Defining a Regulatory Role for the Hsv Glycoprotein B Membrane Proximal Region in Membrane Association

Total Page:16

File Type:pdf, Size:1020Kb

Defining a Regulatory Role for the Hsv Glycoprotein B Membrane Proximal Region in Membrane Association University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2013 Defining A Regulatory Role For The Hsv Glycoprotein B Membrane Proximal Region In Membrane Association Spencer Shelly University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Biology Commons, and the Virology Commons Recommended Citation Shelly, Spencer, "Defining A Regulatory Role For The Hsv Glycoprotein B Membrane Proximal Region In Membrane Association" (2013). Publicly Accessible Penn Dissertations. 799. https://repository.upenn.edu/edissertations/799 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/799 For more information, please contact [email protected]. Defining A Regulatory Role For The Hsv Glycoprotein B Membrane Proximal Region In Membrane Association Abstract Herpes simplex virus (HSV) entry requires four essential glycoproteins (gD, gH/gL, and gB) to enable fusion between the virion envelope and the cellular membrane. The fusion cascade is activated by gD binding to one of its receptors, nectin-1 or HVEM. Glycoprotein B (gB), a class III viral fusion protein, mediates the fusion reaction, while data indicates that gH/gL acts as a regulator of gB. gB is trimeric and has a 773 amino acid ectodomain with a highly hydrophobic membrane proximal region (MPR) (residues 731-773) and two fusion loops (FL) per protomer. The post-fusion structure of gB was solved from the gB(730t) construct, which is truncated to remove the hydrophobic MPR residues. In this dissertation I investigated the MPRs influence on gBs ability ot interact with membranes. I hypothesize that the MPR regulates fusion loop exposure by interacting with the fusion loops and masks them until fusion begins. To investigate this process I constructed a series of MPR deletion, truncation, and point mutations using both full-length mammalian expression vectors and purified baculovirus expressed protein. I found that deletions in the MPR from full-length gB resulted in a disruption in cell surface expression in transfected cells. This suggests the MPR is necessary for proper folding or transport of gB. Soluble gB MPR truncations [gB(759t), gB(749t), gB(739t)] were expressed and purified using the baculovirus expression system, and compared to MPR-less gB(730t) and full MPR containing gB(773t). I found that gB containing an MPR segment were all compromised in their ability to bind liposomes in comparison to gB(730t), which lacks any MPR residues. Supporting our hypothesis we found that residues 731 to 739 were sufficient epr vent liposome association and mutation of two aromatic residues, F732 and F739, to alanine in gB(739t) restored gBs ability to bind liposomes. Together, my data suggests the MPR does indeed regulate gBs ability to associate with liposomes, and that aromatic residues in the MPR are important for this function. This supports our model that the MPR masks the gB FLs to prevent premature membrane association and adds another layer of regulation to the HSV entry cascade. Degree Type Dissertation Degree Name Doctor of Philosophy (PhD) Graduate Group Cell & Molecular Biology First Advisor Roselyn J. Eisenberg Second Advisor Gary H. Cohen Keywords entry, fusion, herpesvirus, hsv Subject Categories Biology | Virology This dissertation is available at ScholarlyCommons: https://repository.upenn.edu/edissertations/799 DEFINING A REGULATORY ROLE FOR THE HSV GLYCOPROTEIN B MEMBRANE PROXIMAL REGION IN MEMBRANE ASSOCIATION Spencer S. Shelly A DISSERTATION in Cell and Molecular Biology Presented to the Faculties of the University of Pennsylvania in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy 2013 Supervisor of Dissertation Co-Supervisor of Dissertation ___________________________ _________________________ Roselyn J. Eisenberg Gary H. Cohen Professor of Microbiology Professor of Microbiology Graduate Group Chairperson ___________________________ Daniel S. Kessler, Associate Professor of Cell and Developmental Biology Dissertation Committee Paul F. Bates, Professor of Microbiology Jeffrey M. Bergelson, Professor of Pediatrics Harvey M. Friedman, Professor of Medicine Yan Yuan, Professor of Microbiology DEDICATION For my wife and daughter ii ACKNOWLEDGMENTS The work presented in this thesis could not have been completed without the tremendous support provided by many individuals. I would like to express my gratitude to my advisors, Gary Cohen and Roselyn Eisenberg, for allowing me to join the laboratory and for the tremendous support and guidance they provided in all aspects of this work. Their continuous enthusiasm and dedication is inspiring. Also, I need to thank Michael Atchison for his personal support and his commitment to the VMD/PhD program. I must also thank the members of my thesis committee Paul Bates, Jeff Bergelson, Harvey Freidman, and Yan Yuan for helping me throughout this process. My thanks to Sara Cherry and the members of the Cherry laboratory where I learned a great deal. I also thank the fantastic group of people that makes up the Cohen/Eisenberg laboratory. A big thanks to the entire lab past and present, including Doina Atanasiu, Chwan Hong Foo, John Gallagher, Huan Lou, Manuel Ponce-de-Leon, Wan Ting Saw, Katie Stiles, and Chuck Whitbeck. I’d especially like to thank Tina Cairns who helped with many aspects of this project, and made completion of this work possible. I’m incredibly grateful for my wonderful wife and daughter, and their constant support and encouragement. Lastly, I would like to thank my parents for their unconditional support. iii ABSTRACT DEFINING A REGULATORY ROLE FOR THE HSV GLYCOPROTEIN B MEMBRANE PROXIMAL REGION IN MEMBRANE ASSOCIATION Spencer S. Shelly Roselyn J. Eisenberg Gary H. Cohen Herpes simplex virus (HSV) entry requires four essential glycoproteins (gD, gH/gL, and gB) to enable fusion between the virion envelope and the cellular membrane. The fusion cascade is activated by gD binding to one of its receptors, nectin-1 or HVEM. Glycoprotein B (gB), a class III viral fusion protein, mediates the fusion reaction, while data indicates that gH/gL acts as a regulator of gB. gB is trimeric and has a 773 amino acid ectodomain with a highly hydrophobic membrane proximal region (MPR) (residues 731-773) and two fusion loops (FL) per protomer. The post-fusion structure of gB was solved from the gB(730t) construct, which is truncated to remove the hydrophobic MPR residues. In this dissertation I investigated the MPRs influence on gBs ability to interact with membranes. I hypothesize that the MPR regulates fusion loop exposure by interacting with the fusion loops and masks them until fusion begins. To investigate this process I constructed a series of MPR deletion, truncation, and point mutations using iv both full-length mammalian expression vectors and purified baculovirus expressed protein. I found that deletions in the MPR from full-length gB resulted in a disruption in cell surface expression in transfected cells. This suggests the MPR is necessary for proper folding or transport of gB. Soluble gB MPR truncations [gB(759t), gB(749t), gB(739t)] were expressed and purified using the baculovirus expression system, and compared to MPR-less gB(730t) and full MPR containing gB(773t). I found that gB containing an MPR segment were all compromised in their ability to bind liposomes in comparison to gB(730t), which lacks any MPR residues. Supporting our hypothesis we found that residues 731 to 739 were sufficient prevent liposome association and mutation of two aromatic residues, F732 and F739, to alanine in gB(739t) restored gBs ability to bind liposomes. Together, my data suggests the MPR does indeed regulate gBs ability to associate with liposomes, and that aromatic residues in the MPR are important for this function. This supports our model that the MPR masks the gB FLs to prevent premature membrane association and adds another layer of regulation to the HSV entry cascade. v TABLE OF CONTENTS Page DEDICATION ............................................................................................................... ii ACKNOWLEDGEMENTS ......................................................................................... iii ABSTRACT ................................................................................................................... iv TABLE OF CONTENTS ............................................................................................. vi LIST OF TABLES ...................................................................................................... viii LIST OF ILLUSTRATIONS ....................................................................................... ix CHAPTER 1: GENERAL INTRODUCTION ............................................................1 A. Herpesvirus overview ...........................................................................................1 B. Virion structure .....................................................................................................4 C. HSV replication cycle ...........................................................................................5 D. Viral entry strategies .............................................................................................9 E. Viral fusion proteins ............................................................................................11 a. Class I .......................................................................................................12
Recommended publications
  • Virus Entry: What Has Ph Got to Do with It?
    TURNING POINTS Virus entry: What has pH got to do with it? Ari Helenius After several years of working on membrane For almost two years, the virus entry path- Over the following months, we validated proteins using the Semliki Forest virus (SFV) as way and mechanism of host penetration con- all the predictions of our pH hypothesis. For a model, I decided in 1976 to focus my research tinued to elude us. However, in the spring example, we could demonstrate a membrane on the mechanisms by which animal viruses of 1978, I had the chance to participate in a fusion reaction in vitro by simply mixing such as SFV enter and infect their host cells. Dahlem Conference in Berlin called ‘Transport liposomes and SFV, and briefly dropping the pH I had just moved from Finland with the lab of of Macromolecules in Cellular Systems’. It was to 6 or below. Moreover, when we added acidic Kai Simons to the newly founded European an opportune time for a meeting on this topic: medium to cells, we could ‘fool’ surface-bound Molecular Biology Laboratory (EMBL) in pathways of vesicle transport were being dis- viruses into fusing with the plasma membrane; Heidelberg, Germany. I was eager to tackle a covered, receptor-mediated trafficking pro- the entry block caused by weak bases was challenging problem — and one that did not cesses were beginning to be described, and the bypassed and the cells became infected. require work with detergents. important role of clathrin-coated vesicles was Joined by Judy White, Mark Marsh and Relying almost exclusively on electron finally properly appreciated.
    [Show full text]
  • Formation of Influenza Virus Particles Lacking Hemagglutinin on the Viral Envelope Asit K
    University of Nebraska - Lincoln DigitalCommons@University of Nebraska - Lincoln Papers in Veterinary and Biomedical Science Veterinary and Biomedical Sciences, Department of 1986 Formation of Influenza Virus Particles Lacking Hemagglutinin on the Viral Envelope Asit K. Pattnaik University of Nebraska-Lincoln, [email protected] Donald J. Brown University of California at Los Angeles Debi P. Nayak University of California at Los Angeles Follow this and additional works at: http://digitalcommons.unl.edu/vetscipapers Part of the Biochemistry, Biophysics, and Structural Biology Commons, Cell and Developmental Biology Commons, Immunology and Infectious Disease Commons, Medical Sciences Commons, Veterinary Microbiology and Immunobiology Commons, and the Veterinary Pathology and Pathobiology Commons Pattnaik, Asit K.; Brown, Donald J.; and Nayak, Debi P., "Formation of Influenza Virus Particles Lacking Hemagglutinin on the Viral Envelope" (1986). Papers in Veterinary and Biomedical Science. 198. http://digitalcommons.unl.edu/vetscipapers/198 This Article is brought to you for free and open access by the Veterinary and Biomedical Sciences, Department of at DigitalCommons@University of Nebraska - Lincoln. It has been accepted for inclusion in Papers in Veterinary and Biomedical Science by an authorized administrator of DigitalCommons@University of Nebraska - Lincoln. JOURNAL OF VIROLOGY, Dec. 1986, p. 994-1001 Vol. 60, No.3 0022-S38X1861120994-08 $02.00/0 Copyright © 1986, American Society for Microbiology Formation of Influenza Virus Particles
    [Show full text]
  • How Influenza Virus Uses Host Cell Pathways During Uncoating
    cells Review How Influenza Virus Uses Host Cell Pathways during Uncoating Etori Aguiar Moreira 1 , Yohei Yamauchi 2 and Patrick Matthias 1,3,* 1 Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; [email protected] 2 Faculty of Life Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK; [email protected] 3 Faculty of Sciences, University of Basel, 4031 Basel, Switzerland * Correspondence: [email protected] Abstract: Influenza is a zoonotic respiratory disease of major public health interest due to its pan- demic potential, and a threat to animals and the human population. The influenza A virus genome consists of eight single-stranded RNA segments sequestered within a protein capsid and a lipid bilayer envelope. During host cell entry, cellular cues contribute to viral conformational changes that promote critical events such as fusion with late endosomes, capsid uncoating and viral genome release into the cytosol. In this focused review, we concisely describe the virus infection cycle and highlight the recent findings of host cell pathways and cytosolic proteins that assist influenza uncoating during host cell entry. Keywords: influenza; capsid uncoating; HDAC6; ubiquitin; EPS8; TNPO1; pandemic; M1; virus– host interaction Citation: Moreira, E.A.; Yamauchi, Y.; Matthias, P. How Influenza Virus Uses Host Cell Pathways during 1. Introduction Uncoating. Cells 2021, 10, 1722. Viruses are microscopic parasites that, unable to self-replicate, subvert a host cell https://doi.org/10.3390/ for their replication and propagation. Despite their apparent simplicity, they can cause cells10071722 severe diseases and even pose pandemic threats [1–3].
    [Show full text]
  • Viral Envelope Are Controlled by the Helper Virus Used to Activate The
    DETERMINING FACTOR IN THE CAPACITY OF ROUS SARCOMA VIRUS TO INDUCE TUMORS IN MAMMALS* By HIDESABURO HANAFUSA AND TERUKO HANAFUSA COLLMGE DE FRANCE, LABORATOIRE DE MIDECINE EXPARIMENTALE, PARIS Communidated by Ahdre Lwoff, January 24, 1966 Since Zilber and Kriukoval and Svet-Moldavsky' demonstrated that some strains of Rous sarcoma virus (RSV) are capable of inducing hemorrhagic cysts or sarcomas in newborn rats, numerous attempts have been made to induce tumors by RSVin various species of mammals.3-9 One of the remarkable aspects revealed by these investigations is the strain differences in RSV for this capacity.6-9 Certain strains, such as the Schmidt-Ruppin strain, are active, while others, such as the Bryan strain, are generally inactive in mammals. The cause of such strain differ- ences has not been elucidated. Our previous studies have shown that the Bryan high-titer strain of RSV is a defective virus which cannot reproduce without the help of any one of the avian leukosis viruses.'0 The defectiveness derives from the inability of this strain of RSV to induce the synthesis of its own viral envelope."I An essential role played by the helper virus is to provide the envelope to the RSV genome, whose replica- tion occurs in the infected cells without the aid of helper virus. Thus, several properties of RSV which presumably depend in some way on the character of the viral envelope are controlled by the helper virus used to activate the RSV.11-13 These properties include the antigenicity, the sensitivity to the interference induced by the helper virus, and the host range among genetically different chick embryos.
    [Show full text]
  • Structure and Dynamics of the SARS-Cov-2 Envelope Protein Monomer
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.10.434722; this version posted March 10, 2021. 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-ND 4.0 International license. Structure and dynamics of the SARS-CoV-2 envelope protein monomer Alexander Kuzmin1, Philipp Orekhov1,2, Roman Astashkin1,3, Valentin Gordeliy1,3,4,5, Ivan Gushchin1,* 1 Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia 2 Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia 3 Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, France 4 Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, Jülich, Germany 5 JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich GmbH, Jülich, Germany. E-mail for correspondence: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2021.03.10.434722; this version posted March 10, 2021. 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-ND 4.0 International license. Abstract Coronaviruses, especially SARS-CoV-2, present an ongoing threat for human wellbeing. Consequently, elucidation of molecular determinants of their function and interaction with host is an important task. Whereas some of the coronaviral proteins are extensively characterized, others remain understudied.
    [Show full text]
  • A Loop Region in the N-Terminal Domain of Ebola Virus VP40 Is Important in Viral Assembly, Budding, and Egress
    Viruses 2014, 6, 3837-3854; doi:10.3390/v6103837 OPEN ACCESS viruses ISSN 1999-4915 www.mdpi.com/journal/viruses Article A Loop Region in the N-Terminal Domain of Ebola Virus VP40 Is Important in Viral Assembly, Budding, and Egress Emmanuel Adu-Gyamfi 1,†,‡, Smita P. Soni 2,‡, Clara S. Jee 1, Michelle A. Digman 3,4, Enrico Gratton 3 and Robert V. Stahelin 1,2,* 1 Department of Chemistry and Biochemistry, the Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA; E-Mails: [email protected] (E.A.-G.); [email protected] (C.S.J.) 2 Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, USA; E-Mail: [email protected] 3 Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA; E-Mails: [email protected] (M.A.D.); [email protected] (E.G.) 4 Centre for Bioactive Discovery in Health and Ageing, School of Science and Technology, University of New England, Armidale NSW 2351, Australia † Current address: Department of Molecular Biosciences/HHMI, Northwestern University, Evanston, IL 60208, USA. ‡ These authors contributed equally to this work. * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +1-574-631-5054; Fax: +1-574-631-7821. External Editor: Jens H. Kuhn Received: 1 July 2014; in revised form: 9 October 2014 / Accepted: 11 October 2014 / Published: 17 October 2014 Abstract: Ebola virus (EBOV) causes viral hemorrhagic fever in humans and can have clinical fatality rates of ~60%. The EBOV genome consists of negative sense RNA that encodes seven proteins including viral protein 40 (VP40).
    [Show full text]
  • The SARS-Coronavirus Infection Cycle: a Survey of Viral Membrane Proteins, Their Functional Interactions and Pathogenesis
    International Journal of Molecular Sciences Review The SARS-Coronavirus Infection Cycle: A Survey of Viral Membrane Proteins, Their Functional Interactions and Pathogenesis Nicholas A. Wong * and Milton H. Saier, Jr. * Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116, USA * Correspondence: [email protected] (N.A.W.); [email protected] (M.H.S.J.); Tel.: +1-650-763-6784 (N.A.W.); +1-858-534-4084 (M.H.S.J.) Abstract: Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a novel epidemic strain of Betacoronavirus that is responsible for the current viral pandemic, coronavirus disease 2019 (COVID- 19), a global health crisis. Other epidemic Betacoronaviruses include the 2003 SARS-CoV-1 and the 2009 Middle East Respiratory Syndrome Coronavirus (MERS-CoV), the genomes of which, particularly that of SARS-CoV-1, are similar to that of the 2019 SARS-CoV-2. In this extensive review, we document the most recent information on Coronavirus proteins, with emphasis on the membrane proteins in the Coronaviridae family. We include information on their structures, functions, and participation in pathogenesis. While the shared proteins among the different coronaviruses may vary in structure and function, they all seem to be multifunctional, a common theme interconnecting these viruses. Many transmembrane proteins encoded within the SARS-CoV-2 genome play important roles in the infection cycle while others have functions yet to be understood. We compare the various structural and nonstructural proteins within the Coronaviridae family to elucidate potential overlaps Citation: Wong, N.A.; Saier, M.H., Jr.
    [Show full text]
  • Envelope and Host Cell Plasma Membranes (H9 Plasma Membranes) ROLAND C
    Proc. Natl. Acad. Sci. USA Vol. 90, pp. 5181-5185, June 1993 Medical Sciences Lipid composition and fluidity of the human immunodeficiency virus envelope and host cell plasma membranes (H9 plasma membranes) ROLAND C. ALOIA*tt§, HuIROu TIANt, AND FRED C. JENSEN1 *Anesthesia Service, J. L. Pettis Veterans Administration Hospital, Loma Linda, CA 92357; Departments of tAnesthesiology and tBiochemistry, Loma Linda University School of Medicine, Loma Linda, CA 92350; and lImmuneResponse Corporation, Carlsbad, CA 92008 Communicated by Robert C. Gallo, January 4, 1993 (receivedfor review August 11, 1992) ABSTRACT Previous studies have indicated that human IIIRF, Sph is 11-13 mol % (6). It is presently unclear what immunodeficiency virus (HIV) is enclosed with a lipid envelope physical mechanism(s) account for these lipid variations. similar in composition to cell plasma membranes and to other They could reflect variations in cell lines and growth condi- viruses. Further, the fluidity, as measured by spin resonance tions or genetic differences in HIV isolates, in which enve- spectroscopy, is low and the viral envelope is among the most lope or other proteins select different lipids from the bulk highly ordered membranes analyzed. However, the relation- lipid pool within the cell surface membrane during the ship between viral envelope lipids and those of the host cell is budding process (5). Lipid envelopes ofother viruses possess not known. Here we demonstrate that the phospholipids within a lipid profile different from their host surface membranes, as the envelopes ofHIV-1RF and HIV-2-L are similar to each other has been shown for Semliki Forest virus (12), simian virus 5 but significantly different from their respective host cell surface (13, 14), respiratory syncytial virus (15), Sindbis virus (16), membranes.
    [Show full text]
  • Balancing the Influenza Neuraminidase and Hemagglutinin Responses by Exchanging the Vaccine Virus Backbone
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.08.416099; this version posted December 8, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. Balancing the influenza neuraminidase and hemagglutinin responses by exchanging the vaccine virus backbone Jin Gao, Hongquan Wan, Xing Li, Mira Rakic Martinez, Yamei Gao, Zhiping Ye and Robert Daniels* Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA *Correspondence: [email protected] Running Title: Balancing the NA and HA antigen response Abstract Virions are a common antigen source for many viral vaccines. One limitation to using virions is that the antigen abundance is determined by the content of each protein in the virus. This caveat especially applies to viral-based influenza vaccines where the low abundance of the neuraminidase (NA) surface antigen remains a bottleneck for improving the NA antibody response. Here, we used the WHO recommended H1N1 antigens (2019-2020 season) in a systematic analysis to demonstrate that the NA to hemagglutinin (HA) ratio in virions can be improved by exchanging the viral backbone internal genes, thereby preserving the antigens. The purified inactivated virions with higher NA content show a more spherical morphology, a different balance between the HA receptor binding and NA receptor release functions, and induce a better NA inhibitory antibody response in mice.
    [Show full text]
  • The Role of Viral Glycoproteins and Tegument Proteins in Herpes
    Louisiana State University LSU Digital Commons LSU Doctoral Dissertations Graduate School 2014 The Role of Viral Glycoproteins and Tegument Proteins in Herpes Simplex Virus Type 1 Cytoplasmic Virion Envelopment Dmitry Vladimirovich Chouljenko Louisiana State University and Agricultural and Mechanical College Follow this and additional works at: https://digitalcommons.lsu.edu/gradschool_dissertations Part of the Veterinary Pathology and Pathobiology Commons Recommended Citation Chouljenko, Dmitry Vladimirovich, "The Role of Viral Glycoproteins and Tegument Proteins in Herpes Simplex Virus Type 1 Cytoplasmic Virion Envelopment" (2014). LSU Doctoral Dissertations. 4076. https://digitalcommons.lsu.edu/gradschool_dissertations/4076 This Dissertation is brought to you for free and open access by the Graduate School at LSU Digital Commons. It has been accepted for inclusion in LSU Doctoral Dissertations by an authorized graduate school editor of LSU Digital Commons. For more information, please [email protected]. THE ROLE OF VIRAL GLYCOPROTEINS AND TEGUMENT PROTEINS IN HERPES SIMPLEX VIRUS TYPE 1 CYTOPLASMIC VIRION ENVELOPMENT A Dissertation Submitted to the Graduate Faculty of the Louisiana State University and Agricultural and Mechanical College in partial fulfillment of the requirements for the degree of Doctor of Philosophy in The Interdepartmental Program in Veterinary Medical Sciences through the Department of Pathobiological Sciences by Dmitry V. Chouljenko B.Sc., Louisiana State University, 2006 August 2014 ACKNOWLEDGMENTS First and foremost, I would like to thank my parents for their unwavering support and for helping to cultivate in me from an early age a curiosity about the natural world that would directly lead to my interest in science. I would like to express my gratitude to all of the current and former members of the Kousoulas laboratory who provided valuable advice and insights during my tenure here, as well as the members of GeneLab for their assistance in DNA sequencing.
    [Show full text]
  • 7094.Full.Pdf
    The Journal of Immunology Human Cytomegalovirus Envelope Glycoproteins B and H Are Necessary for TLR2 Activation in Permissive Cells1 Karl W. Boehme,* Mario Guerrero,*† and Teresa Compton2*† Human CMV (HCMV) is a ubiquitous member of the Herpesviridae family and an opportunistic pathogen that poses significant health risks for immunocompromised patients. HCMV pathogenesis is intimately tied to the immune status of the host, thus characterization of the innate immune response to HCMV infection is critical for understanding disease progression. Previously, we identified TLR2 as a host factor that detects and initiates inflammatory cytokine secretion in response to HCMV independent of viral replication. In this study, we show that two entry-mediating envelope gp, gp B (gB) and gp H (gH), display determinants recognized by TLR2. Neutralizing Abs against TLR2, gB and gH inhibit inflammatory cytokine responses to HCMV infection, suggesting that inflammatory cytokine stimulation by HCMV is mediated by interactions between these envelope gp and TLR2. Furthermore, both gB and gH coimmunoprecipitate with TLR2 and TLR1, indicating that these envelope gp directly interact with TLR2 and that a TLR2/TLR1 heterodimer is a functional sensor for HCMV. Because our previous studies were conducted in model cell lines, we also show that TLR2 is expressed by HCMV permissive human fibroblast cell strains, and that TLR2 is a functional sensor in these cells. This study further elucidates the importance and potency of envelope gp as a class of molecules displaying pathogen-associated molecular patterns that are recognized with immediate kinetics by TLRs in permissive cells. The Journal of Immunology, 2006, 177: 7094–7102.
    [Show full text]
  • Viroporins: Structure, Function and Potential As Antiviral Targets
    This is a repository copy of Viroporins: Structure, function and potential as antiviral targets. White Rose Research Online URL for this paper: http://eprints.whiterose.ac.uk/88837/ Version: Accepted Version Article: Scott, C and Griffin, S orcid.org/0000-0002-7233-5243 (2015) Viroporins: Structure, function and potential as antiviral targets. Journal of General Virology, 96 (8). pp. 2000-2027. ISSN 0022-1317 https://doi.org/10.1099/vir.0.000201 © 2015 The Authors. Published by the Microbiology Society. This is an author produced version of a paper published in Journal of General Virology. Uploaded in accordance with the publisher's self-archiving policy. Reuse Unless indicated otherwise, fulltext items are protected by copyright with all rights reserved. The copyright exception in section 29 of the Copyright, Designs and Patents Act 1988 allows the making of a single copy solely for the purpose of non-commercial research or private study within the limits of fair dealing. The publisher or other rights-holder may allow further reproduction and re-use of this version - refer to the White Rose Research Online record for this item. Where records identify the publisher as the copyright holder, users can verify any specific terms of use on the publisher’s website. Takedown If you consider content in White Rose Research Online to be in breach of UK law, please notify us by emailing [email protected] including the URL of the record and the reason for the withdrawal request. [email protected] https://eprints.whiterose.ac.uk/ Journal of General Virology Viroporins: structure, function and potential as antiviral targets --Manuscript Draft-- Manuscript Number: VIR-D-15-00200R1 Full Title: Viroporins: structure, function and potential as antiviral targets Article Type: Review Section/Category: High Priority Review Corresponding Author: Stephen D.
    [Show full text]