SIVMAC Vpx Improves the Transduction of Dendritic Cells with Nonintegrative HIV-1-Derived Vectors

Total Page:16

File Type:pdf, Size:1020Kb

SIVMAC Vpx Improves the Transduction of Dendritic Cells with Nonintegrative HIV-1-Derived Vectors Gene Therapy (2009) 16, 159–163 & 2009 Macmillan Publishers Limited All rights reserved 0969-7128/09 $32.00 www.nature.com/gt SHORT COMMUNICATION SIVMAC Vpx improves the transduction of dendritic cells with nonintegrative HIV-1-derived vectors G Berger1,2,3, C Goujon1,2,3, J-L Darlix1,2,3 and A Cimarelli1,2,3 1LaboRetro, Department of Human Virology, Ecole Normale Supe´rieure de Lyon, Lyon, France; 2INSERM, U758, Lyon, France and 3University of Lyon, Lyon1, IFR128 BioSciences Lyon-Gerland, Lyon-Biopole, Lyon, France Lentiviral vector (LV)-mediated gene therapy bears an removes a specific restriction to lentiviral infection in these intrinsic risk of insertional mutagenesis following integration cells. Here, we describe that the transduction efficiency of into the host genome. Nonintegrative LVs may offer an DCs with nonintegrative HIV-1 vectors can also be improved alternative avenue at least in nondividing cells where via Vpx-VLPs that promote the accumulation of complete episomal viral DNA persists stably. Owing to their central and episomal viral DNA. In this setting, Vpx increases both role in immune system functions, differentiated dendritic cells the number of transduced cells and the levels of transgene (DCs) offer an interesting cell target for these vectors. We expression. Thus, these results describe a simple procedure have previously described that the transduction of DCs with by which transduction of differentiated DCs can be achieved wild-type HIV-1-derived vectors can be considerably im- at low viral inputs with safer LVs to improve both the number proved by providing DCs with noninfectious virion-like of transduced cells and the levels of transgene expression. particles (VLPs) carrying Vpx (Vpx-VLPs), a nonstructural Gene Therapy (2009) 16, 159–163; doi:10.1038/gt.2008.128; protein coded by members of the SIVSM/HIV-2 lineage that published online 31 July 2008 Keywords: nonintegrative; HIV-1 lentiviral vectors; DCs; Vpx One of the major interests of lentiviral vectors (LVs) in susceptible to lentiviral infection.11,12 We have previously gene therapy is their ability to provide long-lasting reported that members of the SIVSM/HIV-2 lineage code transgene expression following integration into the host for a protein, Vpx, are capable of relieving the restriction genome. This very property may lead to severe con- that is responsible for the poor susceptibility of DCs to sequences on the cell’s physiology owing to viral integra- lentiviral-mediated modification.13,14 Indeed, Vpx aug- tion into genes that deregulate the normal cellular ments the efficacy of infection of DCs with integration- transcriptional program.1 Given the propensity of lentivir- competent HIV-1 vectors when it is added in trans onto al integrases (IN) for active transcriptional units,2 DCs via noninfectious virion-like particles (VLPs) de- this represents a non-negligible issue. To circumvent rived from SIVMAC (Vpx-VLPs). In this setting, Vpx this problem, several groups have evaluated the use of promotes the accumulation of viral DNA. Thus, we integration-incompetent LVs.3–8 Gene expression from studied whether similar effects could be observed these vectors is driven from viral DNA molecules that following modification of DCs with nonintegrative remain in an episomal form after their entry into the HIV-1-based LV (as depicted in Figure 1a). nucleus as 1 or 2 long terminal repeat circles (1- or 2-LTRs, Viral integrases (IN) exert several functions during the respectively). These episomes are considered as a dead- viral life cycle, ranging from viral particle formation to end product in the normal retroviral infection process, but reverse transcription, and possibly nuclear import. Thus, they are a functional substrate for the transcriptional IN mutants have been divided into two classes according machinery.9 As they lack an origin of replication, these to the specific or pleiotropic defects they impose on forms are transient in dividing cells, but remain stably mutant viruses (class I or II, respectively18). Mutants in present as episomes in the nucleus of nondividing cells. the catalytic site of HIV-1 IN belong to the former. We Dendritic cells (DCs) are an interesting cell type for first constructed an integration defective HIV-1 LV by gene therapy purposes in light of their master role in the introducing a single point mutation in the catalytic site of regulation of immune system responses.10 Although viral the IN (IN-D116A). This mutation did not impair viral infection of DCs can be obtained with lentiviral-derived production and yielded similar infectious titers as vectors, the efficacy of this modification is generally low compared to wild-type (WT) on HeLa cells, if transduced unless high viral inputs are used, because DCs are poorly cells were examined 3 days after infection. However, a sharp loss of green fluorescent protein (GFP)-expressing cells was specifically observed in cells transduced with Correspondence: Dr A Cimarelli, LaboRetro, Department of Human the IN-D116A mutant when cells were analyzed 11 days Virology, ENS-Lyon INSERM, U758, 46 Alle´e d’Italie, 69364 Lyon, after infection (Figure 1b). This result is not surprising France. E-mail: [email protected] for dividing cells transduced with a nonintegrative LV, as Received 6 May 2008; revised 23 June 2008; accepted 7 July 2008; episomal DNA is expected to dilute over cell divisions. published online 31 July 2008 Indeed, when the amount of integrated viral DNA was Vpx in the transduction of DCs by nonintegrative HIV-1 LVs G Berger et al 160 Figure 1 Schematic representation of the lentiviral vectors (LVs) used in the study and characterization of a nonintegrative LV. (a) Integration-deficient LVs were engineered by introducing a single point mutation in the catalytic site of integrase (IN-D116A) in the context of an HIV-1-based packaging construct (8.2).15 LVs were obtained after transfection of 293 T cells with the packaging construct coding gag-pro-pol and all the accessory proteins (Tat, Rev, Vif, Vpr and Nef; not marked for clarity), a miniviral genome bearing a CMV-GFP expression cassette and a construct coding the vesicular stomatitis virus G envelope protein, as described.15 Noninfectious virion-like particles (referred to as Vpx-VLPs) were similarly produced by transfection of an SIVMAC-based construct coding gag-pro-pol and viral accessory proteins (SIV3+, Tat, Rev, Vif, Vpx, Vpr and Nef16). No viral genome was transfected in this case. We have previously documented that Vpx is the protein responsible for the positive effect of VLPs on the infectivity of incoming HIV-1 LVs.13 Virions were purified by ultracentrifugation on a double step sucrose cushion (45–25%) and normalized by protein content against standards of known infectivity, or by their infectious titers on HeLa cells, as previously described.17 Infections were carried out on 105 cells for 2 h prior to extensive cell washing. The percentage of transduced GFP-positive cells was determined 3 days after infection by flow cytometry. When indicated, Vpx- VLPs were provided together with GFP-coding vectors at a multiplicity of infection (MOI) of 2. (b) HeLa cells were transduced with normalized amounts of wild-type (WT) and IN-D116A LVs and analyzed by flow cytometry 3 and 11 days after infection. Control infections were performed in the presence of the reverse-transcriptase inhibitor azidothymidine (AZT) at 10 mg/ml. (c) The presence of integrated provirus was analyzed on transduced cells infected at MOI 10 by Alu-PCR, as described previously.9 Briefly, the first round of PCR was carried out for 20 cycles with a primer specific for the proviral DNA and a second round specific for Alu sequences in the genome. Fivefold dilutions of the first PCR were then used as the template for a second semiquantitative PCR using two internal primers specific for viral DNA. Amplified products were transferred onto a nylon membrane and hybridized with 32P-labeled specific probes prior to Phosphorimager quantification. A representative gel is shown here together with a graph presenting data obtained from three independent experiments. CMV, cPPT, central polypurine tract; GFP, green fluorescent protein; p.i., post-infection; RRE, Rev-responsive element; *, self-inactivating. determined by Alu-PCR, none was observed in cells effect was observed on stimulated PBLs. Of note, transduced with the IN-D116A mutant (Figure 1c). transduction with the IN-D116A mutant virus yielded a Next, the ability of normalized WT and IN-D116A substantially lower percentage of transduced cells than mutant LVs was analyzed on primary DCs, macrophages WT, even if analysis was carried out 3 days post- and phytohemagglutinin-stimulated primary blood lym- infection, that is, prior to substantial dilution of episomal phocytes (PBLs) (Figure 2a). As we had previously DNA following cell division. We believe that this result reported, Vpx-VLPs increased by over 10-fold the can be due to lower transcriptional activity from the percentage of GFP-positive DCs following infection with episomal DNA in PBLs as opposed to the integrated WT HIV-1 vectors.13 When compared to WT, similar forms. In addition to their positive effect on the amounts of GFP-expressing cells were also obtained percentage of HIV-1-LVs transduced cells, Vpx-VLPs following transduction of DCs with the IN-D116A also increased the median fluorescence intensity of GFP- mutant. Similar to WT, the addition of Vpx-VLPs during expressing cells (MFI; Figure 2b), suggesting that in cells IN-D116A LV infection dramatically increased the per- in which they exert a positive effect on viral infection, centage of transduced cells at all viral inputs tested (by Vpx-VLPs also act positively on the levels of transgene 10- to 20-fold). Accordingly, the percentage of macro- expressed intracellularly. Modified DCs could be kept in phages transduced with the IN-D116A mutant in the culture for over a month, retaining stable expression of presence of Vpx-VLPs increased, although to a lower the transgene (Figure 2c).
Recommended publications
  • THE ROLE of the HERPES SIMPLEX VIRUS TYPE 1 UL28 PROTEIN in TERMINASE COMPLEX ASSEMBLY and FUNCTION by Jason Don Heming Bachelor
    THE ROLE OF THE HERPES SIMPLEX VIRUS TYPE 1 UL28 PROTEIN IN TERMINASE COMPLEX ASSEMBLY AND FUNCTION by Jason Don Heming Bachelor of Science, Clarion University of Pennsylvania, 2004 Submitted to the Graduate Faculty of the School of Medicine in partial fulfillment of the requirements for the degree of Doctor of Philosophy University of Pittsburgh 2013 UNIVERSITY OF PITTSBURGH SCHOOL OF MEDICINE This dissertation was presented by Jason Don Heming It was defended on April 18, 2013 and approved by Michael Cascio, Associate Professor, Bayer School of Natural and Environmental Sciences James Conway, Associate Professor, Department of Structural Biology Neal DeLuca, Professor, Department of Microbiology and Molecular Genetics Saleem Khan, Professor, Department of Microbiology and Molecular Genetics Dissertation Advisor: Fred Homa, Associate Professor, Department of Microbiology and Molecular Genetics ii Copyright © by Jason Don Heming 2013 iii THE ROLE OF THE HERPES SIMPLEX VIRUS TYPE 1 UL28 PROTEIN IN TERMINASE COMPLEX ASSEMBLY AND FUNCTION Jason Don Heming, PhD University of Pittsburgh, 2013 Herpes simplex virus type I (HSV-1) is the causative agent of several pathologies ranging in severity from the common cold sore to life-threatening encephalitic infection. During productive lytic infection, over 80 viral proteins are expressed in a highly regulated manner, resulting in the replication of viral genomes and assembly of progeny virions. Cleavage and packaging of replicated, concatemeric viral DNA into newly assembled capsids is critical to virus proliferation and requires seven viral genes: UL6, UL15, UL17, UL25, UL28, UL32, and UL33. Analogy with the well-characterized cleavage and packaging systems of double-stranded DNA bacteriophage suggests that HSV-1 encodes for a viral terminase complex to perform these essential functions, and several studies have indicated that this complex consists of the viral UL15, UL28, and UL33 proteins.
    [Show full text]
  • HIV Accessory Proteins: Review Leading Roles for the Supporting Cast
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector Cell, Vol. 82, 189-192, July 28, 1995, Copyright © 1995 by Cell Press HIV Accessory Proteins: Review Leading Roles for the Supporting Cast Didier Trono ment into coated pits. The lymphoid-specific p56~k tyrosine Infectious Disease Laboratory kinase bound to the CD4 cytoplasmic tail plays such an The Salk Institute for Biological Studies inhibitory role. While Lck is released by Nef, this seems 10010 North Torrey Pines Road to be an epiphenomenon, because Nef-induced CD4 La Jolla, California 92037 down-modulation is observed in cells that do not express Lck and on truncated CD4 molecules that do not bind the kinase. In yet another model that would be unprecedented, Considering that a set of three genes is sufficient for the Nef could physically connect CD4 with a component of replication of most retroviruses, the human and simian the internalization pathway. Future experiments should immunodeficiency viruses (HIV and SIV) exhibit a surpris- test these various possibilities, in particular by determining ing degree of genomic complexity. HIV-1 contains no less conclusively whether Nef, directly or indirectly, interacts than six open reading frames in addition to the prototypic with CD4 or with the endocytic apparatus. Irrespective of gag, pol, and env coding sequences (see Figure 1). Two its mechanism, what' advantage does Nef-mediated CD4 of these supplementary genes, tat and rev, are absolutely down-regulation confer to the virus? Does it rapidly block necessary for virus growth: Tat is a major transactivator detrimental superinfection events? Does it prevent virus of the proviral long terminal repeat (LTR), and Rev acts aggregation on the surface of cells during budding, analo- posttranscriptionally to ensure the switch from the early gous to the role played by neuraminidase for influenza to the late phase of viral gene expression.
    [Show full text]
  • The Impact of Herpes Therapy on Genital and Systemic Immunology
    The impact of herpes therapy on genital and systemic immunology by Tae Joon Yi A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Immunology University of Toronto © Copyright by Tae Joon Yi 2013 The impact of herpes therapy on genital and systemic immunology Tae Joon Yi Doctor of Philosophy Department of Immunology University of Toronto 2013 Abstract HIV infects more than 34 million people globally. Herpes simplex virus type 2 (HSV-2) has been associated with a 3-fold increase in the rate of HIV acquisition, which may be due to physical breaks in the mucosal barrier during symptomatic episodes and/or an increased number of HIV target cells being exposed in the genital tract. Although clinical trials have demonstrated that herpes suppression in HIV uninfected individuals had no impact on HIV incidence, acyclovir was associated with a decrease in plasma HIV viral load and delayed disease progression in HSV-2 co-infected individuals. In addition, many HIV infected individuals display persistent systemic immune activation, which correlates with disease progression, despite successful antiretroviral therapy (ART). It is hypothesized that HSV could be one of the drivers of this activation. To further assess these relationships, my thesis focused on examining the impact of herpes therapy on genital tract immunology and systemic immune activation in HIV uninfected women and a comparison of systemic immune activation in individuals co-infected with HIV and HSV-2 on ART randomized to valacyclovir or placebo. A randomized, placebo-controlled, double-blinded, cross-over trial was conducted in HSV-2 infected, HIV uninfected, women using valacyclovir.
    [Show full text]
  • Acquired Immunodeficiency Syndrome: See AIDS ADCC: See Antibody
    Index Acquired immunodeficiency syndrome: see AIDS Antibodies (cont.) ADCC: see Antibody-mediated cytotoxicity to hemagglutinin, 166 Adenoviruses, 3; see also specific types HIV, 33, 58. 64 Afriamycin, 58 immunoglobulin G, 329 AIDS, 2, 55-56, 76, 95, 257; see also HIV influenza virus neuraminidase and, 229 antigenic characteristics of, 55 monoclonal: see Monoclonal antibodies anxiety about, 57, 58 neuraminidase inhibiting, 184 control of, 57-58 neutralizing: see Neutralizing antibodies defined, 36 PIV3-induced, 144 demographic characteristics of, 40-41 preformed, 94, 95 diagnosis of, 36 protective, 64 epidemiology of in U.S., 36-44 rhinovirus, 231 evolution and, 27 rotavirus. 282 genetic basis of, 78 serotype-specific, 283 geographical distribution of, 42-43 to thymosin alpha I, 56 incidence of, 36 virion interaction with, 236 management of, 57-58 virus-specific, 66 molecular characteristics of, 55 Antibody-complement-mediated lysis, 70 molecular virology of, 56-57 Antibody-mediated cytotoxicity (ADCC). 103 mortality from, 36, 58 Antibody-mediated immunity, 113 neuropathologic lesions in, 100 Antibody-producing B cells. 284 new therapeutic approaches to, 58-59 Antigenic heterogeneity nonstructural-regulatory genes in, 82 of influenza A, 137-138 opportunistic diseases associated with, 41-44 of influenza B, 137 patient exposure groups for, 37-40 Antigenicity, 84 perinatal transmission of. 45 of FMDV, 247-248 prevention of. 58 of rhinoviruses, 226 progression of disease in, 108 Antigenic shift, 4 transmission of, 44-46, 55 Antigenic variations, 2, 20 treatment of, 58-59 of F glycoprotein, 154-155 vaccines for, 6. 59 of FMDV, 247-255 AIDS-related complex (ARC), 55 of influenza virus, 69 AIDS-related lymphadenopathy, 55 of lentiviruses, 66-69 AL V: see Avian leukosis viruses (AL V) of NDV.
    [Show full text]
  • An Investigation of the Mechanisms Underlying HIV-1-Mediated Inflammasome Activation
    An investigation of the mechanisms underlying HIV-1-mediated inflammasome activation Christopher JK Ward Doctorate of Philosophy in Medicine Supervised by Dr. Martha Triantafilou & Prof. Kathy Triantafilou Submitted July 2017 i DECLARATION This work has not been submitted in substance for any other degree or award at this or any other university or place of learning, nor is being submitted concurrently in candidature for any degree or other award. Signed ……………………………………………………… (candidate) Date 01.07.2017 STATEMENT 1 This thesis is being submitted in partial fulfilment of the requirements for the degree of Doctorate of Philosophy (Medicine) Signed ………………………………………….…………… (candidate) Date 01.07.2017 STATEMENT 2 This thesis is the result of my own independent work/investigation, except where otherwise stated, and the thesis has not been edited by a third party beyond what is permitted by Cardiff University’s Policy on the Use of Third Party Editors by Research Degree Students. Other sources are acknowledged by explicit references. The views expressed are my own. Signed ……………………………………….……….…… (candidate) Date 01.07.2017 STATEMENT 3 I hereby give consent for my thesis, if accepted, to be available online in the University’s Open Access repository and for inter-library loan, and for the title and summary to be made available to outside organisations. Signed ……………………………………………..…..….. (candidate) Date 01.07.2017 ii Table of Contents Table of Contents ...........................................................................................................
    [Show full text]
  • Primate Immunodeficiency Virus Vpx and Vpr Counteract Transcriptional Repression of Proviruses by the HUSH Complex
    bioRxiv preprint doi: https://doi.org/10.1101/293001; this version posted April 3, 2018. 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. Primate immunodeficiency virus Vpx and Vpr counteract transcriptional repression of proviruses by the HUSH complex Leonid Yurkovetskiy1, Mehmet Hakan Guney1, Kyusik Kim1, Shih Lin Goh1, Sean McCauley1, Ann Dauphin1, William Diehl1, and Jeremy LuBan1,2* 1Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA 2Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA *Correspondence: [email protected] (J.L.) bioRxiv preprint doi: https://doi.org/10.1101/293001; this version posted April 3, 2018. 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. 1 Drugs that inhibit HIV-1 replication and prevent progression to AIDS do not 2 eliminate HIV-1 proviruses from the chromosomes of long-lived CD4+ memory T 3 cells. To escape eradication by these antiviral drugs, or by the host immune 4 system, HIV-1 exploits poorly defined host factors that silence provirus 5 transcription. These same factors, though, must be overcome by all retroviruses, 6 including HIV-1 and other primate immunodeficiency viruses, in order to activate 7 provirus transcription and produce new virus.
    [Show full text]
  • A Review of HIV Restriction Factors and Viral Countering Mechanisms
    A review of HIV restriction factors and viral countering mechanisms By Qimeng Gao A thesis submitted to the Johns Hopkins University in conformity with the requirements for the degree of Master of Health Science from the Department of Molecular Microbiology and Immunology. Baltimore, Maryland May, 2014 © 2014 Qimeng Gao All Rights Reserved Abstract Human immune system is powerful and it has evolved over the past thousands of years to protect us from various foreign pathogens. In fact, very few pathogens can threaten a man with a competent immune system. The notorious Human Immunodeficiency Virus may be among those very few pathogens as it attacks human immune system; however, it does not mean men are left utterly weaponless in the face of HIV infection. The adaptive arm of the human immune system has historically received more attention, given that most vaccines to viral pathogens are based on this type of immune response. In the context of HIV infection, however, attempts to induce antibody responses or cell-mediated immunity with vaccine have yielded little success. As a result, research resource has shifted to look at the first-line protection that precedes the adaptive immunity. Restriction factors are among this innate arm of the immune system. Since 2002, many restriction factors have been described, many in the context of their antiretroviral activities. In this thesis essay, I attempted to cover some of the best-studied HIV restriction factors to date, including their potential antiviral mechanisms and how virus has developed ways to circumvent their inhibition effects. Many restriction factors are now on the verge of being translated into clinical products, so I tried to include some of the latest translational applications of restriction factors in this article.
    [Show full text]
  • Hepatitis B Virus–Induced Imbalance of Inflammatory and Antiviral
    Hepatitis B Virus−Induced Imbalance of Inflammatory and Antiviral Signaling by Differential Phosphorylation of STAT1 in Human Monocytes This information is current as of September 25, 2021. Hongxiao Song, Guangyun Tan, Yang Yang, An Cui, Haijun Li, Tianyang Li, Zhihui Wu, Miaomiao Yang, Guoyue Lv, Xiumei Chi, Junqi Niu, Kangshun Zhu, Ian Nicholas Crispe, Lishan Su and Zhengkun Tu J Immunol published online 6 March 2019 Downloaded from http://www.jimmunol.org/content/early/2019/03/05/jimmun ol.1800848 http://www.jimmunol.org/ Supplementary http://www.jimmunol.org/content/suppl/2019/03/05/jimmunol.180084 Material 8.DCSupplemental Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on September 25, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2019 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published March 6, 2019, doi:10.4049/jimmunol.1800848 The Journal of Immunology Hepatitis B Virus–Induced Imbalance of Inflammatory and Antiviral Signaling by Differential Phosphorylation of STAT1 in Human Monocytes Hongxiao Song,* Guangyun Tan,* Yang Yang,* An Cui,* Haijun Li,* Tianyang Li,* Zhihui Wu,* Miaomiao Yang,* Guoyue Lv,† Xiumei Chi,‡ Junqi Niu,‡ Kangshun Zhu,x Ian Nicholas Crispe,*,{ Lishan Su,*,‖ and Zhengkun Tu*,‡ It is not clear how hepatitis B virus (HBV) modulates host immunity during chronic infection.
    [Show full text]
  • Cell Cycle Inhibitory Effects of HIV and SIV Vpr and Vpx in the Yeast Schizosaccharomyces Pombe
    VIROLOGY 230, 103–112 (1997) ARTICLE NO. VY978459 Cell Cycle Inhibitory Effects of HIV and SIV Vpr and Vpx in the Yeast Schizosaccharomyces pombe CHENGSHENG ZHANG, COLIN RASMUSSEN,*,†,‡,1 and LUNG-JI CHANG Department of Medical Microbiology and Immunology, *Department of Anatomy and Cell Biology, †Department of Biochemistry, and ‡Department of Oncology, University of Alberta, Edmonton, Alberta, Canada T6G 2H7 Received November 25, 1996; returned to author for revision December 24, 1996; accepted January 23, 1997 The Vpr gene of human immunodeficiency virus type 1 and type 2 (HIV-1, HIV-2) and simian immunodeficiency virus (SIV) encodes a small nuclear protein which is virion-associated and assists nuclear transport of the preintegration complex. Expression of HIV-1 Vpr has been shown to induce differentiation and prevent proliferation of human cells. HIV-1 Vpr has also been shown to arrest cell growth and cause morphological defects in yeast. In contrast, the Vpx gene of HIV-2 and SIV, which shares sequence homology with Vpr, does not seem to inhibit proliferation of human cells. It has been suggested that the cell cycle arrest effect of Vpr and Vpx is species and cell-type dependent. In this study, we have taken advantage of a conditional expression system to characterize the growth inhibitory effects of Vpr and Vpx of HIV-1, HIV-2, and SIV in the fission yeast Schizosaccharomyces pombe. Our results show that both Vpr and/or Vpx of HIV-1, HIV-2, and SIV arrest cell growth in S. pombe, and HIV-1 Vpr is more cytotoxic than HIV-2 or SIV Vpr or Vpx.
    [Show full text]
  • HIV As the Cause of AIDS
    THE LANCET HIV series HIV as the cause of AIDS Françoise Barré-Sinoussi The two known types of HIV are members of a family of primate lentiviruses. HIV, like other retroviruses, contains a virus capsid, which consists of the major capsid protein, the nucleocapsid protein, the diploid single-stranded RNA genome, and the viral enzymes protease, reverse transcriptase, and integrase. HIV isolates show extensive genetic variability, resulting from the relatively low fidelity of reverse transcriptase in conjunction with the extremely high turnover of virions in vivo. These features of HIVs may have strong implications for vaccine development. Simian immunodeficiency viruses from naturally infected animals differ from HIV in one fundamental respect: they do not cause disease in their natural hosts. Study of these viruses may therefore lead to information about the interaction between lentiviruses and host immune response that could be exploited to combat AIDS. Since the beginning of the AIDS RNA epidemic researchers have made gp120 great efforts to understand the Env gp41 nature of the disease and of its causal agent, the human immuno- PROT deficiency virus (HIV). The two known types of HIV—HIV-1 and INT HIV-21,2—belong to a family of primate lentiviruses whose other members infect African green RT monkeys (SIVagm), sootey mangabey monkeys (SIVsm), p17 mandrills (SIVmnd), sykes (MA) monkeys (SIVsyk), and p24 (CA) Gag chimpanzees (SIVcpz).3 I shall p7 review the structure and molecular p9 features of HIV and the other (NC) primate lentiviruses and their Figure 1: Schematic diagram of an HIV virion and electronmicrograph phylogenetic relationship and Location of the structural proteins (see text) is indicated.
    [Show full text]
  • A Hsp40 Chaperone Protein Interacts with and Modulates the Cellular Distribution of the Primase Protein of Human Cytomegalovirus
    A Hsp40 Chaperone Protein Interacts with and Modulates the Cellular Distribution of the Primase Protein of Human Cytomegalovirus Yonggang Pei1., Wenmin Fu1., Ed Yang2., Ao Shen1,2, Yuan-Chuan Chen2, Hao Gong2, Jun Chen1, Jun Huang1, Gengfu Xiao1, Fenyong Liu1,2* 1 State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China, 2 Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United States of America Abstract Genomic DNA replication is a universal and essential process for all herpesvirus including human cytomegalovirus (HCMV). HCMV UL70 protein, which is believed to encode the primase activity of the viral DNA replication machinery and is highly conserved among herpesviruses, needs to be localized in the nucleus, the site of viral DNA synthesis. No host factors that facilitate the nuclear import of UL70 have been reported. In this study, we provided the first direct evidence that UL70 specifically interacts with a highly conserved and ubiquitously expressed member of the heat shock protein Hsp40/DNAJ family, DNAJB6, which is expressed as two isoforms, a and b, as a result of alternative splicing. The interaction of UL70 with a common region of DNAJB6a and b was identified by both a two hybrid screen in yeast and coimmunoprecipitation in human cells. In transfected cells, UL70 was primarily co-localized with DNAJB6a in the nuclei and with DNAJB6b in the cytoplasm, respectively. The nuclear import of UL70 was increased in cells in which DNAJB6a was up-regulated or DNAJB6b was down-regulated, and was reduced in cells in which DNAJB6a was down-regulated or DNAJB6b was up-regulated.
    [Show full text]
  • Proteomic Approaches to Uncovering Virus–Host Protein Interactions During the Progression of Viral Infection
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Expert Rev Manuscript Author Proteomics. Manuscript Author Author manuscript; available in PMC 2016 June 24. Published in final edited form as: Expert Rev Proteomics. 2016 March ; 13(3): 325–340. doi:10.1586/14789450.2016.1147353. Proteomic approaches to uncovering virus–host protein interactions during the progression of viral infection Krystal K Lum and Ileana M Cristea Department of Molecular Biology, Princeton University, Princeton, NJ, USA Abstract The integration of proteomic methods to virology has facilitated a significant breadth of biological insight into mechanisms of virus replication, antiviral host responses and viral subversion of host defenses. Throughout the course of infection, these cellular mechanisms rely heavily on the formation of temporally and spatially regulated virus–host protein–protein interactions. Reviewed here are proteomic-based approaches that have been used to characterize this dynamic virus–host interplay. Specifically discussed are the contribution of integrative mass spectrometry, antibody- based affinity purification of protein complexes, cross-linking and protein array techniques for elucidating complex networks of virus–host protein associations during infection with a diverse range of RNA and DNA viruses. The benefits and limitations of applying proteomic methods to virology are explored, and the contribution of these approaches to important biological discoveries and to inspiring new tractable avenues for the design of antiviral therapeutics is highlighted. Keywords virus–host interactions; mass spectrometry; viral proteomics; AP-MS; IP-MS; interactome Introduction Viruses are fascinatingly diverse in composition, shape, size, tropism, and pathogenesis. Infectious virus particles can have core capsids that can be structurally helical, while others are icosahedral.
    [Show full text]