1
|
Richardo T, Liu X, Döhner K, Chao TY, Buch A, Binz A, Pohlmann A, de le Roi M, Baumgärtner W, Brand K, Bauerfeind R, Förster R, Sodeik B, Halle S. Herpes simplex virus assembly and spread in murine skin after infection from the outside. J Virol 2025; 99:e0163824. [PMID: 39945537 PMCID: PMC11915863 DOI: 10.1128/jvi.01638-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025] Open
Abstract
Herpes simplex viruses (HSV) cause many skin diseases, particularly in immunocompromised patients. HSV-1 infection of murine skin recapitulates many aspects of human pathology. However, many protocols rely on mechanical or enzymatic skin disruption to induce lesions, although this can alter skin homeostasis and prime antiviral inflammation before inoculation. To investigate the initial events following HSV-1 primary skin infection before the onset of symptoms, we developed a novel murine ex vivo explant model using gentle depilation without further scarification and infected keratinocytes from the outside with minimal tissue damage. Two-photon microscopy showed that HSV-1 spread exclusively in the epidermis. The infection centers increased in number and size over time and contained hundreds of infected keratinocytes. We investigated the HSV-1 spread at the cellular level, using reporter strains with fluorescently tagged capsid protein VP26, and observed the formation of nuclear capsid assembly sites and nuclear capsid egress and the recruitment of the inner tegument protein pUL37GFP, the outer tegument protein VP11/12GFP, and the envelope protein gDGFP to cytoplasmic capsids. By using electron microscopy, the skin appeared intact, and keratinocytes contained many nuclear capsids, primary virions in the nuclear envelope, cytosolic membrane-associated capsids, and enveloped virions. Our protocol provides a robust and reproducible approach to investigate the very early events of HSV-1 spread in the skin, to characterize the phenotypes of HSV-1 mutants in terminally differentiated skin tissues, and to evaluate potentially antiviral small molecules in a preclinical ex vivo infection model. IMPORTANCE This study describes a novel murine ex vivo skin explant model to investigate early events in HSV-1 infection without causing significant tissue damage. To infect from the outside, via the apical keratinocytes, this method relies on gentle depilation, which maintains skin integrity. HSV-1 spread exclusively within the epidermis, with infection centers increasing over time and involving hundreds of keratinocytes. Using advanced microscopy techniques, we tracked HSV-1 spread at the cellular level and intracellular assembly of all intermediate virus structures. This model offers a valuable tool for studying the initial stages of HSV-1 infection, assessing viral mutant phenotypes, and testing antiviral compounds in a more physiological context to provide critical insights into HSV-1 pathogenesis and therapeutic strategies.
Collapse
Affiliation(s)
- Timmy Richardo
- Institute of Virology, Hannover Medical School, Hannover, Germany
- RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Xiaokun Liu
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany
- RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Tsung-Yu Chao
- Institute of Virology, Hannover Medical School, Hannover, Germany
- RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anne Binz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Anja Pohlmann
- Institute of Virology, Hannover Medical School, Hannover, Germany
- RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Madeleine de le Roi
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Korbinian Brand
- Institute of Clinical Chemistry and Laboratory Medicine, Hannover Medical School, Hannover, Germany
| | - Rudolf Bauerfeind
- Research Core Unit Laser Microscopy, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner Site, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner Site, Hannover, Germany
| | - Stephan Halle
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Moschonas GD, Delhaye L, Cooreman R, Hüsers F, Bhat A, Stylianidou Z, De Bousser E, De Pryck L, Grzesik H, De Sutter D, Parthoens E, De Smet AS, Maciejczuk A, Lippens S, Callewaert N, Vandekerckhove L, Debyser Z, Sodeik B, Eyckerman S, Saelens X. MX2 forms nucleoporin-comprising cytoplasmic biomolecular condensates that lure viral capsids. Cell Host Microbe 2024; 32:1705-1724.e14. [PMID: 39389033 DOI: 10.1016/j.chom.2024.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/01/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024]
Abstract
Human myxovirus resistance 2 (MX2) can restrict HIV-1 and herpesviruses at a post-entry step through a process requiring an interaction between MX2 and the viral capsids. The involvement of other host cell factors, however, remains poorly understood. Here, we mapped the proximity interactome of MX2, revealing strong enrichment of phenylalanine-glycine (FG)-rich proteins related to the nuclear pore complex as well as proteins that are part of cytoplasmic ribonucleoprotein granules. MX2 interacted with these proteins to form multiprotein cytoplasmic biomolecular condensates that were essential for its anti-HIV-1 and anti-herpes simplex virus 1 (HSV-1) activity. MX2 condensate formation required the disordered N-terminal region and MX2 dimerization. Incoming HIV-1 and HSV-1 capsids associated with MX2 at these dynamic cytoplasmic biomolecular condensates, preventing nuclear entry of their viral genomes. Thus, MX2 forms cytoplasmic condensates that likely act as nuclear pore decoys, trapping capsids and inducing premature viral genome release to interfere with nuclear targeting of HIV-1 and HSV-1.
Collapse
Affiliation(s)
- George D Moschonas
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Louis Delhaye
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Robin Cooreman
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Franziska Hüsers
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; RESIST-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Anayat Bhat
- Department of Pharmacological and Pharmaceutical Sciences, Laboratory of Molecular Virology and Gene Therapy, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Zoe Stylianidou
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Elien De Bousser
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Laure De Pryck
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Hanna Grzesik
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Delphine De Sutter
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Eef Parthoens
- VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium; VIB BioImaging Core, VIB, 9052 Ghent, Belgium
| | - Anne-Sophie De Smet
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Aleksandra Maciejczuk
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Saskia Lippens
- VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, 9052 Ghent, Belgium; VIB BioImaging Core, VIB, 9052 Ghent, Belgium
| | - Nico Callewaert
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Zeger Debyser
- Department of Pharmacological and Pharmaceutical Sciences, Laboratory of Molecular Virology and Gene Therapy, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; RESIST-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; DZIF-German Centre for Infection Research, Partner site Hannover-Braunschweig, Germany
| | - Sven Eyckerman
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium.
| | - Xavier Saelens
- VIB Center for Medical Biotechnology, VIB, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark-Zwijnaarde 75, 9052 Ghent, Belgium.
| |
Collapse
|
3
|
Cao S, Zhou M, Ji S, Ma D, Zhu S. Recent Advances in the Study of Alphaherpesvirus Latency and Reactivation: Novel Guidance for the Design of Herpesvirus Live Vector Vaccines. Pathogens 2024; 13:779. [PMID: 39338969 PMCID: PMC11435198 DOI: 10.3390/pathogens13090779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Alphaherpesviruses, including herpes simplex virus type 1 (HSV-1), herpes simplex virus type 2 (HSV-2), and varicella-zoster virus (VZV), infect a diverse array of hosts, spanning both humans and animals. Alphaherpesviruses have developed a well-adapted relationship with their hosts through long-term evolution. Some alphaherpesviruses exhibit a typical neurotropic characteristic, which has garnered widespread attention and in-depth research. Virus latency involves the retention of viral genomes without producing infectious viruses. However, under stress, this can be reversed, resulting in lytic infection. Such reactivation events can lead to recurrent infections, manifesting as diseases like herpes labialis, genital herpes, and herpes zoster. Reactivation is a complex process influenced by both viral and host factors, and identifying how latency and reactivation work is vital to developing new antiviral therapies. Recent research highlights a complex interaction among the virus, neurons, and the immune system in regulating alphaherpesvirus latency and reactivation. Neurotropic alphaherpesviruses can breach host barriers to infect neurons, proliferate extensively within their cell bodies, and establish latent infections or spread further. Whether infecting neurons or spreading further, the virus undergoes transmission along axons or dendrites, making this process an indispensable part of the viral life cycle and a critical factor influencing the virus's invasion of the nervous system. Research on the transmission process of neurotropic alphaherpesviruses within neurons can not only deepen our understanding of the virus but can also facilitate the targeted development of corresponding vaccines. This review concentrates on the relationship between the transmission, latency, and activation of alphaherpesviruses within neurons, summarizes recent advancements in the field, and discusses how these findings can inform the design of live virus vaccines for alphaherpesviruses.
Collapse
Affiliation(s)
- Shinuo Cao
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China; (S.C.); (M.Z.)
| | - Mo Zhou
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China; (S.C.); (M.Z.)
| | - Shengwei Ji
- Department of Veterinary Medicine, Agriculture College of Yanbian University, Yanji 133000, China;
| | - Dongxue Ma
- Department of Veterinary Medicine, Agriculture College of Yanbian University, Yanji 133000, China;
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China; (S.C.); (M.Z.)
| |
Collapse
|
4
|
Döhner K, Serrero MC, Viejo-Borbolla A, Sodeik B. A Hitchhiker's Guide Through the Cell: The World According to the Capsids of Alphaherpesviruses. Annu Rev Virol 2024; 11:215-238. [PMID: 38954634 DOI: 10.1146/annurev-virology-100422-022751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The nucleoplasm, the cytosol, the inside of virions, and again the cytosol comprise the world in which the capsids of alphaherpesviruses encounter viral and host proteins that support or limit them in performing their tasks. Here, we review the fascinating conundrum of how specific protein-protein interactions late in alphaherpesvirus infection orchestrate capsid nuclear assembly, nuclear egress, and cytoplasmic envelopment, but target incoming capsids to the nuclear pores in naive cells to inject the viral genomes into the nucleoplasm for viral transcription and replication. Multiple capsid interactions with viral and host proteins have been characterized using viral mutants and assays that reconstitute key stages of the infection cycle. Keratinocytes, fibroblasts, mucosal epithelial cells, neurons, and immune cells employ cell type-specific intrinsic and cytokine-induced resistance mechanisms to restrict several stages of the viral infection cycle. However, concomitantly, alphaherpesviruses have evolved countermeasures to ensure efficient capsid function during infection.
Collapse
Affiliation(s)
- Katinka Döhner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- RESIST Cluster of Excellence, Hannover Medical School, Hannover, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany;
| | - Manutea Christophe Serrero
- Department of Biomedicine and Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
- RESIST Cluster of Excellence, Hannover Medical School, Hannover, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany;
| | - Abel Viejo-Borbolla
- RESIST Cluster of Excellence, Hannover Medical School, Hannover, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany;
| | - Beate Sodeik
- DZIF German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany
- RESIST Cluster of Excellence, Hannover Medical School, Hannover, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany;
| |
Collapse
|
5
|
Ramirez JM, Calderon-Zavala AC, Balaram A, Heldwein EE. In vitro reconstitution of herpes simplex virus 1 fusion identifies low pH as a fusion co-trigger. mBio 2023; 14:e0208723. [PMID: 37874146 PMCID: PMC10746285 DOI: 10.1128/mbio.02087-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/25/2023] Open
Abstract
IMPORTANCE HSV-1 causes lifelong, incurable infections and diseases ranging from mucocutaneous lesions to fatal encephalitis. Fusion of viral and host membranes is a critical step in HSV-1 infection of target cells that requires multiple factors on both the viral and host sides. Due to this complexity, many fundamental questions remain unanswered, such as the identity of the viral and host factors that are necessary and sufficient for HSV-1-mediated membrane fusion and the nature of the fusion trigger. Here, we developed a simplified in vitro fusion assay to examine the fusion requirements and identified low pH as a co-trigger for virus-mediated fusion in vitro. We hypothesize that low pH has a critical role in cell entry and, potentially, pathogenesis.
Collapse
Affiliation(s)
- J. Martin Ramirez
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Medical Scientist Training Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ariana C. Calderon-Zavala
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ariane Balaram
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ekaterina E. Heldwein
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Medical Scientist Training Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Döhner K, Serrero MC, Sodeik B. The role of nuclear pores and importins for herpes simplex virus infection. Curr Opin Virol 2023; 62:101361. [PMID: 37672874 DOI: 10.1016/j.coviro.2023.101361] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023]
Abstract
Microtubule transport and nuclear import are functionally connected, and the nuclear pore complex (NPC) can interact with microtubule motors. For several alphaherpesvirus proteins, nuclear localization signals (NLSs) and their interactions with specific importin-α proteins have been characterized. Here, we review recent insights on the roles of microtubule motors, capsid-associated NLSs, and importin-α proteins for capsid transport, capsid docking to NPCs, and genome release into the nucleoplasm, as well as the role of importins for nuclear viral transcription, replication, capsid assembly, genome packaging, and nuclear capsid egress. Moreover, importin-α proteins exert antiviral effects by promoting the nuclear import of transcription factors inducing the expression of interferons (IFN), cytokines, and IFN-stimulated genes, and the IFN-inducible MxB restricts capsid docking to NPCs.
Collapse
Affiliation(s)
- Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany; Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany.
| | - Manutea C Serrero
- Institute of Virology, Hannover Medical School, Hannover, Germany; RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany; RESIST - Cluster of Excellence, Hannover Medical School, Hannover, Germany; DZIF - German Centre for Infection Research, Braunschweig, Hannover, Germany.
| |
Collapse
|
7
|
Association of THBS3 with Glycoprotein D Promotes Pseudorabies Virus Attachment, Fusion, and Entry. J Virol 2023; 97:e0187122. [PMID: 36648234 PMCID: PMC9972988 DOI: 10.1128/jvi.01871-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Pseudorabies virus (PRV) is a neurotropic virus causing obvious neurological disorders and reproductive failure in pigs. PRV entry into target cells is a complex multistep process initiated by interacting viral envelope glycoproteins with cellular receptors. In the current study, we found that thrombospondin 3 (THBS3) plays an important role in PRV entry into target cells, indicating that THBS3 is a new PRV coreceptor. To confirm this hypothesis, the knockdown of THBS3 in several permissive cells inhibited PRV primary infection, and overexpression of THBS3 in PK15 cells promoted PRV infection. CRISPR-Cas9 knockout markedly reduced PRV infection in PK15 cells. Antibodies against THBS3 blocked PRV infection in naturally permissive target cells. Moreover, soluble THBS3 protein neutralized the infectivity of PRV. Mechanistically, THBS3 interacted with the PRV gD via its N and C termini to facilitate PRV binding in permissive and nonpermissive cells. Also, in the absence of Nectin-1, THBS3 promoted cell-to-cell fusion mediated by virus glycoproteins. While THBS3 alone could not increase virus entry, overexpression of it in the presence of Nectin-1 promoted virus entry into CHO-K1 cells. Our results have identified THBS3 as a critical player in PRV binding and subsequent membrane fusion and entry. IMPORTANCE Herpesvirus entry occurs through a cascade of virus-cell interactions, and multiple surface glycoproteins play a role in virus binding and entry during the virus invasion process. Early studies showed that attachment to cells by PRV, as well as other alphaherpesviruses, is mediated by interactions between the viral glycoprotein gC and cell membrane proteoglycans carrying heparan sulfate chains (HSPGs). However, gD may also be involved in virus binding in an HSPG-independent manner. To date, the respective cellular receptors are still unknown. In this report, we identified a host molecule, THBS3, involved in gD-mediated PRV binding and subsequent membrane fusion and entry, which increases our understanding of the initial events in alpha herpesvirus infections.
Collapse
|
8
|
Kazmierski J, Elsner C, Döhner K, Xu S, Ducroux A, Pott F, Jansen J, Thorball CW, Zeymer O, Zhou X, Fedorov R, Fellay J, Löffler MW, Weber ANR, Sodeik B, Goffinet C. A Baseline Cellular Antiviral State Is Maintained by cGAS and Its Most Frequent Naturally Occurring Variant rs610913. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:535-547. [PMID: 35851540 DOI: 10.4049/jimmunol.2100685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 05/13/2022] [Indexed: 10/17/2023]
Abstract
Upon recognition of aberrantly located DNA, the innate immune sensor cyclic GMP-AMP synthase (cGAS) activates stimulator of IFN genes (STING)/IFN regulatory factor (IRF)3-driven antiviral responses. In this study, we characterized the ability of a specific variant of the human cGAS-encoding gene MB21D1, rs610913, to alter cGAS-mediated DNA sensing and viral infection. rs610913 is a frequent G>T polymorphism resulting in a P261H exchange in the cGAS protein. Data from the International Collaboration for the Genomics of HIV suggested that rs610913 nominally associates with HIV-1 acquisition in vivo. Molecular modeling of cGAS(P261H) hinted toward the possibility for an additional binding site for a potential cellular cofactor in cGAS dimers. However, cGAS(wild-type [WT]) or cGAS(P261H)-reconstituted THP-1 cGAS knockout cells shared steady-state expression of IFN-stimulated genes, as opposed to cells expressing the enzymatically inactive cGAS(G212A/S213A). Accordingly, cGAS(WT) and cGAS(P261H) cells were less susceptible to lentiviral transduction and infection with HIV-1, HSV-1, and Chikungunya virus as compared with cGAS knockout or cGAS(G212A/S213A) cells. Upon DNA challenge, innate immune activation appeared to be mildly reduced upon expression of cGAS(P261H) compared with cGAS(WT). Finally, DNA challenge of PBMCs from donors homozygously expressing rs610913 provoked a trend toward a slightly reduced type I IFN response as compared with PBMCs from GG donors. Taken together, the steady-state activity of cGAS maintains a baseline antiviral state rendering cells more refractory to IFN-stimulated gene-sensitive viral infections. rs610913 failed to grossly differ phenotypically from the WT gene, suggesting that cGAS(P261H) and WT cGAS share a similar ability to sense viral infections in vivo.
Collapse
Affiliation(s)
- Julia Kazmierski
- Institute of Virology, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- Institute of Experimental Virology, Twincore Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Carina Elsner
- Institute of Experimental Virology, Twincore Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Shuting Xu
- Institute of Experimental Virology, Twincore Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Aurélie Ducroux
- Institute of Experimental Virology, Twincore Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Fabian Pott
- Institute of Virology, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- Institute of Experimental Virology, Twincore Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Jenny Jansen
- Institute of Virology, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Christian W Thorball
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ole Zeymer
- Institute for Biophysical Chemistry, Research Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
- RESIST-Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Xiaoyi Zhou
- Institute for Biophysical Chemistry, Research Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
- RESIST-Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Roman Fedorov
- Institute for Biophysical Chemistry, Research Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
- RESIST-Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Jacques Fellay
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Markus W Löffler
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
- iFIT-Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- iFIT-Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
- CMFI-Cluster of Excellence (EXC 2124) "Controlling Microbes to Fight Infection," University of Tübingen, Tübingen, Germany; and
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- RESIST-Cluster of Excellence, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Partner Site, Hannover, Germany
| | - Christine Goffinet
- Institute of Virology, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- Institute of Experimental Virology, Twincore Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| |
Collapse
|
9
|
Serrero MC, Girault V, Weigang S, Greco TM, Ramos-Nascimento A, Anderson F, Piras A, Hickford Martinez A, Hertzog J, Binz A, Pohlmann A, Prank U, Rehwinkel J, Bauerfeind R, Cristea IM, Pichlmair A, Kochs G, Sodeik B. The interferon-inducible GTPase MxB promotes capsid disassembly and genome release of herpesviruses. eLife 2022; 11:e76804. [PMID: 35475759 PMCID: PMC9150894 DOI: 10.7554/elife.76804] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022] Open
Abstract
Host proteins sense viral products and induce defence mechanisms, particularly in immune cells. Using cell-free assays and quantitative mass spectrometry, we determined the interactome of capsid-host protein complexes of herpes simplex virus and identified the large dynamin-like GTPase myxovirus resistance protein B (MxB) as an interferon-inducible protein interacting with capsids. Electron microscopy analyses showed that cytosols containing MxB had the remarkable capability to disassemble the icosahedral capsids of herpes simplex viruses and varicella zoster virus into flat sheets of connected triangular faces. In contrast, capsids remained intact in cytosols with MxB mutants unable to hydrolyse GTP or to dimerize. Our data suggest that MxB senses herpesviral capsids, mediates their disassembly, and thereby restricts the efficiency of nuclear targeting of incoming capsids and/or the assembly of progeny capsids. The resulting premature release of viral genomes from capsids may enhance the activation of DNA sensors, and thereby amplify the innate immune responses.
Collapse
Affiliation(s)
- Manutea C Serrero
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
| | | | - Sebastian Weigang
- Institute of Virology, Freiburg University Medical Center, University of FreiburgFreiburgGermany
| | - Todd M Greco
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | | | - Fenja Anderson
- Institute of Virology, Hannover Medical SchoolHannoverGermany
| | - Antonio Piras
- Institute of Virology, Technical University MunichMunichGermany
| | | | - Jonny Hertzog
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Anne Binz
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner SiteHannoverGermany
| | - Anja Pohlmann
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner SiteHannoverGermany
| | - Ute Prank
- Institute of Virology, Hannover Medical SchoolHannoverGermany
| | - Jan Rehwinkel
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Rudolf Bauerfeind
- Research Core Unit Laser Microscopy, Hannover Medical SchoolHannoverGermany
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Andreas Pichlmair
- Institute of Virology, Technical University MunichMunichGermany
- German Center for Infection Research (DZIF), Munich Partner siteMunichGermany
| | - Georg Kochs
- Institute of Virology, Freiburg University Medical Center, University of FreiburgFreiburgGermany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner SiteHannoverGermany
| |
Collapse
|
10
|
A Multi-Laboratory Comparison of Methods for Detection and Quantification of African Swine Fever Virus. Pathogens 2022; 11:pathogens11030325. [PMID: 35335649 PMCID: PMC8949307 DOI: 10.3390/pathogens11030325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/01/2023] Open
Abstract
African swine fever is a viral disease of the family Suidae. Methods to detect and quantify African swine fever virus (ASFV) include qPCR and virus infectivity assays. Individual laboratories often use in-house procedures for these assays, which can hamper the comparison of results. The objective of this study was to estimate the probability of ASFV detection using these assays, and to determine the inter-test correlations between results. This was achieved by testing a panel of 80 samples at three reference laboratories. Samples were analysed using nucleic acid extraction and qPCR, as well as virus infectivity assays. For qPCR, a very high probability (ranging from 0.96 to 1.0) of detecting ASFV DNA was observed for all tested systems. For virus infectivity assays in cells, the probability of detecting infectious ASFV varied from 0.68 to 0.90 and was highest using pulmonary alveolar macrophages, followed by MARC145 cells, peripheral blood monocytes, and finally wild boar lung cells. Intraclass correlation coefficient estimates of 0.97 (0.96–0.98) between qPCR methods, 0.80 (0.74–0.85) to 0.94 (0.92–0.96) between virus infectivity assays, and 0.77 (0.68–0.83) to 0.95 (0.93–0.96) between qPCR methods and virus infectivity assays were obtained. These findings show that qPCR gives the highest probability for the detection of ASFV.
Collapse
|
11
|
Wilson DW. Motor Skills: Recruitment of Kinesins, Myosins and Dynein during Assembly and Egress of Alphaherpesviruses. Viruses 2021; 13:v13081622. [PMID: 34452486 PMCID: PMC8402756 DOI: 10.3390/v13081622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
The alphaherpesviruses are pathogens of the mammalian nervous system. Initial infection is commonly at mucosal epithelia, followed by spread to, and establishment of latency in, the peripheral nervous system. During productive infection, viral gene expression, replication of the dsDNA genome, capsid assembly and genome packaging take place in the infected cell nucleus, after which mature nucleocapsids emerge into the cytoplasm. Capsids must then travel to their site of envelopment at cytoplasmic organelles, and enveloped virions need to reach the cell surface for release and spread. Transport at each of these steps requires movement of alphaherpesvirus particles through a crowded and viscous cytoplasm, and for distances ranging from several microns in epithelial cells, to millimeters or even meters during egress from neurons. To solve this challenging problem alphaherpesviruses, and their assembly intermediates, exploit microtubule- and actin-dependent cellular motors. This review focuses upon the mechanisms used by alphaherpesviruses to recruit kinesin, myosin and dynein motors during assembly and egress.
Collapse
Affiliation(s)
- Duncan W. Wilson
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; ; Tel.: +1-718-430-2305
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
12
|
Richards A, Berth SH, Brady S, Morfini G. Engagement of Neurotropic Viruses in Fast Axonal Transport: Mechanisms, Potential Role of Host Kinases and Implications for Neuronal Dysfunction. Front Cell Neurosci 2021; 15:684762. [PMID: 34234649 PMCID: PMC8255969 DOI: 10.3389/fncel.2021.684762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/17/2021] [Indexed: 11/28/2022] Open
Abstract
Much remains unknown about mechanisms sustaining the various stages in the life cycle of neurotropic viruses. An understanding of those mechanisms operating before their replication and propagation could advance the development of effective anti-viral strategies. Here, we review our current knowledge of strategies used by neurotropic viruses to undergo bidirectional movement along axons. We discuss how the invasion strategies used by specific viruses might influence their mode of interaction with selected components of the host’s fast axonal transport (FAT) machinery, including specialized membrane-bounded organelles and microtubule-based motor proteins. As part of this discussion, we provide a critical evaluation of various reported interactions among viral and motor proteins and highlight limitations of some in vitro approaches that led to their identification. Based on a large body of evidence documenting activation of host kinases by neurotropic viruses, and on recent work revealing regulation of FAT through phosphorylation-based mechanisms, we posit a potential role of host kinases on the engagement of viruses in retrograde FAT. Finally, we briefly describe recent evidence linking aberrant activation of kinase pathways to deficits in FAT and neuronal degeneration in the context of human neurodegenerative diseases. Based on these findings, we speculate that neurotoxicity elicited by viral infection may involve deregulation of host kinases involved in the regulation of FAT and other cellular processes sustaining neuronal function and survival.
Collapse
Affiliation(s)
- Alexsia Richards
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States
| | - Sarah H Berth
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Scott Brady
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
13
|
Elsner C, Ponnurangam A, Kazmierski J, Zillinger T, Jansen J, Todt D, Döhner K, Xu S, Ducroux A, Kriedemann N, Malassa A, Larsen PK, Hartmann G, Barchet W, Steinmann E, Kalinke U, Sodeik B, Goffinet C. Absence of cGAS-mediated type I IFN responses in HIV-1-infected T cells. Proc Natl Acad Sci U S A 2020; 117:19475-19486. [PMID: 32709741 PMCID: PMC7431009 DOI: 10.1073/pnas.2002481117] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The DNA sensor cGAS catalyzes the production of the cyclic dinucleotide cGAMP, resulting in type I interferon responses. We addressed the functionality of cGAS-mediated DNA sensing in human and murine T cells. Activated primary CD4+ T cells expressed cGAS and responded to plasmid DNA by upregulation of ISGs and release of bioactive interferon. In mouse T cells, cGAS KO ablated sensing of plasmid DNA, and TREX1 KO enabled cells to sense short immunostimulatory DNA. Expression of IFIT1 and MX2 was downregulated and upregulated in cGAS KO and TREX1 KO T cell lines, respectively, compared to parental cells. Despite their intact cGAS sensing pathway, human CD4+ T cells failed to mount a reverse transcriptase (RT) inhibitor-sensitive immune response following HIV-1 infection. In contrast, infection of human T cells with HSV-1 that is functionally deficient for the cGAS antagonist pUL41 (HSV-1ΔUL41N) resulted in a cGAS-dependent type I interferon response. In accordance with our results in primary CD4+ T cells, plasmid challenge or HSV-1ΔUL41N inoculation of T cell lines provoked an entirely cGAS-dependent type I interferon response, including IRF3 phosphorylation and expression of ISGs. In contrast, no RT-dependent interferon response was detected following transduction of T cell lines with VSV-G-pseudotyped lentiviral or gammaretroviral particles. Together, T cells are capable to raise a cGAS-dependent cell-intrinsic response to both plasmid DNA challenge or inoculation with HSV-1ΔUL41N. However, HIV-1 infection does not appear to trigger cGAS-mediated sensing of viral DNA in T cells, possibly by revealing viral DNA of insufficient quantity, length, and/or accessibility to cGAS.
Collapse
Affiliation(s)
- Carina Elsner
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
- Institute for Virology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Aparna Ponnurangam
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
| | - Julia Kazmierski
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Thomas Zillinger
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Jenny Jansen
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Daniel Todt
- Department of Molecular and Medical Virology, Ruhr University Bochum, 44801 Bochum, Germany
- European Virus Bioinformatics Center, 07743 Jena, Germany
| | - Katinka Döhner
- Institute of Virology, Hanover Medical School, 30625 Hanover, Germany
| | - Shuting Xu
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
| | - Aurélie Ducroux
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
| | - Nils Kriedemann
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
| | - Angelina Malassa
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
| | - Pia-Katharina Larsen
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Winfried Barchet
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, University of Bonn, 53127 Bonn, Germany
- German Center for Infection Research, 50935 Cologne-Bonn, Germany
| | - Eike Steinmann
- Department of Molecular and Medical Virology, Ruhr University Bochum, 44801 Bochum, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany
| | - Beate Sodeik
- Institute of Virology, Hanover Medical School, 30625 Hanover, Germany
- Cluster of Excellence Resolving Infection Susceptibility (Excellence Cluster 2155), Hanover Medical School, 30625 Hanover, Germany
| | - Christine Goffinet
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hanover, Germany;
- Institute of Virology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| |
Collapse
|
14
|
Wang Y, Huang L, Wang Y, Luo W, Li F, Xiao J, Qin S, Wang Z, Song X, Wang Y, Jin F, Wang Y. Single-cell RNA-sequencing analysis identifies host long noncoding RNA MAMDC2-AS1 as a co-factor for HSV-1 nuclear transport. Int J Biol Sci 2020; 16:1586-1603. [PMID: 32226304 PMCID: PMC7097924 DOI: 10.7150/ijbs.42556] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/02/2020] [Indexed: 12/29/2022] Open
Abstract
Herpes simplex virus (HSV) type 1 (HSV-1) infection exhibited high heterogeneity at individual cells level, including the different gene expression patterns and varying amounts of progeny virus. However, the underlying mechanism of such variability remains obscure. The importance of host long noncoding RNAs (lncRNAs) in virus infection had been recognized, while the contribution of lncRNAs to the heterogeneous infection remains unknown. Herein, a prior single-cell RNA sequencing data using HSV-1 reporter strain expressing ICP4-YFP was re-analyzed to obtain the differentially expressed lncRNA between the successfully initiated viral gene expression (ICP4-YFP+) cells and the aborted infection cells (ICP4-YFP-). The ICP4-YFP+ population show a higher abundance of MAMDC2 antisense 1 (MAMDC2-AS1) lncRNA than ICP4-YFP- population. MAMDC2-AS1 silencing reduces the expression of HSV-1 immediate early (IE) genes and limit HSV-1 infection in human host cells. Consistently, ectopic expression of MAMDC2-AS1 enhances HSV-1 IE genes transcription and facilitates the formation of HSV-1-induced plaques. Mechanically, both RNA-pull down and RNA immunoprecipitation assays show that MAMDC2-AS1 interacts with the RNA binding protein heat shock protein 90α (Hsp90α), a molecular chaperone involving in the nuclear import of HSV-1. The MAMDC2-AS1-Hsp90α interaction facilitates the nuclear transport of viral tegument protein VP16, the core factor initiating the expression of HSV-1 IE genes. The transcription factor YY1 mediates the induction of MAMDC2-AS1 upon HSV-1 infection. Our study elucidates the contribution of lncRNA to HSV-1 infection susceptibility in human cells and the role of Hsp90α RNA binding activity in HSV-1 infection.
Collapse
Affiliation(s)
- Yiliang Wang
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Lianzhou Huang
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China.,College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Yun Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, PR China
| | - Weisheng Luo
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Feng Li
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Ji Xiao
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Shurong Qin
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Zhaoyang Wang
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Xiaowei Song
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Yuan Wang
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Fujun Jin
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, PR China
| | - Yifei Wang
- College of Life science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| |
Collapse
|
15
|
Diwaker D, Wilson DW. Microtubule-Dependent Trafficking of Alphaherpesviruses in the Nervous System: The Ins and Outs. Viruses 2019; 11:v11121165. [PMID: 31861082 PMCID: PMC6950448 DOI: 10.3390/v11121165] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/13/2019] [Accepted: 12/15/2019] [Indexed: 12/12/2022] Open
Abstract
The Alphaherpesvirinae include the neurotropic pathogens herpes simplex virus and varicella zoster virus of humans and pseudorabies virus of swine. These viruses establish lifelong latency in the nuclei of peripheral ganglia, but utilize the peripheral tissues those neurons innervate for productive replication, spread, and transmission. Delivery of virions from replicative pools to the sites of latency requires microtubule-directed retrograde axonal transport from the nerve terminus to the cell body of the sensory neuron. As a corollary, during reactivation newly assembled virions must travel along axonal microtubules in the anterograde direction to return to the nerve terminus and infect peripheral tissues, completing the cycle. Neurotropic alphaherpesviruses can therefore exploit neuronal microtubules and motors for long distance axonal transport, and alternate between periods of sustained plus end- and minus end-directed motion at different stages of their infectious cycle. This review summarizes our current understanding of the molecular details by which this is achieved.
Collapse
Affiliation(s)
- Drishya Diwaker
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Duncan W. Wilson
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
- Correspondence: ; Tel.: +1-(718)-430-2305
| |
Collapse
|
16
|
Grosche L, Döhner K, Düthorn A, Hickford-Martinez A, Steinkasserer A, Sodeik B. Herpes Simplex Virus Type 1 Propagation, Titration and Single-step Growth Curves. Bio Protoc 2019; 9:e3441. [PMID: 33654936 DOI: 10.21769/bioprotoc.3441] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/31/2019] [Accepted: 11/18/2019] [Indexed: 01/13/2023] Open
Abstract
Given the endemic seroprevalence of herpes simplex viruses (HSV), its associated human diseases, and the emergence of acyclovir-resistant strains, there is a continuous need for better antiviral therapies. Towards this aim, identifying mechanistic details of how HSV-1 manipulates infected cells, how it modulates the immune responses, and how it causes diseases are essential. Measuring titers and growth kinetics of clinical isolates and viral mutants are important for a thorough characterization of viral phenotypes in vitro and in vivo. We provide protocols for the preparation as well as titration of HSV-1 stocks, and explain how to perform single-step growth curves to characterize the functions of viral proteins or host factors during infection. In particular, we describe methods to prepare and characterize high-titer HSV-1 stocks with low genome to titer ratios that are required for infection studies in cell culture and animal experiments.
Collapse
Affiliation(s)
- Linda Grosche
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katinka Döhner
- Institute of Virology, OE5230, Hannover Medical School, Hannover, Germany
| | - Alexandra Düthorn
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | | | - Beate Sodeik
- Institute of Virology, OE5230, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Bearer EL, Wu C. Herpes Simplex Virus, Alzheimer's Disease and a Possible Role for Rab GTPases. Front Cell Dev Biol 2019; 7:134. [PMID: 31448273 PMCID: PMC6692634 DOI: 10.3389/fcell.2019.00134] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/04/2019] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus (HSV) is a common pathogen, infecting 85% of adults in the United States. After reaching the nucleus of the long-lived neuron, HSV may enter latency to persist throughout the life span. Re-activation of latent herpesviruses is associated with progressive cognitive impairment and Alzheimer's disease (AD). As an enveloped DNA virus, HSV exploits cellular membrane systems for its life cycle, and thereby comes in contact with the Rab family of GTPases, master regulators of intracellular membrane dynamics. Knock-down and overexpression of specific Rabs reduce HSV production. Disheveled membrane compartments could lead to AD because membrane sorting and trafficking are crucial for synaptic vesicle formation, neuronal survival signaling and Abeta production. Amyloid precursor protein (APP), a transmembrane glycoprotein, is the parent of Abeta, the major component of senile plaques in AD. Up-regulation of APP expression due to HSV is significant since excess APP interferes with Rab5 endocytic trafficking in neurons. Here, we show that purified PC12-cell endosomes transport both anterograde and retrograde when injected into the squid giant axon at rates similar to isolated HSV. Intracellular HSV co-fractionates with these endosomes, contains APP, Rab5 and TrkA, and displays a second membrane. HSV infected PC12 cells up-regulate APP expression. Whether interference with Rabs has a specific effect on HSV or indirectly affects membrane compartment dynamics co-opted by virus needs further study. Ultimately Rabs, their effectors or their membrane-binding partners may serve as handles to reduce the impact of viral re-activation on cognitive function, or even as more general-purpose anti-microbial therapies.
Collapse
Affiliation(s)
- Elaine L. Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Chengbiao Wu
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
18
|
The Carboxyl Terminus of Tegument Protein pUL21 Contributes to Pseudorabies Virus Neuroinvasion. J Virol 2019; 93:JVI.02052-18. [PMID: 30651360 DOI: 10.1128/jvi.02052-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/08/2019] [Indexed: 12/21/2022] Open
Abstract
Following its entry into cells, pseudorabies virus (PRV) utilizes microtubules to deliver its nucleocapsid to the nucleus. Previous studies have shown that PRV VP1/2 is an effector of dynein-mediated capsid transport. However, the mechanism of PRV for recruiting microtubule motor proteins for successful neuroinvasion and neurovirulence is not well understood. Here, we provide evidence that PRV pUL21 is an inner tegument protein. We tested its interaction with the cytoplasmic light chains using a bimolecular fluorescence complementation (BiFC) assay and observed that PRV pUL21 interacts with Roadblock-1. This interaction was confirmed by coimmunoprecipitation (co-IP) assays. We also determined the efficiency of retrograde and anterograde axonal transport of PRV strains in explanted neurons using a microfluidic chamber system and investigated pUL21's contribution to PRV neuroinvasion in vivo Further data showed that the carboxyl terminus of pUL21 is essential for its interaction with Roadblock-1, and this domain contributes to PRV retrograde axonal transport in vitro and in vivo Our findings suggest that the carboxyl terminus of pUL21 contributes to PRV neuroinvasion.IMPORTANCE Herpesviruses are a group of DNA viruses that infect both humans and animals. Alphaherpesviruses are distinguished by their ability to establish latent infection in peripheral neurons. After entering neurons, the herpesvirus capsid interacts with cellular motor proteins and undergoes retrograde transport on axon microtubules. This elaborate process is vital to the herpesvirus lifecycle, but the underlying mechanism remains poorly understood. Here, we determined that pUL21 is an inner tegument protein of pseudorabies virus (PRV) and that it interacts with the cytoplasmic dynein light chain Roadblock-1. We also observed that pUL21 promotes retrograde transport of PRV in neuronal cells. Furthermore, our findings confirm that pUL21 contributes to PRV neuroinvasion in vivo Importantly, the carboxyl terminus of pUL21 is responsible for interaction with Roadblock-1, and this domain contributes to PRV neuroinvasion. This study offers fresh insights into alphaherpesvirus neuroinvasion and the interaction between virus and host during PRV infection.
Collapse
|
19
|
Nathan L, Daniel S. Single Virion Tracking Microscopy for the Study of Virus Entry Processes in Live Cells and Biomimetic Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1215:13-43. [PMID: 31317494 PMCID: PMC7122913 DOI: 10.1007/978-3-030-14741-9_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The most widely-used assays for studying viral entry, including infectivity, cofloatation, and cell-cell fusion assays, yield functional information but provide low resolution of individual entry steps. Structural characterization provides high-resolution conformational information, but on its own is unable to address the functional significance of these conformations. Single virion tracking microscopy techniques provide more detail on the intermediate entry steps than infection assays and more functional information than structural methods, bridging the gap between these methods. In addition, single virion approaches also provide dynamic information about the kinetics of entry processes. This chapter reviews single virion tracking techniques and describes how they can be applied to study specific virus entry steps. These techniques provide information complementary to traditional ensemble approaches. Single virion techniques may either probe virion behavior in live cells or in biomimetic platforms. Synthesizing information from ensemble, structural, and single virion techniques ultimately yields a more complete understanding of the viral entry process than can be achieved by any single method alone.
Collapse
Affiliation(s)
- Lakshmi Nathan
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA.
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
20
|
Ibáñez FJ, Farías MA, Gonzalez-Troncoso MP, Corrales N, Duarte LF, Retamal-Díaz A, González PA. Experimental Dissection of the Lytic Replication Cycles of Herpes Simplex Viruses in vitro. Front Microbiol 2018; 9:2406. [PMID: 30386309 PMCID: PMC6198116 DOI: 10.3389/fmicb.2018.02406] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022] Open
Abstract
Herpes simplex viruses type 1 and type 2 (HSV-1 and HSV-2) produce lifelong infections and are highly prevalent in the human population. Both viruses elicit numerous clinical manifestations and produce mild-to-severe diseases that affect the skin, eyes, and brain, among others. Despite the existence of numerous antivirals against HSV, such as acyclovir and acyclovir-related analogs, virus variants that are resistant to these compounds can be isolated from immunosuppressed individuals. For such isolates, second-line drugs can be used, yet they frequently produce adverse side effects. Furthermore, topical antivirals for treating cutaneous HSV infections usually display poor to moderate efficacy. Hence, better or novel anti-HSV antivirals are needed and details on their mechanisms of action would be insightful for improving their efficacy and identifying specific molecular targets. Here, we review and dissect the lytic replication cycles of herpes simplex viruses, discussing key steps involved in cell infection and the processes that yield new virions. Additionally, we review and discuss rapid, easy-to-perform and simple experimental approaches for studying key steps involved in HSV replication to facilitate the identification of the mechanisms of action of anti-HSV compounds.
Collapse
Affiliation(s)
- Francisco J Ibáñez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maria P Gonzalez-Troncoso
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás Corrales
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
21
|
Promyelocytic leukemia (PML) nuclear bodies (NBs) induce latent/quiescent HSV-1 genomes chromatinization through a PML NB/Histone H3.3/H3.3 Chaperone Axis. PLoS Pathog 2018; 14:e1007313. [PMID: 30235352 PMCID: PMC6168178 DOI: 10.1371/journal.ppat.1007313] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/02/2018] [Accepted: 08/31/2018] [Indexed: 12/31/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) latency establishment is tightly controlled by promyelocytic leukemia (PML) nuclear bodies (NBs) (or ND10), although their exact contribution is still elusive. A hallmark of HSV-1 latency is the interaction between latent viral genomes and PML NBs, leading to the formation of viral DNA-containing PML NBs (vDCP NBs), and the complete silencing of HSV-1. Using a replication-defective HSV-1-infected human primary fibroblast model reproducing the formation of vDCP NBs, combined with an immuno-FISH approach developed to detect latent/quiescent HSV-1, we show that vDCP NBs contain both histone H3.3 and its chaperone complexes, i.e., DAXX/ATRX and HIRA complex (HIRA, UBN1, CABIN1, and ASF1a). HIRA also co-localizes with vDCP NBs present in trigeminal ganglia (TG) neurons from HSV-1-infected wild type mice. ChIP and Re-ChIP show that vDCP NBs-associated latent/quiescent viral genomes are chromatinized almost exclusively with H3.3 modified on its lysine (K) 9 by trimethylation, consistent with an interaction of the H3.3 chaperones with multiple viral loci and with the transcriptional silencing of HSV-1. Only simultaneous inactivation of both H3.3 chaperone complexes has a significant impact on the deposition of H3.3 on viral genomes, suggesting a compensation mechanism. In contrast, the sole depletion of PML significantly impacts the chromatinization of the latent/quiescent viral genomes with H3.3 without any overall replacement with H3.1. vDCP NBs-associated HSV-1 genomes are not definitively silenced since the destabilization of vDCP NBs by ICP0, which is essential for HSV-1 reactivation in vivo, allows the recovery of a transcriptional lytic program and the replication of viral genomes. Consequently, the present study demonstrates a specific chromatin regulation of vDCP NBs-associated latent/quiescent HSV-1 through an H3.3-dependent HSV-1 chromatinization involving the two H3.3 chaperones DAXX/ATRX and HIRA complexes. Additionally, the study reveals that PML NBs are major actors in latent/quiescent HSV-1 H3.3 chromatinization through a PML NB/histone H3.3/H3.3 chaperone axis. An understanding of the molecular mechanisms contributing to the persistence of a virus in its host is essential to be able to control viral reactivation and its associated diseases. Herpes simplex virus 1 (HSV-1) is a human pathogen that remains latent in the PNS and CNS of the infected host. The latency is unstable, and frequent reactivations of the virus are responsible for PNS and CNS pathologies. It is thus crucial to understand the physiological, immunological and molecular levels of interplay between latent HSV-1 and the host. Promyelocytic leukemia (PML) nuclear bodies (NBs) control viral infections by preventing the onset of lytic infection. In previous studies, we showed a major role of PML NBs in favoring the establishment of a latent state for HSV-1. A hallmark of HSV-1 latency establishment is the formation of PML NBs containing the viral genome, which we called “viral DNA-containing PML NBs” (vDCP NBs). The genome entrapped in the vDCP NBs is transcriptionally silenced. This naturally occurring latent/quiescent state could, however, be transcriptionally reactivated. Therefore, understanding the role of PML NBs in controlling the establishment of HSV-1 latency and its reactivation is essential to design new therapeutic approaches based on the prevention of viral reactivation.
Collapse
|
22
|
The Apical Region of the Herpes Simplex Virus Major Capsid Protein Promotes Capsid Maturation. J Virol 2018; 92:JVI.00821-18. [PMID: 29976665 DOI: 10.1128/jvi.00821-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/27/2018] [Indexed: 01/12/2023] Open
Abstract
The herpesvirus capsid assembles in the nucleus as an immature procapsid precursor built around viral scaffold proteins. The event that initiates procapsid maturation is unknown, but it is dependent upon activation of the VP24 internal protease. Scaffold cleavage triggers angularization of the shell and its decoration with the VP26 and pUL25 capsid-surface proteins. In both the procapsid and mature angularized capsid, the apical region of the major capsid protein (VP5) is surface exposed. We investigated whether the VP5 apical region contributes to intracellular transport dynamics following entry into primary sensory neurons and also tested the hypothesis that conserved negatively charged amino acids in the apical region contribute to VP26 acquisition. To our surprise, neither hypothesis proved true. Instead, mutation of glutamic acid residues in the apical region delayed viral propagation and induced focal capsid accumulations in nuclei. Examination of capsid morphogenesis based on epitope unmasking, capsid composition, and ultrastructural analysis indicated that these clusters consisted of procapsids. The results demonstrate that, in addition to established events that occur inside the capsid, the exterior capsid shell promotes capsid morphogenesis and maturation.IMPORTANCE Herpesviruses assemble capsids and encapsidate their genomes by a process that is unlike those of other mammalian viruses but is similar to those of some bacteriophage. Many important aspects of herpesvirus morphogenesis remain enigmatic, including how the capsid shell matures into a stable angularized configuration. Capsid maturation is triggered by activation of a protease that cleaves an internal protein scaffold. We report on the fortuitous discovery that a region of the major capsid protein that is exposed on the outer surface of the capsid also contributes to capsid maturation, demonstrating that the morphogenesis of the capsid shell from its procapsid precursor to the mature angularized form is dependent upon internal and external components of the megastructure.
Collapse
|
23
|
Interaction between the cellular E3 ubiquitin ligase SIAH-1 and the viral immediate-early protein ICP0 enables efficient replication of Herpes Simplex Virus type 2 in vivo. PLoS One 2018; 13:e0201880. [PMID: 30080903 PMCID: PMC6078308 DOI: 10.1371/journal.pone.0201880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/24/2018] [Indexed: 01/17/2023] Open
Abstract
Herpes Simplex Virus type 2 (HSV-2) is a neurotropic human pathogen. Upon de novo infection, the viral infected cell protein 0 (ICP0) is immediately expressed and interacts with various cellular components during the viral replication cycle. ICP0 is a multifunctional regulatory protein that has been shown to be important for both efficient viral replication and virus reactivation from latency. In particular, as previously demonstrated in transfected tissue culture models, ICP0 interacts with the cellular E3 ubiquitin ligase SIAH-1, which targets ICP0 for proteasomal degradation. However, the consequence of this virus-host interaction during the establishment of HSV-2 infection in vivo has not yet been elucidated. Here we confirmed that ICP0 of HSV-2 interacts with SIAH-1 via two conserved PxAxVxP amino acid binding motifs. We also demonstrate in vitro that a SIAH-1 binding-deficient HSV-2 strain, constructed by homologous recombination technology, exhibits an attenuated growth curve and impaired DNA and protein synthesis. This attenuated phenotype was also confirmed in an in vivo ocular infection mouse model. Specifically, viral load of the SIAH-1 binding-deficient HSV-2 mutant was significantly reduced in the trigeminal ganglia and brain stem at day 5 and 7 post infection. Our findings indicate that the interplay between ICP0 and SIAH-1 is important for efficient HSV-2 replication in vivo, thereby affecting viral dissemination kinetics in newly infected organisms, and possibly revealing novel targets for antiviral therapy.
Collapse
|
24
|
Walsh D, Naghavi MH. Exploitation of Cytoskeletal Networks during Early Viral Infection. Trends Microbiol 2018; 27:39-50. [PMID: 30033343 DOI: 10.1016/j.tim.2018.06.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/12/2018] [Accepted: 06/29/2018] [Indexed: 12/12/2022]
Abstract
Being dependent upon host transport systems to navigate the cytoplasm, viruses have evolved various strategies to manipulate cytoskeletal functions. Generally, viruses use the actin cytoskeleton to control entry and short-range transport at the cell periphery and exploit microtubules (MTs) for longer-range cytosolic transport, in some cases to reach the nucleus. While earlier studies established the fundamental importance of these networks to successful infection, the mechanistic details and true extent to which viruses usurp highly specialized host cytoskeletal regulators and motor adaptors is only beginning to emerge. This review outlines our current understanding of how cytoskeletal regulation contributes specifically to the early stages of viral infection, with a primary focus on retroviruses and herpesviruses as examples of recent advances in this area.
Collapse
Affiliation(s)
- Derek Walsh
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mojgan H Naghavi
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
25
|
Imaging, Tracking and Computational Analyses of Virus Entry and Egress with the Cytoskeleton. Viruses 2018; 10:v10040166. [PMID: 29614729 PMCID: PMC5923460 DOI: 10.3390/v10040166] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Viruses have a dual nature: particles are “passive substances” lacking chemical energy transformation, whereas infected cells are “active substances” turning-over energy. How passive viral substances convert to active substances, comprising viral replication and assembly compartments has been of intense interest to virologists, cell and molecular biologists and immunologists. Infection starts with virus entry into a susceptible cell and delivers the viral genome to the replication site. This is a multi-step process, and involves the cytoskeleton and associated motor proteins. Likewise, the egress of progeny virus particles from the replication site to the extracellular space is enhanced by the cytoskeleton and associated motor proteins. This overcomes the limitation of thermal diffusion, and transports virions and virion components, often in association with cellular organelles. This review explores how the analysis of viral trajectories informs about mechanisms of infection. We discuss the methodology enabling researchers to visualize single virions in cells by fluorescence imaging and tracking. Virus visualization and tracking are increasingly enhanced by computational analyses of virus trajectories as well as in silico modeling. Combined approaches reveal previously unrecognized features of virus-infected cells. Using select examples of complementary methodology, we highlight the role of actin filaments and microtubules, and their associated motors in virus infections. In-depth studies of single virion dynamics at high temporal and spatial resolutions thereby provide deep insight into virus infection processes, and are a basis for uncovering underlying mechanisms of how cells function.
Collapse
|
26
|
Infection and Transport of Herpes Simplex Virus Type 1 in Neurons: Role of the Cytoskeleton. Viruses 2018; 10:v10020092. [PMID: 29473915 PMCID: PMC5850399 DOI: 10.3390/v10020092] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 02/16/2018] [Accepted: 02/20/2018] [Indexed: 12/22/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a neuroinvasive human pathogen that has the ability to infect and replicate within epithelial cells and neurons and establish a life-long latent infection in sensory neurons. HSV-1 depends on the host cellular cytoskeleton for entry, replication, and exit. Therefore, HSV-1 has adapted mechanisms to promote its survival by exploiting the microtubule and actin cytoskeletons to direct its active transport, infection, and spread between neurons and epithelial cells during primary and recurrent infections. This review will focus on the currently known mechanisms utilized by HSV-1 to harness the neuronal cytoskeleton, molecular motors, and the secretory and exocytic pathways for efficient virus entry, axonal transport, replication, assembly, and exit from the distinct functional compartments (cell body and axon) of the highly polarized sensory neurons.
Collapse
|
27
|
Cytoskeletons in the Closet-Subversion in Alphaherpesvirus Infections. Viruses 2018; 10:v10020079. [PMID: 29438303 PMCID: PMC5850386 DOI: 10.3390/v10020079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/30/2018] [Accepted: 02/07/2018] [Indexed: 12/14/2022] Open
Abstract
Actin filaments, microtubules and intermediate filaments form the cytoskeleton of vertebrate cells. Involved in maintaining cell integrity and structure, facilitating cargo and vesicle transport, remodelling surface structures and motility, the cytoskeleton is necessary for the successful life of a cell. Because of the broad range of functions these filaments are involved in, they are common targets for viral pathogens, including the alphaherpesviruses. Human-tropic alphaherpesviruses are prevalent pathogens carried by more than half of the world’s population; comprising herpes simplex virus (types 1 and 2) and varicella-zoster virus, these viruses are characterised by their ability to establish latency in sensory neurons. This review will discuss the known mechanisms involved in subversion of and transport via the cytoskeleton during alphaherpesvirus infections, focusing on protein-protein interactions and pathways that have recently been identified. Studies on related alphaherpesviruses whose primary host is not human, along with comparisons to more distantly related beta and gammaherpesviruses, are also presented in this review. The need to decipher as-yet-unknown mechanisms exploited by viruses to hijack cytoskeletal components—to reveal the hidden cytoskeletons in the closet—will also be addressed.
Collapse
|
28
|
Döhner K, Ramos-Nascimento A, Bialy D, Anderson F, Hickford-Martinez A, Rother F, Koithan T, Rudolph K, Buch A, Prank U, Binz A, Hügel S, Lebbink RJ, Hoeben RC, Hartmann E, Bader M, Bauerfeind R, Sodeik B. Importin α1 is required for nuclear import of herpes simplex virus proteins and capsid assembly in fibroblasts and neurons. PLoS Pathog 2018; 14:e1006823. [PMID: 29304174 PMCID: PMC5773220 DOI: 10.1371/journal.ppat.1006823] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/18/2018] [Accepted: 12/16/2017] [Indexed: 01/28/2023] Open
Abstract
Herpesviruses are large DNA viruses which depend on many nuclear functions, and therefore on host transport factors to ensure specific nuclear import of viral and host components. While some import cargoes bind directly to certain transport factors, most recruit importin β1 via importin α. We identified importin α1 in a small targeted siRNA screen to be important for herpes simplex virus (HSV-1) gene expression. Production of infectious virions was delayed in the absence of importin α1, but not in cells lacking importin α3 or importin α4. While nuclear targeting of the incoming capsids, of the HSV-1 transcription activator VP16, and of the viral genomes were not affected, the nuclear import of the HSV-1 proteins ICP4 and ICP0, required for efficient viral transcription, and of ICP8 and pUL42, necessary for DNA replication, were reduced. Furthermore, quantitative electron microscopy showed that fibroblasts lacking importin α1 contained overall fewer nuclear capsids, but an increased proportion of mature nuclear capsids indicating that capsid formation and capsid egress into the cytoplasm were impaired. In neurons, importin α1 was also not required for nuclear targeting of incoming capsids, but for nuclear import of ICP4 and for the formation of nuclear capsid assembly compartments. Our data suggest that importin α1 is specifically required for the nuclear localization of several important HSV1 proteins, capsid assembly, and capsid egress into the cytoplasm, and may become rate limiting in situ upon infection at low multiplicity or in terminally differentiated cells such as neurons. Nuclear pore complexes are highly selective gateways that penetrate the nuclear envelope for bidirectional trafficking between the cytoplasm and the nucleoplasm. Viral and host cargoes have to engage specific transport factors to achieve active nuclear import and export. Like many human and animal DNA viruses, herpesviruses are critically dependent on many functions of the host cell nucleus. Alphaherpesviruses such as herpes simplex virus (HSV) cause many diseases upon productive infection in epithelial cells, fibroblasts and neurons. Here, we asked which nuclear transport factors of the host cells help HSV-1 to translocate viral components into the nucleus for viral gene expression, nuclear capsid assembly, capsid egress into the cytoplasm, and production of infectious virions. Our data show that HSV-1 requires the nuclear import factor importin α1 for efficient replication and virus assembly in fibroblasts and in mature neurons. To our knowledge this is the first time that a specific importin α isoform is shown to be required for herpesvirus infection. Our study fosters our understanding on how the different but highly homologous importin α isoforms could fulfill specific functions in vivo which are only understood for a very limited number of host and viral cargos.
Collapse
Affiliation(s)
- Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Dagmara Bialy
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Fenja Anderson
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Franziska Rother
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Thalea Koithan
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Kathrin Rudolph
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Ute Prank
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anne Binz
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Stefanie Hügel
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rob C. Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Enno Hartmann
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Rudolf Bauerfeind
- Research Core Unit Laser Microscopy, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
29
|
Milev MP, Yao X, Berthoux L, Mouland AJ. Impacts of virus-mediated manipulation of host Dynein. DYNEINS 2018. [PMCID: PMC7150161 DOI: 10.1016/b978-0-12-809470-9.00010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In general viruses' modus operandi to propagate is achieved by the co-opting host cell components, membranes, proteins, and machineries to their advantage. This is true for virtually every aspect of a virus' replication cycle from virus entry to the budding or release of progeny virus particles. In this chapter, we will discuss new information on the impacts of virus-mediated manipulation of Dynein motor complexes and associated machineries and factors. We will highlight how these host cell components impact on pathogenicity and immune responses, as many of the virus-mediated hijacked components also play pivotal roles in immune responses to pathogen insult. There are several comprehensive reviews that define virus–Dynein interactions including the first edition of this book that describes how viruses manipulate the host cell machineries their advantage. An updated table is included to summarize these virus–host interactions. Notably, barriers to intracellular translocation represent major hurdles to viral components during de novo infection and during active replication and the generation of progeny virus particles. Clearly, the subversion of host cell molecular motor protein activities takes advantage of constitutive and regulated membrane trafficking events and will target virus components to intracytoplasmic locales and membrane assembly. Broadening our understanding of the interplay between viruses, Dynein and the cytoskeleton will likely inform on new types of therapies. Continual enhancement of the breadth of new information on how viruses manipulate host cell biology will inevitably aid in the identification of new targets that can be poisoned to block old, new, and emerging viruses alike in their tracks.
Collapse
|
30
|
Human Embryonic Stem Cell-Derived Neurons Are Highly Permissive for Varicella-Zoster Virus Lytic Infection. J Virol 2017; 92:JVI.01108-17. [PMID: 29046461 DOI: 10.1128/jvi.01108-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/03/2017] [Indexed: 12/31/2022] Open
Abstract
Varicella-zoster virus (VZV) is highly cell associated when grown in culture and has a much higher (4,000- to 20,000-fold increased) particle-to-PFU ratio in vitro than herpes simplex virus (HSV). In contrast, VZV is highly infectious in vivo by airborne transmission. Neurons are major targets for VZV in vivo; in neurons, the virus can establish latency and reactivate to produce infectious virus. Using neurons derived from human embryonic stem cells (hESC) and cell-free wild-type (WT) VZV, we demonstrated that neurons are nearly 100 times more permissive for WT VZV infection than very-early-passage human embryonic lung cells or MRC-5 diploid human fibroblasts, the cells used for vaccine production or virus isolation. The peak titers achieved after infection were ∼10-fold higher in human neurons than in MRC-5 cells, and the viral genome copy number-to-PFU ratio for VZV in human neurons was 500, compared with 50,000 for MRC-5 cells. Thus, VZV may not necessarily have a higher particle-to-PFU ratio than other herpesviruses; instead, the cells previously used to propagate virus in vitro may have been suboptimal. Furthermore, based on electron microscopy, neurons infected with VZV produced fewer defective or incomplete viral particles than MRC-5 cells. Our data suggest that neurons derived from hESC may have advantages compared to other cells for studies of VZV pathogenesis, for obtaining stocks of virus with high titers, and for isolating VZV from clinical specimens.IMPORTANCE Varicella-zoster virus (VZV) causes chickenpox and shingles. Cell-free VZV has been difficult to obtain, both for in vitro studies and for vaccine production. While numerous cells lines have been tested for their ability to produce high titers of VZV, the number of total virus particles relative to the number of viral particles that can form plaques in culture has been reported to be extremely high relative to that in other viruses. We show that VZV grows to much higher titers in human neurons than in other cell types in vitro and that the number of total virus genomes relative to the number of viral particles that can form plaques in culture is much lower in human neurons than other cultured cells. These findings indicate that human neurons may be useful for studying VZV in vitro, for growing preparations of virus with high titers, and for isolating the virus from human samples.
Collapse
|
31
|
Buch A, Müller O, Ivanova L, Döhner K, Bialy D, Bosse JB, Pohlmann A, Binz A, Hegemann M, Nagel CH, Koltzenburg M, Viejo-Borbolla A, Rosenhahn B, Bauerfeind R, Sodeik B. Inner tegument proteins of Herpes Simplex Virus are sufficient for intracellular capsid motility in neurons but not for axonal targeting. PLoS Pathog 2017; 13:e1006813. [PMID: 29284065 PMCID: PMC5761964 DOI: 10.1371/journal.ppat.1006813] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/10/2018] [Accepted: 12/14/2017] [Indexed: 02/07/2023] Open
Abstract
Upon reactivation from latency and during lytic infections in neurons, alphaherpesviruses assemble cytosolic capsids, capsids associated with enveloping membranes, and transport vesicles harboring fully enveloped capsids. It is debated whether capsid envelopment of herpes simplex virus (HSV) is completed in the soma prior to axonal targeting or later, and whether the mechanisms are the same in neurons derived from embryos or from adult hosts. We used HSV mutants impaired in capsid envelopment to test whether the inner tegument proteins pUL36 or pUL37 necessary for microtubule-mediated capsid transport were sufficient for axonal capsid targeting in neurons derived from the dorsal root ganglia of adult mice. Such neurons were infected with HSV1-ΔUL20 whose capsids recruited pUL36 and pUL37, with HSV1-ΔUL37 whose capsids associate only with pUL36, or with HSV1-ΔUL36 that assembles capsids lacking both proteins. While capsids of HSV1-ΔUL20 were actively transported along microtubules in epithelial cells and in the somata of neurons, those of HSV1-ΔUL36 and -ΔUL37 could only diffuse in the cytoplasm. Employing a novel image analysis algorithm to quantify capsid targeting to axons, we show that only a few capsids of HSV1-ΔUL20 entered axons, while vesicles transporting gD utilized axonal transport efficiently and independently of pUL36, pUL37, or pUL20. Our data indicate that capsid motility in the somata of neurons mediated by pUL36 and pUL37 does not suffice for targeting capsids to axons, and suggest that capsid envelopment needs to be completed in the soma prior to targeting of herpes simplex virus to the axons, and to spreading from neurons to neighboring cells.
Collapse
Affiliation(s)
- Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
- DZIF—German Center for Infection Research, Hannover, Germany
| | - Oliver Müller
- Institute for Information Processing, Leibniz University, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Lyudmila Ivanova
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Dagmara Bialy
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Jens B. Bosse
- Heinrich-Pette-Institute, Leibniz-Institute for Experimental Virology, Hamburg, Germany
| | - Anja Pohlmann
- Institute of Virology, Hannover Medical School, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Anne Binz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Maike Hegemann
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | | | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
| | - Bodo Rosenhahn
- Institute for Information Processing, Leibniz University, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Rudolf Bauerfeind
- Research Core Unit Laser Microscopy, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
- DZIF—German Center for Infection Research, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| |
Collapse
|
32
|
Budida R, Stankov MV, Döhner K, Buch A, Panayotova-Dimitrova D, Tappe KA, Pohlmann A, Sodeik B, Behrens GMN. Herpes simplex virus 1 interferes with autophagy of murine dendritic cells and impairs their ability to stimulate CD8 + T lymphocytes. Eur J Immunol 2017; 47:1819-1834. [PMID: 28771693 DOI: 10.1002/eji.201646908] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/31/2017] [Accepted: 08/01/2017] [Indexed: 12/17/2022]
Abstract
The MHC class I presentation is responsible for the presentation of viral proteins to CD8+ T lymphocytes and mainly depends on the classical antigen processing pathway. Recently, a second pathway involving autophagy has been implicated in this process. Here, we show an increase in the capacity of murine dendritic cells (DCs) to present viral antigens on MHC class I after infection with a mutant herpes simplex virus 1 (HSV-1-Δ34.5), lacking infected cell protein 34.5 (ICP34.5), when compared to its parental HSV-1 strain. The ICP34.5 protein counteracts host cell translational arrest and suppresses macroautophagy, and the lack of this protein resulted in a low viral protein abundance, which was processed and presented in an efficient way. Our study demonstrates an important role of autophagy in processing endogenous viral proteins in HSV-1-infected DCs.
Collapse
Affiliation(s)
- Ramachandramouli Budida
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Metodi V Stankov
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Kim A Tappe
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Anja Pohlmann
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany.,DZIF-German Center for Infection Research, Hannover-Braunschweig site, Hannover, Germany
| | - Georg M N Behrens
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany.,DZIF-German Center for Infection Research, Hannover-Braunschweig site, Hannover, Germany
| |
Collapse
|
33
|
Herpes Simplex Virus 1 Small Capsomere-Interacting Protein VP26 Regulates Nucleocapsid Maturation. J Virol 2017; 91:JVI.01068-17. [PMID: 28679756 DOI: 10.1128/jvi.01068-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 01/22/2023] Open
Abstract
VP26 is a herpes simplex virus 1 (HSV-1) small capsomere-interacting protein. In this study, we investigated the function of VP26 in HSV-1-infected cells with the following results. (i) The VP26 null mutation significantly impaired incorporation of minor capsid protein UL25 into nucleocapsids (type C capsids) in the nucleus. (ii) The VP26 mutation caused improper localization of UL25 in discrete punctate domains containing multiple capsid proteins (e.g., the VP5 major capsid protein) in the nucleus; these domains corresponded to capsid aggregates. (iii) The VP26 mutation significantly impaired packaging of replicated viral DNA genomes into capsids but had no effect on viral DNA concatemer cleavage. (iv) The VP26 mutation reduced the frequency of type C capsids, which contain viral DNA but not scaffolding proteins, and produced an accumulation of type A capsids, which lack both viral DNA and scaffold proteins, and had no effect on accumulation of type B capsids, which lack viral DNA but retain cleaved scaffold proteins. Collectively, these results indicated that VP26 was required for efficient viral DNA packaging and proper localization of nuclear capsids. The phenotype of the VP26 null mutation was similar to that reported previously of the UL25 null mutation and of UL25 mutations that preclude UL25 binding to capsids. Thus, VP26 appeared to regulate nucleocapsid maturation by promoting incorporation of UL25 into capsids, which is likely to be required for proper capsid nuclear localization.IMPORTANCE HSV-1 VP26 has been reported to be important for viral replication and virulence in cell cultures and/or mouse models. However, little is known about the function of VP26 during HSV-1 replication, in particular, in viral nucleocapsid maturation although HSV-1 nucleocapsids are estimated to contain 900 copies of VP26. In this study, we present data suggesting that VP26 promoted packaging of HSV-1 DNA genomes into capsids by regulating incorporation of capsid protein UL25 into capsids, which was reported to increase stability of the capsid structure. We also showed that VP26 was required for proper localization of capsids in the infected cell nucleus. This is the first report showing that HSV-1 VP26 is a regulator for nucleocapsid maturation.
Collapse
|
34
|
Ganapathiraju MK, Karunakaran KB, Correa-Menéndez J. Predicted protein interactions of IFITMs may shed light on mechanisms of Zika virus-induced microcephaly and host invasion. F1000Res 2016; 5:1919. [PMID: 29333229 PMCID: PMC5747333 DOI: 10.12688/f1000research.9364.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/04/2017] [Indexed: 06/16/2024] Open
Abstract
After the first reported case of Zika virus (ZIKV) in Brazil, in 2015, a significant increase in the reported cases of microcephaly was observed. Microcephaly is a neurological condition in which the infant's head is significantly smaller with complications in brain development. Recently, two small membrane-associated interferon-inducible transmembrane proteins (IFITM1 and IFITM3) have been shown to repress members of the flaviviridae family which includes ZIKV. However, the exact mechanisms leading to the inhibition of the virus are yet unknown. Here, we assembled an interactome of IFITM1 and IFITM3 with known protein-protein interactions (PPIs) collected from publicly available databases and novel PPIs predicted using the High-confidence Protein-Protein Interaction Prediction (HiPPIP) model. We analyzed the functional and pathway associations of the interacting proteins, and found that there are several immunity pathways (toll-like receptor signaling, cd28 signaling in T-helper cells, crosstalk between dendritic cells and natural killer cells), neuronal pathways (axonal guidance signaling, neural tube closure and actin cytoskeleton signaling) and developmental pathways (neural tube closure, embryonic skeletal system development) that are associated with these interactors. Our novel PPIs associate cilia dysfunction in ependymal cells to microcephaly, and may also shed light on potential targets of ZIKV for host invasion by immunosuppression and cytoskeletal rearrangements. These results could help direct future research in elucidating the mechanisms underlying host defense to ZIKV and other flaviviruses.
Collapse
Affiliation(s)
- Madhavi K. Ganapathiraju
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kalyani B. Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India
| | | |
Collapse
|
35
|
Ganapathiraju MK, Karunakaran KB, Correa-Menéndez J. Predicted protein interactions of IFITMs may shed light on mechanisms of Zika virus-induced microcephaly and host invasion. F1000Res 2016; 5:1919. [PMID: 29333229 PMCID: PMC5747333 DOI: 10.12688/f1000research.9364.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/04/2017] [Indexed: 12/22/2022] Open
Abstract
After the first reported case of Zika virus (ZIKV) in Brazil, in 2015, a significant increase in the reported cases of microcephaly was observed. Microcephaly is a neurological condition in which the infant’s head is significantly smaller with complications in brain development. Recently, two small membrane-associated interferon-inducible transmembrane proteins (IFITM1 and IFITM3) have been shown to repress members of the flaviviridae family which includes ZIKV. However, the exact mechanisms leading to the inhibition of the virus are yet unknown. Here, we assembled an interactome of IFITM1 and IFITM3 with known protein-protein interactions (PPIs) collected from publicly available databases and novel PPIs predicted using the High-confidence Protein-Protein Interaction Prediction (HiPPIP) model. We analyzed the functional and pathway associations of the interacting proteins, and found that there are several immunity pathways (toll-like receptor signaling, cd28 signaling in T-helper cells, crosstalk between dendritic cells and natural killer cells), neuronal pathways (axonal guidance signaling, neural tube closure and actin cytoskeleton signaling) and developmental pathways (neural tube closure, embryonic skeletal system development) that are associated with these interactors. Our novel PPIs associate cilia dysfunction in ependymal cells to microcephaly, and may also shed light on potential targets of ZIKV for host invasion by immunosuppression and cytoskeletal rearrangements. These results could help direct future research in elucidating the mechanisms underlying host defense to ZIKV and other flaviviruses.
Collapse
Affiliation(s)
- Madhavi K Ganapathiraju
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India
| | | |
Collapse
|
36
|
Ashford P, Hernandez A, Greco TM, Buch A, Sodeik B, Cristea IM, Grünewald K, Shepherd A, Topf M. HVint: A Strategy for Identifying Novel Protein-Protein Interactions in Herpes Simplex Virus Type 1. Mol Cell Proteomics 2016; 15:2939-53. [PMID: 27384951 PMCID: PMC5013309 DOI: 10.1074/mcp.m116.058552] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Indexed: 11/12/2022] Open
Abstract
Human herpesviruses are widespread human pathogens with a remarkable impact on worldwide public health. Despite intense decades of research, the molecular details in many aspects of their function remain to be fully characterized. To unravel the details of how these viruses operate, a thorough understanding of the relationships between the involved components is key. Here, we present HVint, a novel protein-protein intraviral interaction resource for herpes simplex virus type 1 (HSV-1) integrating data from five external sources. To assess each interaction, we used a scoring scheme that takes into consideration aspects such as the type of detection method and the number of lines of evidence. The coverage of the initial interactome was further increased using evolutionary information, by importing interactions reported for other human herpesviruses. These latter interactions constitute, therefore, computational predictions for potential novel interactions in HSV-1. An independent experimental analysis was performed to confirm a subset of our predicted interactions. This subset covers proteins that contribute to nuclear egress and primary envelopment events, including VP26, pUL31, pUL40, and the recently characterized pUL32 and pUL21. Our findings support a coordinated crosstalk between VP26 and proteins such as pUL31, pUS9, and the CSVC complex, contributing to the development of a model describing the nuclear egress and primary envelopment pathways of newly synthesized HSV-1 capsids. The results are also consistent with recent findings on the involvement of pUL32 in capsid maturation and early tegumentation events. Further, they open the door to new hypotheses on virus-specific regulators of pUS9-dependent transport. To make this repository of interactions readily accessible for the scientific community, we also developed a user-friendly and interactive web interface. Our approach demonstrates the power of computational predictions to assist in the design of targeted experiments for the discovery of novel protein-protein interactions.
Collapse
Affiliation(s)
- Paul Ashford
- From the: ‡Institute of Structural and Molecular Biology, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, UK
| | - Anna Hernandez
- From the: ‡Institute of Structural and Molecular Biology, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, UK; §Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Todd Michael Greco
- ¶Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544
| | - Anna Buch
- ‖Institute of Virology, Hannover Medical School, OE 4310, Carl-Neuberg-Str. 1, D-30623, Hannover, Germany
| | - Beate Sodeik
- ‖Institute of Virology, Hannover Medical School, OE 4310, Carl-Neuberg-Str. 1, D-30623, Hannover, Germany
| | - Ileana Mihaela Cristea
- ¶Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544;
| | - Kay Grünewald
- §Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Adrian Shepherd
- From the: ‡Institute of Structural and Molecular Biology, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, UK
| | - Maya Topf
- From the: ‡Institute of Structural and Molecular Biology, Birkbeck College, University of London, Malet Street, London, WC1E 7HX, UK;
| |
Collapse
|
37
|
Conserved Tryptophan Motifs in the Large Tegument Protein pUL36 Are Required for Efficient Secondary Envelopment of Herpes Simplex Virus Capsids. J Virol 2016; 90:5368-5383. [PMID: 27009950 DOI: 10.1128/jvi.03167-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/15/2016] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Herpes simplex virus (HSV) replicates in the skin and mucous membranes, and initiates lytic or latent infections in sensory neurons. Assembly of progeny virions depends on the essential large tegument protein pUL36 of 3,164 amino acid residues that links the capsids to the tegument proteins pUL37 and VP16. Of the 32 tryptophans of HSV-1-pUL36, the tryptophan-acidic motifs (1766)WD(1767) and (1862)WE(1863) are conserved in all HSV-1 and HSV-2 isolates. Here, we characterized the role of these motifs in the HSV life cycle since the rare tryptophans often have unique roles in protein function due to their large hydrophobic surface. The infectivity of the mutants HSV-1(17(+))Lox-pUL36-WD/AA-WE/AA and HSV-1(17(+))Lox-CheVP26-pUL36-WD/AA-WE/AA, in which the capsid has been tagged with the fluorescent protein Cherry, was significantly reduced. Quantitative electron microscopy shows that there were a larger number of cytosolic capsids and fewer enveloped virions compared to their respective parental strains, indicating a severe impairment in secondary capsid envelopment. The capsids of the mutant viruses accumulated in the perinuclear region around the microtubule-organizing center and were not dispersed to the cell periphery but still acquired the inner tegument proteins pUL36 and pUL37. Furthermore, cytoplasmic capsids colocalized with tegument protein VP16 and, to some extent, with tegument protein VP22 but not with the envelope glycoprotein gD. These results indicate that the unique conserved tryptophan-acidic motifs in the central region of pUL36 are required for efficient targeting of progeny capsids to the membranes of secondary capsid envelopment and for efficient virion assembly. IMPORTANCE Herpesvirus infections give rise to severe animal and human diseases, especially in young, immunocompromised, and elderly individuals. The structural hallmark of herpesvirus virions is the tegument, which contains evolutionarily conserved proteins that are essential for several stages of the herpesvirus life cycle. Here we characterized two conserved tryptophan-acidic motifs in the central region of the large tegument protein pUL36 of herpes simplex virus. When we mutated these motifs, secondary envelopment of cytosolic capsids and the production of infectious particles were severely impaired. Our data suggest that pUL36 and its homologs in other herpesviruses, and in particular such tryptophan-acidic motifs, could provide attractive targets for the development of novel drugs to prevent herpesvirus assembly and spread.
Collapse
|
38
|
Jovasevic V, Naghavi MH, Walsh D. Microtubule plus end-associated CLIP-170 initiates HSV-1 retrograde transport in primary human cells. J Cell Biol 2016; 211:323-37. [PMID: 26504169 PMCID: PMC4621836 DOI: 10.1083/jcb.201505123] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Herpes simplex virus particles that enter the cell do not randomly associate with microtubule filaments, but require plus end–binding proteins EB1, CLIP-170, and dynactin to initiate retrograde transport to the nucleus. Dynamic microtubules (MTs) continuously explore the intracellular environment and, through specialized plus end–tracking proteins (+TIPs), engage a variety of targets. However, the nature of cargoes that require +TIP-mediated capture for their movement on MTs remains poorly understood. Using RNA interference and dominant-negative approaches, combined with live cell imaging, we show that herpes simplex virus particles that have entered primary human cells exploit a +TIP complex comprising end-binding protein 1 (EB1), cytoplasmic linker protein 170 (CLIP-170), and dynactin-1 (DCTN1) to initiate retrograde transport. Depletion of these +TIPs completely blocked post-entry long-range transport of virus particles and suppressed infection ∼5,000-fold, whereas transferrin uptake, early endosome organization, and dynein-dependent movement of lysosomes and mitochondria remained unaffected. These findings provide the first insights into the earliest stages of viral engagement of MTs through specific +TIPs, akin to receptors, with therapeutic implications, and identify herpesvirus particles as one of a very limited number of cargoes absolutely dependent on CLIP-170–mediated capture to initiate transport in primary human cells.
Collapse
Affiliation(s)
- Vladimir Jovasevic
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Mojgan H Naghavi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 Department of Biochemistry and Molecular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Derek Walsh
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 Department of Microbiology, School of Medicine, New York University, New York, NY 10016
| |
Collapse
|
39
|
Cytoplasmic isoforms of Kaposi sarcoma herpesvirus LANA recruit and antagonize the innate immune DNA sensor cGAS. Proc Natl Acad Sci U S A 2016; 113:E1034-43. [PMID: 26811480 DOI: 10.1073/pnas.1516812113] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The latency-associated nuclear antigen (LANA) of Kaposi sarcoma herpesvirus (KSHV) is mainly localized and functions in the nucleus of latently infected cells, playing a pivotal role in the replication and maintenance of latent viral episomal DNA. In addition, N-terminally truncated cytoplasmic isoforms of LANA, resulting from internal translation initiation, have been reported, but their function is unknown. Using coimmunoprecipitation and MS, we found the cGMP-AMP synthase (cGAS), an innate immune DNA sensor, to be a cellular interaction partner of cytoplasmic LANA isoforms. By directly binding to cGAS, LANA, and particularly, a cytoplasmic isoform, inhibit the cGAS-STING-dependent phosphorylation of TBK1 and IRF3 and thereby antagonize the cGAS-mediated restriction of KSHV lytic replication. We hypothesize that cytoplasmic forms of LANA, whose expression increases during lytic replication, inhibit cGAS to promote the reactivation of the KSHV from latency. This observation points to a novel function of the cytoplasmic isoforms of LANA during lytic replication and extends the function of LANA from its role during latency to the lytic replication cycle.
Collapse
|
40
|
Jin F, Ma K, Chen M, Zou M, Wu Y, Li F, Wang Y. Pentagalloylglucose Blocks the Nuclear Transport and the Process of Nucleocapsid Egress to Inhibit HSV-1 Infection. Jpn J Infect Dis 2015; 69:135-42. [PMID: 26166506 DOI: 10.7883/yoken.jjid.2015.137] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Herpes simplex virus type 1 (HSV-1), a widespread virus, causes a variety of human viral diseases worldwide. The serious threat of drug-resistance highlights the extreme urgency to develop novel antiviral drugs with different mechanisms of action. Pentagalloylglucose (PGG) is a natural polyphenolic compound with significant anti-HSV activity; however, the mechanisms underlying its antiviral activity need to be defined by further studies. In this study, we found that PGG treatment delays the nuclear transport process of HSV-1 particles by inhibiting the upregulation of dynein (a cellular major motor protein) induced by HSV-1 infection. Furthermore, PGG treatment affects the nucleocapsid egress of HSV-1 by inhibiting the expression and disrupting the cellular localization of pEGFP-UL31 and pEGFP-UL34, which are indispensable for HSV-1 nucleocapsid egress from the nucleus. However, the over-expression of pEGFP-UL31 and pEGFP-UL34 could decrease the antiviral effect of PGG. In this study, for the first time, the antiviral activity of PGG against acyclovir-resistant virus was demonstrated in vitro, and the possible mechanisms of its anti-HSV activities were identified based on the inhibition of nuclear transport and nucleocapsid egress in HSV-1. It was further confirmed that PGG could be a promising candidate for HSV therapy, especially for drug-resistant strains.
Collapse
Affiliation(s)
- Fujun Jin
- Guangzhou Jinan Biomedicine Research and Development Center, National Engineering Research Center of Genetic Medicine, Jinan University
| | | | | | | | | | | | | |
Collapse
|
41
|
Kennedy PGE, Rovnak J, Badani H, Cohrs RJ. A comparison of herpes simplex virus type 1 and varicella-zoster virus latency and reactivation. J Gen Virol 2015; 96:1581-602. [PMID: 25794504 DOI: 10.1099/vir.0.000128] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1; human herpesvirus 1) and varicella-zoster virus (VZV; human herpesvirus 3) are human neurotropic alphaherpesviruses that cause lifelong infections in ganglia. Following primary infection and establishment of latency, HSV-1 reactivation typically results in herpes labialis (cold sores), but can occur frequently elsewhere on the body at the site of primary infection (e.g. whitlow), particularly at the genitals. Rarely, HSV-1 reactivation can cause encephalitis; however, a third of the cases of HSV-1 encephalitis are associated with HSV-1 primary infection. Primary VZV infection causes varicella (chickenpox) following which latent virus may reactivate decades later to produce herpes zoster (shingles), as well as an increasingly recognized number of subacute, acute and chronic neurological conditions. Following primary infection, both viruses establish a latent infection in neuronal cells in human peripheral ganglia. However, the detailed mechanisms of viral latency and reactivation have yet to be unravelled. In both cases latent viral DNA exists in an 'end-less' state where the ends of the virus genome are joined to form structures consistent with unit length episomes and concatemers, from which viral gene transcription is restricted. In latently infected ganglia, the most abundantly detected HSV-1 RNAs are the spliced products originating from the primary latency associated transcript (LAT). This primary LAT is an 8.3 kb unstable transcript from which two stable (1.5 and 2.0 kb) introns are spliced. Transcripts mapping to 12 VZV genes have been detected in human ganglia removed at autopsy; however, it is difficult to ascribe these as transcripts present during latent infection as early-stage virus reactivation may have transpired in the post-mortem time period in the ganglia. Nonetheless, low-level transcription of VZV ORF63 has been repeatedly detected in multiple ganglia removed as close to death as possible. There is increasing evidence that HSV-1 and VZV latency is epigenetically regulated. In vitro models that permit pathway analysis and identification of both epigenetic modulations and global transcriptional mechanisms of HSV-1 and VZV latency hold much promise for our future understanding in this complex area. This review summarizes the molecular biology of HSV-1 and VZV latency and reactivation, and also presents future directions for study.
Collapse
Affiliation(s)
- Peter G E Kennedy
- 1Institute of Infection, Immunity and Inflammation, University of Glasgow, Garscube Campus, Glasgow G61 1QH, UK
| | - Joel Rovnak
- 2Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80521, USA
| | - Hussain Badani
- 3Department of Neurology, University of Colorado Medical School, Aurora, CO 80045, USA
| | - Randall J Cohrs
- 3Department of Neurology, University of Colorado Medical School, Aurora, CO 80045, USA 4Department of Microbiology, University of Colorado Medical School, Aurora, CO 80045, USA
| |
Collapse
|
42
|
Kati S, Hage E, Mynarek M, Ganzenmueller T, Indenbirken D, Grundhoff A, Schulz TF. Generation of high-titre virus stocks using BrK.219, a B-cell line infected stably with recombinant Kaposi's sarcoma-associated herpesvirus. J Virol Methods 2015; 217:79-86. [PMID: 25736227 DOI: 10.1016/j.jviromet.2015.02.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 01/13/2015] [Accepted: 02/23/2015] [Indexed: 11/15/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is a gamma-2-lymphotropic human oncogenic herpesvirus associated with Kaposi's sarcoma (KS) and two B-cell lymphoproliferative diseases, primary effusion lymphoma (PEL) and multicentric Castleman's disease (MCD). KSHV establishes latency soon after infection in vivo and in vitro. Consequently, it is technically difficult to generate high-titre virus stocks required for infection experiments in tissue culture. Currently used methods of KSHV stock production involve induction of the lytic/productive cycle in PEL cell lines or in adherent cell lines harbouring recombinant KSHV genomes. In this study, the BJAB-derived B-cell line BrK.219, which is infected latently with a recombinant KSHV (rKSHV.219), is used to produce high-titre virus stocks. BrK.219 cells enter the lytic KSHV replication cycle upon cross-linking of B-cell receptors (BCRs) with anti-IgM antibodies without the need for additional, potentially toxic chemical inducers. High cell concentrations can be cultured and induced easily in spinner flasks, saving time and resources. The established protocol allows the generation of KSHV virus stocks with titres of up to 10(6) IU/ml in unconcentrated culture supernatants, representing a 10(3)-10(4)-fold improvement compared to conventional methods.
Collapse
Affiliation(s)
- Semra Kati
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str.1, Hannover D-30625, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Elias Hage
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str.1, Hannover D-30625, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Martin Mynarek
- Department of Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Tina Ganzenmueller
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str.1, Hannover D-30625, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Daniela Indenbirken
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Adam Grundhoff
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str.1, Hannover D-30625, Germany; German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany.
| |
Collapse
|
43
|
Krawczyk A, Dirks M, Kasper M, Buch A, Dittmer U, Giebel B, Wildschütz L, Busch M, Goergens A, Schneweis KE, Eis-Hübinger AM, Sodeik B, Heiligenhaus A, Roggendorf M, Bauer D. Prevention of herpes simplex virus induced stromal keratitis by a glycoprotein B-specific monoclonal antibody. PLoS One 2015; 10:e0116800. [PMID: 25587898 PMCID: PMC4294644 DOI: 10.1371/journal.pone.0116800] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 12/15/2014] [Indexed: 10/24/2022] Open
Abstract
The increasing incidence of acyclovir (ACV) and multidrug-resistant strains in patients with corneal HSV-1 infections leading to Herpetic Stromal Keratitis (HSK) is a major health problem in industrialized countries and often results in blindness. To overcome this obstacle, we have previously developed an HSV-gB-specific monoclonal antibody (mAb 2c) that proved to be highly protective in immunodeficient NOD/SCID-mice towards genital infections. In the present study, we examined the effectivity of mAb 2c in preventing the immunopathological disease HSK in the HSK BALB/c mouse model. Therefore, mice were inoculated with HSV-1 strain KOS on the scarified cornea to induce HSK and subsequently either systemically or topically treated with mAb 2c. Systemic treatment was performed by intravenous administration of mAb 2c 24 h prior to infection (pre-exposure prophylaxis) or 24, 40, and 56 hours after infection (post-exposure immunotherapy). Topical treatment was performed by periodical inoculations (5 times per day) of antibody-containing eye drops as control, starting at 24 h post infection. Systemic antibody treatment markedly reduced viral loads at the site of infection and completely protected mice from developing HSK. The administration of the antiviral antibody prior or post infection was equally effective. Topical treatment had no improving effect on the severity of HSK. In conclusion, our data demonstrate that mAb 2c proved to be an excellent drug for the treatment of corneal HSV-infections and for prevention of HSK and blindness. Moreover, the humanized counterpart (mAb hu2c) was equally effective in protecting mice from HSV-induced HSK when compared to the parental mouse antibody. These results warrant the future development of this antibody as a novel approach for the treatment of corneal HSV-infections in humans.
Collapse
Affiliation(s)
- Adalbert Krawczyk
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- * E-mail:
| | - Miriam Dirks
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maren Kasper
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Ulf Dittmer
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Lena Wildschütz
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Martin Busch
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Andre Goergens
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Karl E. Schneweis
- Institute of Virology, University Medical Center Bonn, Bonn, Germany
| | | | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Arnd Heiligenhaus
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Michael Roggendorf
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dirk Bauer
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| |
Collapse
|
44
|
Anderson F, Savulescu AF, Rudolph K, Schipke J, Cohen I, Ibiricu I, Rotem A, Grünewald K, Sodeik B, Harel A. Targeting of viral capsids to nuclear pores in a cell-free reconstitution system. Traffic 2014; 15:1266-81. [PMID: 25131140 DOI: 10.1111/tra.12209] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/13/2014] [Accepted: 08/13/2014] [Indexed: 11/28/2022]
Abstract
Many viruses deliver their genomes into the nucleoplasm for viral transcription and replication. Here, we describe a novel cell-free system to elucidate specific interactions between viruses and nuclear pore complexes (NPCs). Nuclei reconstituted in vitro from egg extracts of Xenopus laevis, an established biochemical system to decipher nuclear functions, were incubated with GFP-tagged capsids of herpes simplex virus, an alphaherpesvirus replicating in the nucleus. Capsid binding to NPCs was analyzed using fluorescence and field emission scanning electron microscopy. Tegument-free capsids or viral capsids exposing inner tegument proteins on their surface bound to nuclei, while capsids inactivated by a high-salt treatment or covered by inner and outer tegument showed less binding. There was little binding of the four different capsid types to nuclei lacking functional NPCs. This novel approach provides a powerful system to elucidate the molecular mechanisms that enable viral structures to engage with NPCs. Furthermore, this assay could be expanded to identify molecular cues triggering viral genome uncoating and nuclear import of viral genomes.
Collapse
Affiliation(s)
- Fenja Anderson
- Institute of Virology, OE 5230, Hannover Medical School, Carl-Neuberg-Straße 1, D-30623, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Herpes simplex virus internalization into epithelial cells requires Na+/H+ exchangers and p21-activated kinases but neither clathrin- nor caveolin-mediated endocytosis. J Virol 2014; 88:13378-95. [PMID: 25210183 DOI: 10.1128/jvi.03631-13] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED Herpes simplex virus 1 (HSV-1) is an alphaherpesvirus that has been reported to infect some epithelial cell types by fusion at the plasma membrane but others by endocytosis. To determine the molecular mechanisms of productive HSV-1 cell entry, we perturbed key endocytosis host factors using specific inhibitors, RNA interference (RNAi), or overexpression of dominant negative proteins and investigated their effects on HSV-1 infection in the permissive epithelial cell lines Vero, HeLa, HEp-2, and PtK2. HSV-1 internalization required neither endosomal acidification nor clathrin- or caveolin-mediated endocytosis. In contrast, HSV-1 gene expression and internalization were significantly reduced after treatment with 5-(N-ethyl-N-isopropyl)amiloride (EIPA). EIPA blocks the activity of Na(+)/H(+) exchangers, which are plasma membrane proteins implicated in all forms of macropinocytosis. HSV-1 internalization furthermore required the function of p21-activated kinases that contribute to macropinosome formation. However, in contrast to some forms of macropinocytosis, HSV-1 did not enlist the activities of protein kinase C (PKC), tyrosine kinases, C-terminal binding protein 1, or dynamin to activate its internalization. These data suggest that HSV-1 depends on Na(+)/H(+) exchangers and p21-activated kinases either for macropinocytosis or for local actin rearrangements required for fusion at the plasma membrane or subsequent passage through the actin cortex underneath the plasma membrane. IMPORTANCE After initial replication in epithelial cells, herpes simplex viruses (HSVs) establish latent infections in neurons innervating these regions. Upon primary infection and reactivation from latency, HSVs cause many human skin and neurological diseases, particularly in immunocompromised hosts, despite the availability of effective antiviral drugs. Many viruses use macropinocytosis for virus internalization, and many host factors mediating this entry route have been identified, although the specific perturbation profiles vary for different host and viral cargo. In addition to an established entry pathway via acidic endosomes, we show here that HSV-1 internalization depended on sodium-proton exchangers at the plasma membrane and p21-activated kinases. These results suggest that HSV-1 requires a reorganization of the cortical actin cytoskeleton, either for productive cell entry via pH-independent fusion from macropinosomes or for fusion at the plasma membrane, and subsequent cytosolic passage to microtubules that mediate capsid transport to the nucleus for genome uncoating and replication.
Collapse
|
46
|
Blyuss KB, Nicholson LB. Understanding the roles of activation threshold and infections in the dynamics of autoimmune disease. J Theor Biol 2014; 375:13-20. [PMID: 25150457 DOI: 10.1016/j.jtbi.2014.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/30/2014] [Accepted: 08/11/2014] [Indexed: 12/21/2022]
Abstract
Onset and development of autoimmunity have been attributed to a number of factors, including genetic predisposition, age and different environmental factors. In this paper we discuss mathematical models of autoimmunity with an emphasis on two particular aspects of immune dynamics: breakdown of immune tolerance in response to an infection with a pathogen, and interactions between T cells with different activation thresholds. We illustrate how the explicit account of T cells with different activation thresholds provides a viable model of immune dynamics able to reproduce several types of immune behaviour, including normal clearance of infection, emergence of a chronic state, and development of a recurrent infection with autoimmunity. We discuss a number of open research problems that can be addressed within the same modelling framework.
Collapse
Affiliation(s)
- K B Blyuss
- Department of Mathematics, University of Sussex, Falmer, Brighton BN1 9QH, UK.
| | - L B Nicholson
- School of Cellular and Molecular Medicine & School of Clinical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK.
| |
Collapse
|
47
|
Radtke K, Anderson F, Sodeik B. A precipitation-based assay to analyze interactions of viral particles with cytosolic host factors. Methods Mol Biol 2014; 1144:191-208. [PMID: 24671685 DOI: 10.1007/978-1-4939-0428-0_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Since viruses are obligate intracellular parasites, viral particles, subviral structures, and viral proteins enlist the support of host proteins to foster intracellular transport, viral gene expression, replication, and evasion from antiviral host responses. We have devised a biochemical in vitro method to analyze specific interactions of cytosolic factors with capsids of herpes simplex virus and to characterize host proteins that specifically coprecipitate with different types of viral particles by immunoblotting, mass spectrometry, and immunoelectron microscopy. Our method bridges the gap between assays such as co-immunoprecipitation and yeast-two-hybrid approaches that determine direct binding between individual subunits of protein complexes and microscopy methods that analyze the dynamic interplay between intact viral particles and host factor complexes in intact cells. Our protocol can be extended to functional analyses of herpesvirus capsids and other viral structures with more complex host structures such as microtubule transport, genome uncoating at nuclear pores, or capsid envelopment at host membranes.
Collapse
Affiliation(s)
- Kerstin Radtke
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, 2900 Blvd. Édouard-Montpetit Montréal, QC, Canada
| | | | | |
Collapse
|
48
|
Shaw AE, Brüning-Richardson A, Morrison EE, Bond J, Simpson J, Ross-Smith N, Alpar O, Mertens PPC, Monaghan P. Bluetongue virus infection induces aberrant mitosis in mammalian cells. Virol J 2013; 10:319. [PMID: 24165208 PMCID: PMC3874736 DOI: 10.1186/1743-422x-10-319] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/27/2013] [Indexed: 12/25/2022] Open
Abstract
Background Bluetongue virus (BTV) is an arbovirus that is responsible for ‘bluetongue’, an economically important disease of livestock. Although BTV is well characterised at the protein level, less is known regarding its interaction with host cells. During studies of virus inclusion body formation we observed what appeared to be a large proportion of cells in mitosis. Although the modulation of the cell cycle is well established for many viruses, this was a novel observation for BTV. We therefore undertook a study to reveal in more depth the impact of BTV upon cell division. Methods We used a confocal microscopy approach to investigate the localisation of BTV proteins in a cellular context with their respective position relative to cellular proteins. In addition, to quantitatively assess the frequency of aberrant mitosis induction by the viral non-structural protein (NS) 2 we utilised live cell imaging to monitor HeLa-mCherry tubulin cells transfected with a plasmid expressing NS2. Results Our data showed that these ‘aberrant mitoses’ can be induced in multiple cell types and by different strains of BTV. Further study confirmed multiplication of the centrosomes, each resulting in a separate mitotic spindle during mitosis. Interestingly, the BTV NS1 protein was strongly localised to the centrosomal regions. In a separate, yet related observation, the BTV NS2 protein was co-localised with the condensed chromosomes to a region suggestive of the kinetochore. Live cell imaging revealed that expression of an EGFP-NS2 fusion protein in HeLa-mCherry tubulin cells also results in mitotic defects. Conclusions We hypothesise that NS2 is a microtubule cargo protein that may inadvertently disrupt the interaction of microtubule tips with the kinetochores during mitosis. Furthermore, the BTV NS1 protein was distinctly localised to a region encompassing the centrosome and may therefore be, at least in part, responsible for the disruption of the centrosome as observed in BTV infected mammalian cells.
Collapse
|
49
|
Zaichick SV, Bohannon KP, Hughes A, Sollars PJ, Pickard GE, Smith GA. The herpesvirus VP1/2 protein is an effector of dynein-mediated capsid transport and neuroinvasion. Cell Host Microbe 2013; 13:193-203. [PMID: 23414759 DOI: 10.1016/j.chom.2013.01.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 11/13/2012] [Accepted: 01/11/2013] [Indexed: 12/23/2022]
Abstract
Microtubule transport of herpesvirus capsids from the cell periphery to the nucleus is imperative for viral replication and, in the case of many alphaherpesviruses, transmission into the nervous system. Using the neuroinvasive herpesvirus, pseudorabies virus (PRV), we show that the viral protein 1/2 (VP1/2) tegument protein associates with the dynein/dynactin microtubule motor complex and promotes retrograde microtubule transport of PRV capsids. Functional activation of VP1/2 requires binding to the capsid protein pUL25 or removal of the capsid-binding domain. A proline-rich sequence within VP1/2 is required for the efficient interaction with the dynein/dynactin microtubule motor complex as well as for PRV virulence and retrograde axon transport in vivo. Additionally, in the absence of infection, functionally active VP1/2 is sufficient to move large surrogate cargoes via the dynein/dynactin microtubule motor complex. Thus, VP1/2 tethers PRV capsids to dynein/dynactin to enhance microtubule transport, neuroinvasion, and pathogenesis.
Collapse
Affiliation(s)
- Sofia V Zaichick
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
50
|
Griffiths SJ, Koegl M, Boutell C, Zenner HL, Crump CM, Pica F, Gonzalez O, Friedel CC, Barry G, Martin K, Craigon MH, Chen R, Kaza LN, Fossum E, Fazakerley JK, Efstathiou S, Volpi A, Zimmer R, Ghazal P, Haas J. A systematic analysis of host factors reveals a Med23-interferon-λ regulatory axis against herpes simplex virus type 1 replication. PLoS Pathog 2013; 9:e1003514. [PMID: 23950709 PMCID: PMC3738494 DOI: 10.1371/journal.ppat.1003514] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/24/2013] [Indexed: 11/24/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a neurotropic virus causing vesicular oral or genital skin lesions, meningitis and other diseases particularly harmful in immunocompromised individuals. To comprehensively investigate the complex interaction between HSV-1 and its host we combined two genome-scale screens for host factors (HFs) involved in virus replication. A yeast two-hybrid screen for protein interactions and a RNA interference (RNAi) screen with a druggable genome small interfering RNA (siRNA) library confirmed existing and identified novel HFs which functionally influence HSV-1 infection. Bioinformatic analyses found the 358 HFs were enriched for several pathways and multi-protein complexes. Of particular interest was the identification of Med23 as a strongly anti-viral component of the largely pro-viral Mediator complex, which links specific transcription factors to RNA polymerase II. The anti-viral effect of Med23 on HSV-1 replication was confirmed in gain-of-function gene overexpression experiments, and this inhibitory effect was specific to HSV-1, as a range of other viruses including Vaccinia virus and Semliki Forest virus were unaffected by Med23 depletion. We found Med23 significantly upregulated expression of the type III interferon family (IFN-λ) at the mRNA and protein level by directly interacting with the transcription factor IRF7. The synergistic effect of Med23 and IRF7 on IFN-λ induction suggests this is the major transcription factor for IFN-λ expression. Genotypic analysis of patients suffering recurrent orofacial HSV-1 outbreaks, previously shown to be deficient in IFN-λ secretion, found a significant correlation with a single nucleotide polymorphism in the IFN-λ3 (IL28b) promoter strongly linked to Hepatitis C disease and treatment outcome. This paper describes a link between Med23 and IFN-λ, provides evidence for the crucial role of IFN-λ in HSV-1 immune control, and highlights the power of integrative genome-scale approaches to identify HFs critical for disease progression and outcome.
Collapse
Affiliation(s)
| | - Manfred Koegl
- Preclinical Target Development and Genomics and Proteomics Core Facilities, German Cancer Research Center, Heidelberg, Germany
| | - Chris Boutell
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Helen L. Zenner
- Division of Virology, Department of Pathology Cambridge University, Cambridge, United Kingdom
| | - Colin M. Crump
- Division of Virology, Department of Pathology Cambridge University, Cambridge, United Kingdom
| | | | - Orland Gonzalez
- Institute for Informatics, Ludwig-Maximilians Universität München, München, Germany
| | - Caroline C. Friedel
- Institute for Informatics, Ludwig-Maximilians Universität München, München, Germany
| | - Gerald Barry
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Kim Martin
- Division of Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Marie H. Craigon
- Division of Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Rui Chen
- Division of Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Lakshmi N. Kaza
- Division of Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Even Fossum
- Division of Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - John K. Fazakerley
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Stacey Efstathiou
- Division of Virology, Department of Pathology Cambridge University, Cambridge, United Kingdom
| | | | - Ralf Zimmer
- Institute for Informatics, Ludwig-Maximilians Universität München, München, Germany
| | - Peter Ghazal
- Division of Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Systems Biology at Edinburgh, University of Edinburgh, Edinburgh, United Kingdom
| | - Jürgen Haas
- Division of Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Max von Pettenkofer Institut, Ludwig-Maximilians Universität München, München, Germany
| |
Collapse
|