA (cccDNA) in the nucleus of infected methylation of HBV cccDNA in a cell‑based model in which hepatocytes (2). HBV cccDNA remains the main hurdle in the HepG2 cells were directly infected with wild‑type HBV eradication of infected HBV as current antiviral agents cannot virions.
    [Show full text]
  • Quantification and Epigenetic Evaluation of the Residual Pool Of
    www.nature.com/scientificreports OPEN Quantifcation and epigenetic evaluation of the residual pool of hepatitis B covalently closed circular DNA in long‑term nucleoside analogue‑treated patients Fanny Lebossé1,2,3, Aurore Inchauspé1,2, Maëlle Locatelli1,2, Clothilde Miaglia1,2,3, Audrey Diederichs1,2, Judith Fresquet1,2, Fleur Chapus1,2, Kamal Hamed4, Barbara Testoni1,2* & Fabien Zoulim1,2,3* Hepatitis B virus (HBV) covalently closed circular (ccc)DNA is the key genomic form responsible for viral persistence and virological relapse after treatment withdrawal. The assessment of residual intrahepatic cccDNA levels and activity after long‑term nucleos(t)ide analogues therapy still represents a technical challenge. Quantitative (q)PCR, rolling circle amplifcation (RCA) and droplet digital (dd)PCR assays were used to quantify residual intrahepatic cccDNA in liver biopsies from 56 chronically HBV infected patients after 3 to 5 years of telbivudine treatment. Activity of residual cccDNA was evaluated by quantifying 3.5 kB HBV RNA (preC/pgRNA) and by assessing cccDNA‑associated histone tails post‑transcriptional modifcations (PTMs) by micro‑chromatin immunoprecipitation. Long‑term telbivudine treatment resulted in serum HBV DNA suppression, with most of the patients reaching undetectable levels. Despite 38 out of 56 patients had undetectable cccDNA when assessed by qPCR, RCA and ddPCR assays detected cccDNA in all‑but‑one negative samples. Low preC/pgRNA level in telbivudine‑treated samples was associated with enrichment for cccDNA histone PTMs related to repressed transcription. No diference in cccDNA levels was found according to serum viral markers evolution. This panel of cccDNA evaluation techniques should provide an added value for the new proof‑of‑concept clinical trials aiming at a functional cure of chronic hepatitis B.
    [Show full text]
  • Human/Mouse/Rat HIF-1 Alpha/HIF1A Antibody
    Human/Mouse/Rat HIF-1 alpha/HIF1A Antibody Monoclonal Mouse IgG1 Clone # 241809 Catalog Number: MAB1536 DESCRIPTION Species Reactivity Human/Mouse/Rat Specificity Detects human, mouse, and rat HIF-1 alpha/HIF1A. Source Monoclonal Mouse IgG1 Clone # 241809 Purification Protein A or G purified from hybridoma culture supernatant Immunogen E. coli-derived recombinant human HIF-1 alpha/HIF1A Arg575-Asn826 Accession # Q16665.1 Formulation Lyophilized from a 0.2 μm filtered solution in PBS with Trehalose. See Certificate of Analysis for details. *Small pack size (-SP) is supplied either lyophilized or as a 0.2 μm filtered solution in PBS. APPLICATIONS Please Note: Optimal dilutions should be determined by each laboratory for each application. General Protocols are available in the Technical Information section on our website. Recommended Sample Concentration Western Blot 1 µg/mL See Below Immunohistochemistry 5-25 µg/mL See Below Immunoprecipitation 1-3 µg/500 µg cell MCF‑7 human breast cancer cell line treated with CoCl2, see our available Western blot lysate detection antibodies Simple Western 10 µg/mL See Below Knockout Validated HIF-1 alpha/HIF1A is specifically detected in HeLa human cervical epithelial carcinoma parental cell line but is not detectable in HIF-1 alpha/HIF1A knockout HeLa cell line. DATA Western Blot Immunohistochemistry Detection of Human, Mouse, HIF-1 alpha/HIF1A in Human Kidney. and Rat HIF-1 alpha/HIF1A by HIF-1 alpha/HIF1A was detected in Western Blot. Western blot immersion fixed paraffin-embedded sections shows lysates of MCF-7 human of human kidney using Mouse Anti- breast cancer cell line, Balb-3T3 Human/Mouse/Rat HIF-1 alpha/HIF1A mouse embryonic fibroblast cell Monoclonal Antibody (Catalog # MAB1536) line, and PC-12 rat adrenal at 5 µg/mL for 1 hour at room temperature pheochromocytoma cell line followed by incubation with the Anti-Mouse untreated (-) or treated (+) with IgG VisUCyte™ HRP Polymer Antibody 150 μM CoCl2 for 8 hours.
    [Show full text]
  • Transcriptome Analysis Uncovers the Diagnostic Value of Mir-192-5P/HNF1A-AS1/VIL1 Panel in Cervical Adenocarcinoma
    www.nature.com/scientificreports OPEN Transcriptome analysis uncovers the diagnostic value of miR‑192‑5p/ HNF1A‑AS1/VIL1 panel in cervical adenocarcinoma Junfen Xu1*, Jian Zou1, Luyao Wu1 & Weiguo Lu1,2* Despite the fact that the incidence of cervical squamous cell carcinoma has decreased, there is an increase in the incidence of cervical adenocarcinoma. However, our knowledge on cervical adenocarcinoma is largely unclear. Transcriptome sequencing was conducted to compare 4 cervical adenocarcinoma tissue samples with 4 normal cervical tissue samples. mRNA, lncRNA, and miRNA signatures were identifed to discriminate cervical adenocarcinoma from normal cervix. The expression of VIL1, HNF1A‑AS1, MIR194‑2HG, SSTR5‑AS1, miR‑192‑5p, and miR‑194‑5p in adenocarcinoma were statistically signifcantly higher than that in normal control samples. The Receiver Operating Characteristic (ROC) curve analysis indicated that combination of miR‑192‑5p, HNF1A‑AS1, and VIL1 yielded a better performance (AUC = 0.911) than any single molecule -and could serve as potential biomarkers for cervical adenocarcinoma. Of note, the combination model also gave better performance than TCT test for cervical adenocarcinoma diagnosis. However, there was no correlation between miR‑192‑5p or HNF1A‑AS1 and HPV16/18 E6 or E7. VIL1 was weakly correlated with HPV18 E7 expression. In summary, our study has identifed miR‑192‑5p/HNF1A‑AS1/VIL1 panel that accurately discriminates adenocarcinoma from normal cervix. Detection of this panel may provide considerable clinical value in the diagnosis of cervical adenocarcinoma. Abbreviations SCC Cervical squamous cell carcinoma NcRNA Non-coding RNA MiRNA MicroRNA LncRNA Long non-coding RNA ROC Receiver operating characteristic AUC​ Area under the ROC curve RT-qPCR Quantitative reverse-transcriptase PCR HNF1A-AS1 LncRNA HNF1A antisense RNA 1 VIL1 Villin 1 Cervical cancer ranks fourth for both cancer incidence and mortality in women worldwide 1.
    [Show full text]
  • Inhibition of Jcpyv Infection Mediated by Targeted Viral Genome Editing
    www.nature.com/scientificreports OPEN Inhibition of JCPyV infection mediated by targeted viral genome editing using CRISPR/Cas9 Received: 31 May 2016 Yi-ying Chou1, Annabel Krupp2, Campbell Kaynor2, Raphaël Gaudin1,†, Minghe Ma1, Accepted: 21 October 2016 Ellen Cahir-McFarland2 & Tom Kirchhausen1,3 Published: 14 November 2016 Progressive multifocal leukoencephalopathy (PML) is a debilitating disease resulting from infection of oligodendrocytes by the JC polyomavirus (JCPyV). Currently, there is no anti-viral therapeutic available against JCPyV infection. The clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR- associated protein 9 (Cas9) system (CRISPR/Cas9) is a genome editing tool capable of introducing sequence specific breaks in double stranded DNA. Here we show that the CRISPR/Cas9 system can restrict the JCPyV life cycle in cultured cells. We utilized CRISPR/Cas9 to target the noncoding control region and the late gene open reading frame of the JCPyV genome. We found significant inhibition of virus replication and viral protein expression in cells recipient of Cas9 together with JCPyV-specific single-guide RNA delivered prior to or after JCPyV infection. The JC polyomavirus (JCPyV), a member of the Polyomaviridae family, is the causative agent of a rare and debili- tating demyelinating disease termed progressive multifocal leukoencephalopathy (PML) caused by infection and destruction of oligodendrocytes in the central nervous system (CNS)1,2. PML can develop in the context of immu- nodeficiency and treatment with immunomodulatory drugs3. Indeed, it is one of the most common CNS-related diseases in AIDS, affecting 5% of the HIV-1 positive patients. The incidence of PML has also risen to 0.2–0.4% in patients receiving immunomodulatory therapies4,5.
    [Show full text]
  • Viral Epigenomes in Human Tumorigenesis
    Oncogene (2010) 29, 1405–1420 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc REVIEW Viral epigenomes in human tumorigenesis AF Fernandez1 and M Esteller1,2 1Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain and 2Institucio Catalana de Recerca i Estudis Avanc¸ats (ICREA), Barcelona, Catalonia, Spain Viruses are associated with 15–20% of human cancers is altered in cancer (Fraga and Esteller, 2005; Chuang worldwide. In the last century, many studies were directed and Jones, 2007; Lujambio et al., 2007). towards elucidating the molecular mechanisms and genetic DNA methylation mainly occurs on cytosines that alterations by which viruses cause cancer. The importance precede guanines to yield 5-methylcytosine; these of epigenetics in the regulation of gene expression has dinucleotide sites are usually referred to as CpGs prompted the investigation of virus and host interactions (Herman and Baylin, 2003). CpGs are asymmetrically not only at the genetic level but also at the epigenetic level. distributed into CpG-poor regions and dense regions In this study, we summarize the published epigenetic called ‘CpG islands’, which are located in the promoter information relating to the genomes of viruses directly or regions of approximately half of all genes. These CpG indirectly associated with the establishment of tumori- islands are usually unmethylated in normal cells, with genic processes. We also review aspects such as viral the exceptions listed below, whereas the sporadic CpG replication and latency associated with epigenetic changes sites in the rest of the genome are generally methylated and summarize what is known about epigenetic alterations (Jones and Takai, 2001).
    [Show full text]