1
|
Spear PG. Opportunities, Technology, and the Joy of Discovery. Annu Rev Virol 2022; 9:1-17. [PMID: 35363539 DOI: 10.1146/annurev-virology-100520-012840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
My grandparents were immigrants. My paternal grandfather was illiterate. Yet my parents were able to complete college and to become teachers. I had a conventional upbringing in a small town in Florida, graduating from high school in 1960. I was fortunate enough to graduate cum laude from Florida State University and to earn other credentials leading to faculty positions at outstanding institutions of higher education: the University of Chicago and Northwestern University. At a time when women were rarely the leaders of research groups, I was able to establish a well-funded research program and to make contributions to our understanding of viral entry into cells. My best research was done after I became confident enough to seek productive interactions with collaborators. I am grateful for the collaborators and collaborations that moved our field forward and for my trainees who have gone on to successes in many different careers. Expected final online publication date for the Annual Review of Virology, Volume 9 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Patricia G Spear
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA;
| |
Collapse
|
2
|
Mittal A, Chauhan A. Aspects of Biological Replication and Evolution Independent of the Central Dogma: Insights from Protein-Free Vesicular Transformations and Protein-Mediated Membrane Remodeling. J Membr Biol 2022; 255:185-209. [PMID: 35333977 PMCID: PMC8951669 DOI: 10.1007/s00232-022-00230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/06/2022] [Indexed: 11/21/2022]
Abstract
Biological membrane remodeling is central to living systems. In spite of serving as “containers” of whole-living systems and functioning as dynamic compartments within living systems, biological membranes still find a “blue collar” treatment compared to the “white collar” nucleic acids and proteins in biology. This may be attributable to the fact that scientific literature on biological membrane remodeling is only 50 years old compared to ~ 150 years of literature on proteins and a little less than 100 years on nucleic acids. However, recently, evidence for symbiotic origins of eukaryotic cells from data only on biological membranes was reported. This, coupled with appreciation of reproducible amphiphilic self-assemblies in aqueous environments (mimicking replication), has already initiated discussions on origins of life beyond nucleic acids and proteins. This work presents a comprehensive compilation and meta-analyses of data on self-assembly and vesicular transformations in biological membranes—starting from model membranes to establishment of Influenza Hemagglutinin-mediated membrane fusion as a prototypical remodeling system to a thorough comparison between enveloped mammalian viruses and cellular vesicles. We show that viral membrane fusion proteins, in addition to obeying “stoichiometry-driven protein folding”, have tighter compositional constraints on their amino acid occurrences than general-structured proteins, regardless of type/class. From the perspective of vesicular assemblies and biological membrane remodeling (with and without proteins) we find that cellular vesicles are quite different from viruses. Finally, we propose that in addition to pre-existing thermodynamic frameworks, kinetic considerations in de novo formation of metastable membrane structures with available “third-party” constituents (including proteins) were not only crucial for origins of life but also continue to offer morphological replication and/or functional mechanisms in modern life forms, independent of the central dogma.
Collapse
Affiliation(s)
- Aditya Mittal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi, 110016, India. .,Supercomputing Facility for Bioinformatics and Computational Biology (SCFBio), IIT Delhi, Hauz Khas, New Delhi, 110016, India.
| | - Akanksha Chauhan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi (IIT Delhi), Hauz Khas, New Delhi, 110016, India
| |
Collapse
|
3
|
Hutson CL, Kondas AV, Ritter JM, Reed Z, Ostergaard SD, Morgan CN, Gallardo-Romero N, Tansey C, Mauldin MR, Salzer JS, Hughes CM, Goldsmith CS, Carroll D, Olson VA. Teaching a new mouse old tricks: Humanized mice as an infection model for Variola virus. PLoS Pathog 2021; 17:e1009633. [PMID: 34547055 PMCID: PMC8454956 DOI: 10.1371/journal.ppat.1009633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 05/11/2021] [Indexed: 01/12/2023] Open
Abstract
Smallpox, caused by the solely human pathogen Variola virus (VARV), was declared eradicated in 1980. While known VARV stocks are secure, smallpox remains a bioterrorist threat agent. Recent U.S. Food and Drug Administration approval of the first smallpox anti-viral (tecovirimat) therapeutic was a successful step forward in smallpox preparedness; however, orthopoxviruses can become resistant to treatment, suggesting a multi-therapeutic approach is necessary. Animal models are required for testing medical countermeasures (MCMs) and ideally MCMs are tested directly against the pathogen of interest. Since VARV only infects humans, a representative animal model for testing therapeutics directly against VARV remains a challenge. Here we show that three different humanized mice strains are highly susceptible to VARV infection, establishing the first small animal model using VARV. In comparison, the non-humanized, immunosuppressed background mouse was not susceptible to systemic VARV infection. Following an intranasal VARV challenge that mimics the natural route for human smallpox transmission, the virus spread systemically within the humanized mouse before mortality (~ 13 days post infection), similar to the time from exposure to symptom onset for ordinary human smallpox. Our identification of a permissive/representative VARV animal model can facilitate testing of MCMs in a manner consistent with their intended use.
Collapse
Affiliation(s)
- Christina L. Hutson
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ashley V. Kondas
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jana M. Ritter
- Infectious Diseases Pathology Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Zachary Reed
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Sharon Dietz Ostergaard
- Comparative Medicine Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Clint N. Morgan
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Nadia Gallardo-Romero
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Cassandra Tansey
- Comparative Medicine Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Matthew R. Mauldin
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Johanna S. Salzer
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Christine M. Hughes
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Cynthia S. Goldsmith
- Infectious Diseases Pathology Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Darin Carroll
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Victoria A. Olson
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
4
|
Leoni V, Gatta V, Palladini A, Nicoletti G, Ranieri D, Dall'Ora M, Grosso V, Rossi M, Alviano F, Bonsi L, Nanni P, Lollini PL, Campadelli-Fiume G. Systemic delivery of HER2-retargeted oncolytic-HSV by mesenchymal stromal cells protects from lung and brain metastases. Oncotarget 2016; 6:34774-87. [PMID: 26430966 PMCID: PMC4741489 DOI: 10.18632/oncotarget.5793] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/04/2015] [Indexed: 01/01/2023] Open
Abstract
Fully retargeted oncolytic herpes simplex viruses (o-HSVs) gain cancer-specificity from redirection of tropism to cancer-specific receptors, and are non-attenuated. To overcome the hurdles of systemic delivery, and enable oncolytic viruses (o-viruses) to reach metastatic sites, carrier cells are being exploited. Mesenchymal stromal cells (MSCs) were never tested as carriers of retargeted o-viruses, given their scarse-null expression of the cancer-specific receptors. We report that MSCs from different sources can be forcedly infected with a HER2-retargeted oncolytic HSV. Progeny virus spread from MSCs to cancer cells in vitro and in vivo. We evaluated the organ distribution and therapeutic efficacy in two murine models of metastatic cancers, following a single i.v. injection of infected MSCs. As expected, the highest concentration of carrier-cells and of viral genomes was in the lungs. Viral genomes persisted throughout the body for at least two days. The growth of ovarian cancer lung metastases in nude mice was strongly inhibited, and the majority of treated mice appeared metastasis-free. The treatment significantly inhibited also breast cancer metastases to the brain in NSG mice, and reduced by more than one-half the metastatic burden in the brain.
Collapse
Affiliation(s)
- Valerio Leoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Valentina Gatta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Arianna Palladini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Giordano Nicoletti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Dario Ranieri
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Massimiliano Dall'Ora
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Valentina Grosso
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Martina Rossi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Francesco Alviano
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Laura Bonsi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Patrizia Nanni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Pier-Luigi Lollini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | | |
Collapse
|
5
|
Walker EB, Pritchard SM, Cunha CW, Aguilar HC, Nicola AV. Polyethylene glycol-mediated fusion of herpes simplex type 1 virions with the plasma membrane of cells that support endocytic entry. Virol J 2015; 12:190. [PMID: 26573723 PMCID: PMC4647588 DOI: 10.1186/s12985-015-0423-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/12/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Mouse B78 cells and Chinese hamster ovary (CHO) cells are important to the study of HSV-1 entry because both are resistant to infection at the level of viral entry. When provided with a gD-receptor such as nectin-1, these cells support HSV-1 entry by an endocytosis pathway. Treating some viruses bound to cells with the fusogen polyethylene glycol (PEG) mediates viral fusion with the cell surface but is insufficient to rescue viral entry. It is unclear whether PEG-mediated fusion of HSV with the plasma membrane of B78 or CHO cells results in successful entry and infection. FINDINGS Treating HSV-1 bound to B78 or CHO cells with PEG allowed viral entry as measured by virus-induced beta-galactosidase activity. Based on the mechanism of PEG action, we propose that entry likely proceeds by direct fusion of HSV particles with the plasma membrane. Under the conditions tested, PEG-mediated infection of CHO cells progressed to the level of HSV late gene expression, while B78 cells supported HSV DNA replication. We tested whether proteolysis or acidification of cell-bound virions could trigger HSV fusion with the plasma membrane. Under the conditions tested, mildly acidic pH of 5-6 or the protease trypsin were not capable of triggering HSV-1 fusion as compared to PEG-treated cell-bound virions. CONCLUSIONS B78 cells and CHO cells, which typically endocytose HSV prior to viral penetration, are capable of supporting HSV-1 entry via direct penetration. HSV capsids delivered directly to the cytosol at the periphery of these cells complete the entry process. B78 and CHO cells may be utilized to screen for factors that trigger entry as a consequence of fusion of virions with the cell surface, and PEG treatment can provide a necessary control.
Collapse
Affiliation(s)
- Erik B Walker
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
| | - Suzanne M Pritchard
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
| | - Cristina W Cunha
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
| | - Hector C Aguilar
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, 99164, USA.
| | - Anthony V Nicola
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, 99164, USA.
| |
Collapse
|
6
|
Variola virus-specific diagnostic assays: characterization, sensitivity, and specificity. J Clin Microbiol 2015; 53:1406-10. [PMID: 25673790 DOI: 10.1128/jcm.03613-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A public health response relies upon rapid and reliable confirmation of disease by diagnostic assays. Here, we detail the design and validation of two variola virus-specific real-time PCR assays, since previous assays cross-reacted with newly identified cowpox viruses. The assay specificity must continually be reassessed as other closely related viruses are identified.
Collapse
|
7
|
Dash P, Thomas PG. Host detection and the stealthy phenotype in influenza virus infection. Curr Top Microbiol Immunol 2015; 386:121-47. [PMID: 25038940 DOI: 10.1007/82_2014_412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The innate host response to influenza virus infection plays a critical role in determining the subsequent course of infection and the clinical outcome of disease. The host has a diverse array of detection and effector mechanisms that are able to recognize and initiate effective antiviral responses. In opposition, the virus utilizes a number of distinct mechanisms to evade host detection and effector activity in order to remain "stealthy" throughout its replication cycle. In this review, we describe these host and viral mechanisms, including the major pattern recognition receptor families (the TLRs, NLRs, and RLRs) in the host and the specific viral proteins such as NS1 that are key players in this interaction. Additionally, we explore nonreductive mechanisms of viral immune evasion and propose areas important for future inquiry.
Collapse
Affiliation(s)
- Pradyot Dash
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | |
Collapse
|
8
|
Abstract
UNLABELLED Nonmuscle myosin heavy chain IIA (NMHC-IIA) has been reported to function as a herpes simplex virus 1 (HSV-1) entry coreceptor by interacting with viral envelope glycoprotein B (gB). Vertebrates have three genetically distinct isoforms of the NMHC-II, designated NMHC-IIA, NMHC-IIB, and NMHC-IIC. COS cells, which are readily infected by HSV-1, do not express NMHC-IIA but do express NMHC-IIB. This observation prompted us to investigate whether NMHC-IIB might associate with HSV-1 gB and be involved in an HSV-1 entry like NMHC-IIA. In these studies, we show that (i) NMHC-IIB coprecipitated with gB in COS-1 cells upon HSV-1 entry; (ii) a specific inhibitor of myosin light chain kinase inhibited cell surface expression of NMHC-IIB in COS-1 cells upon HSV-1 entry as well as HSV-1 infection, as reported with NMHC-IIA; (iii) overexpression of mouse NMHC-IIB in IC21 cells significantly increased their susceptibility to HSV-1 infection; and (iv) knockdown of NMHC-IIB in COS-1 cells inhibited HSV-1 infection as well as cell-cell fusion mediated by HSV-1 envelope glycoproteins. These results supported the hypothesis that, like NMHC-IIA, NMHC-IIB associated with HSV-1 gB and mediated HSV-1 entry. IMPORTANCE Herpes simplex virus 1 (HSV-1) was reported to utilize nonmuscle myosin heavy chain IIA (NMHC-IIA) as an entry coreceptor associating with gB. Vertebrates have three genetically distinct isoforms of NMHC-II. In these isoforms, NMHC-IIB is of special interest since it highly expresses in neuronal tissue, one of the most important cellular targets of HSV-1 in vivo. In this study, we demonstrated that the ability to mediate HSV-1 entry appeared to be conserved in NMHC-II isoforms. These results may provide an insight into the mechanism by which HSV-1 infects a wide variety of cell types in vivo.
Collapse
|
9
|
Liu K, Jiang D, Zhang L, Yao Z, Chen Z, Yu S, Wang X. Identification of B- and T-cell epitopes from glycoprotein B of herpes simplex virus 2 and evaluation of their immunogenicity and protection efficacy. Vaccine 2011; 30:3034-41. [PMID: 22008818 DOI: 10.1016/j.vaccine.2011.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 09/16/2011] [Accepted: 10/05/2011] [Indexed: 10/16/2022]
Abstract
Herpes simplex virus (HSV) infection is a major health concern worldwide. Evidence obtained from animals and humans indicates that B- and T-cell responses contribute to protective immunity against herpes virus infection. Glycoprotein B is a transmembrane envelope component of HSV-1 and HSV-2, which plays an important role in virion morphogenesis and penetration into host cells, and can induce neutralizing antibodies and protective T-cell response when it is used to immunize humans and animals. However, little is known about gB epitopes that are involved in B- and T-cell activities in vitro and in vivo. Thus, the HSV-2 gB sequence was screened using B- and T-cell epitope prediction systems, and the B-cell regions and the HLA-A*0201-restricted epitopes were identified. These B-cell epitopes elicited high IgG antibody titers in Balb/C mice, with a predominantly IgG1 subclass distribution, which indicated a Th2 bias. Specific IgGs induced by these two epitopes were evaluated as the neutralizing antibodies for virus neutralization. The predicted T-cell epitopes stabilized the HLA-A*0201 molecules on T(2) cells, and stimulate interferon-γ-secreting and cytotoxic CD8(+) T cells. Immunization with the predicted peptides reduced virus shedding and protected against lethal viral challenge in mice. The functional epitopes described herein, both B- and T-cell epitopes, are potentially implicated in vaccine development.
Collapse
Affiliation(s)
- Kun Liu
- Department of Immunology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Arii J, Goto H, Suenaga T, Oyama M, Kozuka-Hata H, Imai T, Minowa A, Akashi H, Arase H, Kawaoka Y, Kawaguchi Y. Non-muscle myosin IIA is a functional entry receptor for herpes simplex virus-1. Nature 2010; 467:859-62. [PMID: 20944748 DOI: 10.1038/nature09420] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 08/16/2010] [Indexed: 01/24/2023]
Abstract
Herpes simplex virus-1 (HSV-1), the prototype of the α-herpesvirus family, causes life-long infections in humans. Although generally associated with various mucocutaneous diseases, HSV-1 is also involved in lethal encephalitis. HSV-1 entry into host cells requires cellular receptors for both envelope glycoproteins B (gB) and D (gD). However, the gB receptors responsible for its broad host range in vitro and infection of critical targets in vivo remain unknown. Here we show that non-muscle myosin heavy chain IIA (NMHC-IIA), a subunit of non-muscle myosin IIA (NM-IIA), functions as an HSV-1 entry receptor by interacting with gB. A cell line that is relatively resistant to HSV-1 infection became highly susceptible to infection by this virus when NMHC-IIA was overexpressed. Antibody to NMHC-IIA blocked HSV-1 infection in naturally permissive target cells. Furthermore, knockdown of NMHC-IIA in the permissive cells inhibited HSV-1 infection as well as cell-cell fusion when gB, gD, gH and gL were coexpressed. Cell-surface expression of NMHC-IIA was markedly and rapidly induced during the initiation of HSV-1 entry. A specific inhibitor of myosin light chain kinase, which regulates NM-IIA by phosphorylation, reduced the redistribution of NMHC-IIA as well as HSV-1 infection in cell culture and in a murine model for herpes stromal keratitis. NMHC-IIA is ubiquitously expressed in various human tissues and cell types and, therefore, is implicated as a functional gB receptor that mediates broad HSV-1 infectivity both in vitro and in vivo. The identification of NMHC-IIA as an HSV-1 entry receptor and the involvement of NM-IIA regulation in HSV-1 infection provide an insight into HSV-1 entry and identify new targets for antiviral drug development.
Collapse
Affiliation(s)
- Jun Arii
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Vachirayonstien T, Promkhatkaew D, Bunjob M, Chueyprom A, Chavalittumrong P, Sawanpanyalert P. Molecular evaluation of extracellular activity of medicinal herb Clinacanthus nutans against herpes simplex virus type-2. Nat Prod Res 2010; 24:236-45. [DOI: 10.1080/14786410802393548] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Thaveechai Vachirayonstien
- a Department of Medical Sciences , Medical Biotechnology Center, Ministry of Public Health , Muang, Nonthaburi, Thailand
| | - Duanthanorm Promkhatkaew
- a Department of Medical Sciences , Medical Biotechnology Center, Ministry of Public Health , Muang, Nonthaburi, Thailand
| | - Malee Bunjob
- b Department of Medical Sciences , Medicinal Plant Research Institute, Ministry of Public Health , Muang, Nonthaburi, Thailand
| | - Asawachai Chueyprom
- b Department of Medical Sciences , Medicinal Plant Research Institute, Ministry of Public Health , Muang, Nonthaburi, Thailand
| | - Pranee Chavalittumrong
- b Department of Medical Sciences , Medicinal Plant Research Institute, Ministry of Public Health , Muang, Nonthaburi, Thailand
| | - Pathom Sawanpanyalert
- a Department of Medical Sciences , Medical Biotechnology Center, Ministry of Public Health , Muang, Nonthaburi, Thailand
| |
Collapse
|
12
|
Phosphorylation of the U(L)31 protein of herpes simplex virus 1 by the U(S)3-encoded kinase regulates localization of the nuclear envelopment complex and egress of nucleocapsids. J Virol 2009; 83:5181-91. [PMID: 19279109 DOI: 10.1128/jvi.00090-09] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Herpes simplex virus 1 nucleocapsids bud through the inner nuclear membrane (INM) into the perinuclear space to obtain a primary viral envelope. This process requires a protein complex at the INM composed of the U(L)31 and U(L)34 gene products. While it is clear that the viral kinase encoded by the U(S)3 gene regulates the localization of pU(L)31/pU(L)34 within the INM, the molecular mechanism by which this is accomplished remains enigmatic. Here, we have determined the following. (i) The N terminus of pU(L)31 is indispensable for the protein's normal function and contains up to six serines that are phosphorylated by the U(S)3 kinase during infection. (ii) Phosphorylation at these six serines was not essential for a productive infection but was required for optimal viral growth kinetics. (iii) In the presence of active U(S)3 kinase, changing the serines to alanine caused the pU(L)31/pU(L)34 complex to aggregate at the nuclear rim and caused some virions to accumulate aberrantly in herniations of the nuclear membrane, much as in cells infected with a U(S)3 kinase-dead mutant. (iv) The replacement of the six serines of pU(L)31 with glutamic acid largely restored the smooth distribution of pU(L)34/pU(L)31 at the nuclear membrane and precluded the accumulation of virions in herniations whether or not U(S)3 kinase was active but also precluded the optimal primary envelopment of nucleocapsids. These observations indicate that the phosphorylation of pU(L)31 by pU(S)3 represents an important regulatory event in the virion egress pathway that can account for much of pU(S)3's role in nuclear egress. The data also suggest that the dynamics of pU(L)31 phosphorylation modulate both the primary envelopment and the subsequent fusion of the nascent virion envelope with the outer nuclear membrane.
Collapse
|
13
|
The U(L)31 and U(L)34 gene products of herpes simplex virus 1 are required for optimal localization of viral glycoproteins D and M to the inner nuclear membranes of infected cells. J Virol 2009; 83:4800-9. [PMID: 19279119 DOI: 10.1128/jvi.02431-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
U(L)31 and U(L)34 of herpes simplex virus type 1 form a complex necessary for nucleocapsid budding at the inner nuclear membrane (INM). Previous examination by immunogold electron microscopy and electron tomography showed that pU(L)31, pU(L)34, and glycoproteins D and M are recruited to perinuclear virions and densely staining regions of the INM where nucleocapsids bud into the perinuclear space. We now show by quantitative immunogold electron microscopy coupled with analysis of variance that gD-specific immunoreactivity is significantly reduced at both the INM and outer nuclear membrane (ONM) of cells infected with a U(L)34 null virus. While the amount of gM associated with the nuclear membrane (NM) was only slightly (P = 0.027) reduced in cells infected with the U(L)34 null virus, enrichment of gM in the INM at the expense of that in the ONM was greatly dependent on U(L)34 (P < 0.0001). pU(L)34 also interacted directly or indirectly with immature forms of gD (species expected to reside in the endoplasmic reticulum or nuclear membrane) in lysates of infected cells and with the cytosolic tail of gD fused to glutathione S-transferase in rabbit reticulocyte lysates, suggesting a role for the pU(L)34/gD interaction in recruiting gD to the NM. The effects of U(L)34 on gD and gM localization were not a consequence of decreased total expression of gD and gM, as determined by flow cytometry. Separately, pU(L)31 was dispensable for targeting gD and gM to the two leaflets of the NM but was required for (i) the proper INM-versus-ONM ratio of gD and gM in infected cells and (ii) the presence of electron-dense regions in the INM, representing nucleocapsid budding sites. We conclude that in addition to their roles in nucleocapsid envelopment and lamina alteration, U(L)31 and U(L)34 play separate but related roles in recruiting appropriate components to nucleocapsid budding sites at the INM.
Collapse
|
14
|
Human cytomegalovirus glycoprotein B is required for virus entry and cell-to-cell spread but not for virion attachment, assembly, or egress. J Virol 2009; 83:3891-903. [PMID: 19193805 DOI: 10.1128/jvi.01251-08] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glycoprotein B (gB) homologs are conserved throughout the family Herpesviridae and appear to serve essential, universal functions, as well as specific functions unique to a particular herpesvirus. Genetic analysis is a powerful tool to analyze protein function, and while it has been possible to generate virus mutants, complementation of essential virus knockouts has been problematic. Human cytomegalovirus (HCMV) gB (UL55) plays an essential role in the replication cycle of the virus. To define the function(s) of gB in HCMV infection, the BAC system was used to generate a recombinant virus in which the UL55 gene was replaced with galK (pAD/CreDeltaUL55). UL55 deletions in the viral genome have been made before, demonstrating that UL55 is an essential gene. However, without being able to successfully complement the genetic defect, a phenotypic analysis of the mutant virus was impossible. We generated fibroblasts expressing HCMV gB that complement pAD/CreDeltaUL55 and produce infectious virions lacking the UL55 gene but containing wild-type gB on the virion surface (DeltaUL55-gB HCMV). This is the first successful complementation of an HCMV mutant with a glycoprotein deleted. To characterize DeltaUL55 infection in the absence of gB, noncomplementing cells were infected with DeltaUL55-gB virus. All stages of gene expression were detected, and significant amounts of DNase-resistant viral DNA genomes, representing whole intact virions, were released into the infected cell supernatant. Gradient purification of these virions revealed they lacked gB but contained other viral structural proteins. The gB-null virions were able to attach to the cell surface similarly to wild-type gB-containing virions but were defective in virus entry and cell-to-cell spread. Glycoprotein B-null virions do, however, contain infectious DNA, as IE gene expression can be detected in fibroblasts following treatment of attached gB-null virions with a membrane fusion agent, polyethylene glycol. Taken together, our results indicate that gB is required for virus entry and cell-to-cell spread of the virus. However, HCMV gB is not absolutely required for virus attachment or assembly and egress from infected cells.
Collapse
|
15
|
Dendritic cells mediate herpes simplex virus infection and transmission through the C-type lectin DC-SIGN. J Gen Virol 2008; 89:2398-2409. [DOI: 10.1099/vir.0.2008/003129-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dendritic cells (DCs) are essential for the induction of specific immune responses against invading pathogens. Herpes simplex virus (HSV) is a common human pathogen that causes painful but mild infections of the skin and mucosa, and which results in latency and recurrent infections. Of the two HSV subtypes described, HSV-1 causes mainly oral–facial lesions, whilst HSV-2 is associated with genital herpes. DCs are involved in HSV-induced immune suppression, but little is known about the molecular interactions between DCs and HSV. This study demonstrated that HSV-1 and -2 both interact with the DC-specific C-type lectin DC-SIGN. Further analyses demonstrated that DC-SIGN interacts with the HSV glycoproteins gB and gC. Binding of HSV-1 to immature DCs depended on both DC-SIGN and heparan sulfate proteoglycans. Strikingly, HSV-1 infection of DCs was almost completely inhibited by blocking antibodies against DC-SIGN. Thus, DC-SIGN is an important attachment receptor for HSV-1 on immature DCs and enhances infection of DCs in cis. In addition, DC-SIGN captures HSV-1 for transmission to permissive target cells. These data strongly suggest that DC-SIGN is a potential target to prevent HSV infection and virus dissemination. Further studies will show whether these interactions are involved in HSV-induced immune suppression.
Collapse
|
16
|
Proteolytic cleavage of VP1-2 is required for release of herpes simplex virus 1 DNA into the nucleus. J Virol 2008; 82:3311-9. [PMID: 18216103 DOI: 10.1128/jvi.01919-07] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this report we propose a model in which after the herpes simplex virus (HSV) capsid docks at the nuclear pore, the tegument protein attached to the capsid must be cleaved by a serine or a cysteine protease in order for the DNA to be released into the nucleus. In support of the model are the following results. (i) Exposure of cells at the time of or before infection to l-(tosylamido-2-phenyl) ethyl chloromethyl ketone (TPCK), a serine-cysteine protease inhibitor, prevents the release of viral DNA or expression of viral genes. TPCK does not block viral gene expression after entry of viral DNA into the nucleus. (ii) The tegument protein VP1-2, the product of the U(L)36 gene, is cleaved shortly after the entry of the HSV 1 (HSV-1) virion into the cell. (iii) The proteolytic cleavage of VP1-2 does not occur in cells that are infected with HSV-1 under conditions that prevent the release of the viral DNA into the nucleus. (iv) The proteolytic cleavage of VP1-2 occurs only after the capsid is attached to the nuclear pore. Thus, TPCK prevented the release of HSV-1 DNA into the nucleus when added to medium 1 hour after infection with tsB7 at 39.5 degrees C followed by a shift down to the permissive temperature. The ts lesion maps in the U(L)36 gene. At the nonpermissive temperature, the capsids accumulate at the nuclear pore but the DNA is not released into the nucleus.
Collapse
|
17
|
Ch'ng TH, Spear PG, Struyf F, Enquist LW. Glycoprotein D-independent spread of pseudorabies virus infection in cultured peripheral nervous system neurons in a compartmented system. J Virol 2007; 81:10742-57. [PMID: 17652377 PMCID: PMC2045490 DOI: 10.1128/jvi.00981-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The molecular mechanisms underlying the directional neuron-to-epithelial cell transport of herpesvirus particles during infection are poorly understood. To study the role of the viral glycoprotein D (gD) in the directional spread of herpes simplex virus (HSV) and pseudorabies virus (PRV) infection, a culture system consisting of sympathetic neurons or epithelial cells in different compartments was employed. We discovered that PRV infection could spread efficiently from neurons to cells and back to neurons in the absence of gD, the viral ligand required for entry of extracellular particles. Unexpectedly, PRV infection can also spread transneuronally via axo-axonal contacts. We show that this form of interaxonal spread between neurons is gD independent and is not mediated by extracellular virions. We also found that unlike PRV gD, HSV-1 gD is required for neuron-to-cell spread of infection. Neither of the host cell gD receptors (HVEM and nectin-1) is required in target primary fibroblasts for neuron-to-cell spread of HSV-1 or PRV infection.
Collapse
Affiliation(s)
- T H Ch'ng
- Schultz Laboratory, Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | | | | | |
Collapse
|
18
|
Pheasey N, John J, Love C, Coffin R, Ward JM, Boushaba R, Hoare M, Levy MS. A capillary cytometer method to quantitate viable virus particles based on early detection of viral antigens and cellular events within single cells. J Virol Methods 2006; 137:213-8. [PMID: 16854472 DOI: 10.1016/j.jviromet.2006.06.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Revised: 06/06/2006] [Accepted: 06/13/2006] [Indexed: 11/26/2022]
Abstract
The traditional plaque forming and TCID(50) methods to determine replication competent virus titres rely on several cycles of replication and infection to generate a plaque with an incubation period of 24-72 h post-infection typically required. We developed a method to quantify infective viral particles based on early detection of cellular events by capillary cytometry. The method uses a capillary cytometer as a precise cell counter that can discriminate infected from non-infected cells. The general protocol was developed using a Guava PCA, genetically modified HSV-1 virus and polyclonal antibodies against antigens expressed on the cell membrane. Infection was detected after 1 h incubation and a plateau in the number of infected cells was observed between 7 and 9 h. A good correlation between titres obtained by the plaque forming method and the proposed method was observed for a ratio of infected to total cells between 0.5 and 0.05. The rapid and automated analysis (10 s/1000 events acquired per sample) makes the method particularly useful for high-throughput applications. The proposed method can be extended easily to determine the titre of other viruses providing a powerful tool for virology and antiviral screening.
Collapse
Affiliation(s)
- Nigel Pheasey
- Department of Biochemical Engineering, University College London, Torrington Place, WC1E 7JE, UK
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Li Y, Olson VA, Laue T, Laker MT, Damon IK. Detection of monkeypox virus with real-time PCR assays. J Clin Virol 2006; 36:194-203. [PMID: 16731033 PMCID: PMC9628957 DOI: 10.1016/j.jcv.2006.03.012] [Citation(s) in RCA: 275] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Revised: 03/13/2006] [Accepted: 03/17/2006] [Indexed: 11/16/2022]
Abstract
BACKGROUND Human monkeypox, a zoonotic disease, was first reported outside of Africa during the 2003 US outbreak. OBJECTIVES We present two real-time PCR assays critical for laboratory diagnosis of monkeypox during the 2003 US outbreak. STUDY DESIGN A TaqMan-based assay (E9L-NVAR) targets the orthopoxvirus DNA polymerase gene and detects Eurasian orthopoxviruses other than Variola. A hybridization assay, utilizing a MGB Eclipsetrade mark (Epoch Biosciences) probe, targets an envelope protein gene (B6R) and specifically detects monkeypox virus (MPXV). Assays were validated using coded orthopoxvirus DNA samples and used to evaluate lesion samples from five confirmed US monkeypox cases. RESULTS E9L-NVAR did not detect variola (48 strains), North American orthopoxviruses (2), or DNA derived from non-poxviral rash illnesses. The assay reproducibly identified various concentrations of 13 Eurasian orthopoxvirus strains and was sensitive to 12.5 vaccinia genomes. The B6R assay recognized 15 different MPXV strains, while other orthopoxvirus (9) and bacteria (15) strains did not cross-react. Of the 13 human samples tested from confirmed cases, both assays identified 100% as containing MPXV DNA. CONCLUSIONS E9L-NVAR and B6R assays demonstrate 100% specificity for non-variola Eurasian orthopoxvirus and MPXV, respectively. Using two discrete viral gene targets, these assays together provide a reliable and sensitive method for quickly confirming monkeypox infections.
Collapse
Affiliation(s)
- Yu Li
- Poxvirus Program, Division of Viral and Rickettsial Diseases, National Centers of Infectious Diseases, Centers for Disease Control and Prevention, Mail Stop G-43, 1600 Clifton Road, NE, Atlanta, GA 30333, USA
| | | | | | | | | |
Collapse
|
20
|
Li W, Minova-Foster TJ, Norton DD, Muggeridge MI. Identification of functional domains in herpes simplex virus 2 glycoprotein B. J Virol 2006; 80:3792-800. [PMID: 16571796 PMCID: PMC1440482 DOI: 10.1128/jvi.80.8.3792-3800.2006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glycoprotein B (gB) is one of four membrane proteins that are essential for the entry of herpes simplex viruses (HSV) into cells, and coexpression of the same combination of proteins in transfected cells results in cell fusion. The latter effect is reminiscent of the ability of virus infection to cause cell fusion, particularly since the degree of fusion is greatly increased by syncytial mutations in gB. Despite intensive efforts with the gB homologs of HSV and some other herpesviruses, information about functionally important regions in the 700-amino-acid ectodomain of this protein is very limited at present. This is largely due to the misfolding of the majority of the mutants examined. It was shown previously that the percentage of correctly folded mutants could be increased by targeting only predicted loop regions (i.e., not alpha-helix or beta-strand), and by using this approach new functional domains in HSV-2 gB have now been identified.
Collapse
Affiliation(s)
- Wei Li
- Center for Molecular and Tumor Virology, Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisian 71130, USA
| | | | | | | |
Collapse
|
21
|
Ryckman BJ, Jarvis MA, Drummond DD, Nelson JA, Johnson DC. Human cytomegalovirus entry into epithelial and endothelial cells depends on genes UL128 to UL150 and occurs by endocytosis and low-pH fusion. J Virol 2006; 80:710-22. [PMID: 16378974 PMCID: PMC1346879 DOI: 10.1128/jvi.80.2.710-722.2006] [Citation(s) in RCA: 266] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Human cytomegalovirus (HCMV) replication in epithelial and endothelial cells appears to be important in virus spread, disease, and persistence. It has been difficult to study infection of these cell types because HCMV laboratory strains (e.g., AD169 and Towne) have lost their ability to infect cultured epithelial and endothelial cells during extensive propagation in fibroblasts. Clinical strains of HCMV (e.g., TR and FIX) possess a cluster of genes (UL128 to UL150) that are largely mutated in laboratory strains, and recent studies have indicated that these genes facilitate replication in epithelial and endothelial cells. The mechanisms by which these genes promote infection of these two cell types are unclear. We derived an HCMV UL128-to-UL150 deletion mutant from strain TR, TRdelta4, and studied early events in HCMV infection of epithelial and endothelial cells, and the role of genes UL128 to UL150. Analysis of wild-type TR indicated that HCMV enters epithelial and endothelial cells by endocytosis followed by low-pH-dependent fusion, which is different from the pH-independent fusion with the plasma membrane observed with human fibroblasts. TRdelta4 displayed a number of defects in early infection processes. Adsorption and entry of TRdelta4 on epithelial cells were poor compared with those of TR, but these defects could be overcome with higher doses of virus and the use of polyethylene glycol (PEG) to promote fusion between virion and cellular membranes. High multiplicity and PEG treatment did not promote infection of endothelial cells by TRdelta4, yet virus particles were internalized. Together, these data indicate that genes UL128 to UL150 are required for HCMV adsorption and penetration of epithelial cells and to promote some early stage of virus replication, subsequent to virus entry, in endothelial cells.
Collapse
Affiliation(s)
- Brent J Ryckman
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 Sam Jackson Park Rd., Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
22
|
McShane MP, Longnecker R. Cell-surface expression of a mutated Epstein-Barr virus glycoprotein B allows fusion independent of other viral proteins. Proc Natl Acad Sci U S A 2004; 101:17474-9. [PMID: 15583133 PMCID: PMC536015 DOI: 10.1073/pnas.0404535101] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epstein-Barr virus (EBV) infects human B lymphocytes and epithelial cells. We have compared the requirements for EBV glycoprotein-induced cell fusion between Chinese hamster ovary effecter cells and human B lymphoblasts or epithelial cells by using a virus-free cell fusion assay. EBV-encoded gB, gH, gL, and gp42 glycoproteins were required for efficient B cell fusion, whereas EBV gB, gH, and gL glycoproteins were required for Chinese hamster ovary effecter cell fusion with epithelial cell lines (AGS and SCC68) or the human embryonic kidney cell line 293-P. Fusion with human embryonic kidney 293-P cells was greater than fusion observed with B cells, indicative of an important role for cell contact. An antibody directed against the gH and gL complex inhibited epithelial cell fusion. Increased surface expression of gB alone as a result of truncations or point mutants in the carboxyl-terminal tail allowed gB-mediated fusion with epithelial cells, albeit at a lower level than with coexpression of gB, gH, and gL. Overall, gB appears to be the critical component for EBV glycoprotein-mediated cell fusion.
Collapse
Affiliation(s)
- Marisa P McShane
- Department of Microbiology and Immunology, The Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | | |
Collapse
|
23
|
Jones TR, Lee SW, Johann SV, Razinkov V, Visalli RJ, Feld B, Bloom JD, O'Connell J. Specific inhibition of human cytomegalovirus glycoprotein B-mediated fusion by a novel thiourea small molecule. J Virol 2004; 78:1289-300. [PMID: 14722284 PMCID: PMC321382 DOI: 10.1128/jvi.78.3.1289-1300.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A novel small molecule inhibitor of human cytomegalovirus (HCMV) was identified as the result of screening a chemical library by using a whole-virus infected-cell assay. Synthetic chemistry efforts yielded the analog designated CFI02, a compound whose potency had been increased about 100-fold over an initial inhibitor. The inhibitory concentration of CFI02 in various assays is in the low nanomolar range. CFI02 is a selective and potent inhibitor of HCMV; it has no activity against other CMVs, alphaherpesviruses, or unrelated viruses. Mechanism-of-action studies indicate that CFI02 acts very early in the replication cycle, inhibiting virion envelope fusion with the cell plasma membrane. Mutants resistant to CFI02 have mutations in the abundant virion envelope glycoprotein B that are sufficient to confer resistance. Taken together, the data suggest that CFI02 inhibits glycoprotein B-mediated HCMV virion fusion. Furthermore, CFI02 inhibits the cell-cell spread of HCMV. This is the first study of a potent and selective small molecule inhibitor of CMV fusion and cell-cell spread.
Collapse
Affiliation(s)
- Thomas R Jones
- Infectious Disease Section, Wyeth Research, Pearl River, New York 10965, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Domingo C, Gadea I, Pardeiro M, Castilla C, Fernández S, Fernández-Clua MA, De la Cruz Troca JJ, Punzón C, Soriano F, Fresno M, Tabarés E. Immunological properties of a DNA plasmid encoding a chimeric protein of herpes simplex virus type 2 glycoprotein B and glycoprotein D. Vaccine 2003; 21:3565-74. [PMID: 12922084 DOI: 10.1016/s0264-410x(03)00423-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A DNA plasmid containing a chimeric sequence encoding both herpes simplex virus type 2 (HSV-2) glycoprotein B (gB) and glycoprotein D (gD) external domains (pcgDB) was used to immunize BALB/c mice against genital HSV-2 infection. To determine the efficacy of this vaccine, groups of mice immunized with the pcgDB plasmid were compared with animals immunized with plasmids corresponding to the individual proteins (pcgBt or pcgDt), administered separately or in combination (pcgBt + pcgDt). We studied the response of the different mouse groups to viral challenge by analyzing clinical disease (vaginitis), serum antibody levels, as well as lymphoproliferative responses and cytokine production by spleen cells. Increased IFN-gamma levels correlated with prolonged survival in mice immunized with the plasmid pcgDB, relative to mice immunized with plasmids coding for the individual proteins alone or in combination. Our results show that immunization with the plasmid encoding the chimeric protein is advantageous over separate proteins. These findings may have important implications for the development of multivalent DNA vaccines against HSV and other complex pathogens.
Collapse
Affiliation(s)
- C Domingo
- Departamento de Medicina Preventiva y Salud Pública (Microbiología), Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Herpes simplex virus (HSV) is a neurotropic DNA virus with many favorable properties as a gene delivery vector. HSV is highly infectious, so HSV vectors are efficient vehicles for the delivery of exogenous genetic material to cells. Viral replication is readily disrupted by null mutations in immediate early genes that in vitro can be complemented in trans, enabling straightforward production of high-titre pure preparations of non-pathogenic vector. The genome is large (152 Kb) and many of the viral genes are dispensable for replication in vitro, allowing their replacement with large or multiple transgenes. Latent infection with wild-type virus results in episomal viral persistence in sensory neuronal nuclei for the duration of the host lifetime. Transduction with replication-defective vectors causes a latent-like infection in both neural and non-neural tissue; the vectors are non-pathogenic, unable to reactivate and persist long-term. The latency active promoter complex can be exploited in vector design to achieve long-term stable transgene expression in the nervous system. HSV vectors transduce a broad range of tissues because of the wide expression pattern of the cellular receptors recognized by the virus. Increasing understanding of the processes involved in cellular entry has allowed preliminary steps to be taken towards targeting the tropism of HSV vectors. Using replication-defective HSV vectors, highly encouraging results have emerged from recent pre-clinical studies on models of neurological disease, including glioma, peripheral neuropathy, chronic pain and neurodegeneration. Consequently, HSV vectors encoding appropriate transgenes to tackle these pathogenic processes are poised to enter clinical trials.
Collapse
Affiliation(s)
- Edward A Burton
- Department of Clinical Neurology, University of Oxford, United Kingdom
| | | | | |
Collapse
|
26
|
Dan K, Miyashita K, Seto Y, Fujita H, Yamase T. The memory effect of heteropolyoxotungstate (PM-19) pretreatment on infection by herpes simplex virus at the penetration stage. Pharmacol Res 2002; 46:357-61. [PMID: 12361699 DOI: 10.1016/s1043661802001706] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The keggin-type heteropolyoxotungstate K(7)[PTi(2)W(10)O(40)].6H(2)O (PM-19) is a potent polyoxometalate (PM) inhibitor of the replication of herpes simplex virus (HSV). Pretreatment of Vero cells with PM-19 prior to HSV-2 infection enhanced the antiviral potency of PM-19 almost 10-fold compared with treatment of the cells only after infection. The pretreatment effect of PM-19 is called "the memory effect". The memory effect was reflected by inhibition of plaque formation and decrease of intracellular virus DNA quantity, and was strongest when PM-19 was present during the penetration stage of HSV-2 infection. The effect was maintained under conditions of fusion induced by polyethyleneglycol treatment. This suggests that PM-19 does not act at the fusion stage of infection. Using the infectious center assay method, it was clarified that a second round of infection was inhibited by about 30% in the presence of PM-19 at the penetration stage compared with the virus control in nontreated cells. The inhibition was enhanced to about 60% by PM-19 pretreatment prior to infection. This suggests that PM-19 pretreatment of the cells protects them against HSV-2 infection.
Collapse
Affiliation(s)
- Katsuaki Dan
- Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160, Japan.
| | | | | | | | | |
Collapse
|
27
|
Hung SL, Cheng YY, Wang YH, Chang KW, Chen YT. Expression and roles of herpesvirus entry mediators A and C in cells of oral origin. ORAL MICROBIOLOGY AND IMMUNOLOGY 2002; 17:215-23. [PMID: 12121471 DOI: 10.1034/j.1399-302x.2002.170403.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The roles of viral glycoprotein D (gD) and cellular herpesvirus entry mediators A (HveA) and C (HveC) in herpes simplex virus entry into oral cells were determined. Studies with purified truncated forms of gD-1, HveA and HveC indicated that these molecules may be involved in herpes simplex virus entry into oral cells. Moreover, HveA was expressed similarly in primary cultures of gingival keratinocytes and fibroblasts, whereas HveC was expressed at higher levels in gingival keratinocytes, as determined by RT-PCR and immunocytochemical staining. Further analysis using immunohistochemistry demonstrated that both HveA and HveC were expressed in epithelial cells, fibroblasts and vascular endothelial cells in gingival tissues. However, only HveC was detected in nerve fibers. Also, HveA was detected throughout the epidermis, whereas HveC was pronounced in the strata basale and spinosum. In conclusion, this study characterized HveA and HveC, molecules that may participate in entry of herpes simplex virus into oral cells.
Collapse
MESH Headings
- Adult
- Cell Adhesion Molecules/biosynthesis
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/physiology
- Fibroblasts/enzymology
- Fibroblasts/virology
- Gingiva/cytology
- Gingiva/enzymology
- Gingiva/virology
- Herpesvirus 1, Human/chemistry
- Herpesvirus 1, Human/pathogenicity
- Humans
- Immunohistochemistry
- KB Cells/virology
- Keratinocytes/enzymology
- Keratinocytes/virology
- Mouth Mucosa/cytology
- Mouth Mucosa/enzymology
- Mouth Mucosa/virology
- Nectins
- RNA, Messenger/analysis
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Member 14
- Receptors, Virus/biosynthesis
- Receptors, Virus/genetics
- Receptors, Virus/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Viral Envelope Proteins/biosynthesis
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/physiology
Collapse
Affiliation(s)
- S-L Hung
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
28
|
Hung SL, Wang YH, Chen HW, Lee PL, Chen YT. Analysis of herpes simplex virus entering into cells of oral origin. Virus Res 2002; 86:59-69. [PMID: 12076830 DOI: 10.1016/s0168-1702(02)00055-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The entry of herpes simplex virus (HSV) into an oral epithelial cell line, primary normal human oral keratinocytes (NHOK) and gingival fibroblasts (GF) was examined. Infection of these cells by HSV-1 and HSV-2 was blocked by heparin. Further examination indicated that heparin reduced viral attachment but not penetration. Moreover, neomycin inhibited HSV-1 infection more effectively than HSV-2 infection in GF, but not in NHOK. In conclusion, our results elucidated some aspects of the HSV entry process into oral cells and revealed some differences in HSV entering into NHOK and GF.
Collapse
Affiliation(s)
- Shan Ling Hung
- Institute of Oral Biology, National Yang-Ming University, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, Taiwan.
| | | | | | | | | |
Collapse
|
29
|
Abstract
Herpes simplex virus (HSV) is an encapsulated DNA virus, with many favourable properties for use as a gene transfer vector. For gene therapy applications, it may be desirable to restrict transgene expression to pre-defined subsets of cells. One potential method for achieving targeted transgene expression using the HSV vector system might involve dictating the cell types to which the vector will transfer the therapeutic transgene of interest. HSV delivers its genetic payload to cells directly through the plasmalemma; the mechanisms are complex and involve multiple viral and cell surface determinants. We have investigated several ways in which each component of the cell entry cascade may be manipulated in order to restrict viral DNA and transgene delivery to particular cellular populations. Our results indicate that targeted transduction may be a viable approach to achieving our goal of targeted HSV-mediated transgene expression.
Collapse
Affiliation(s)
- E A Burton
- University of Pittsburgh School of Medicine, Department of Molecular Genetics and Biochemistry, E1240 Biomedical Sciences Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | | | | | | |
Collapse
|
30
|
Kingham BF, Zelnık V, Kopáček J, Majerčiak V, Ney E, Schmidt CJ. The genome of herpesvirus of turkeys: comparative analysis with Marek's disease viruses. J Gen Virol 2001; 82:1123-1135. [PMID: 11297687 DOI: 10.1099/0022-1317-82-5-1123] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The complete coding sequence of the herpesvirus of turkeys (HVT) unique long (U(L)) region along with the internal repeat regions has been determined. This allows completion of the HVT nucleotide sequence by linkage to the sequence of the unique short (U(S)) region. The genome is approximately 160 kbp and shows extensive similarity in organization to the genomes of Marek's disease virus serotypes 1 and 2 (MDV-1, MDV-2) and other alphaherpesviruses. The HVT genome contains 75 ORFs, with three ORFs present in two copies. Sixty-seven ORFs were identified readily as homologues of other alphaherpesvirus genes. Seven of the remaining eight ORFs are homologous to genes in MDV, but are absent from other herpesviruses. These include a gene with similarity to cellular lipases. The final, HVT-unique gene is a virus homologue of the cellular NR-13 gene, the product of which belongs to the Bcl family of proteins that regulate apoptosis. No other herpesvirus sequenced to date contains a homologue of this gene. Of potential significance is the absence of a complete block of genes within the HVT internal repeat that is present in MDV-1. These include the pp38 and meq genes, which have been implicated in MDV-1-induced T-cell lymphoma. By implication, other genes present in this region of MDV-1, but missing in HVT, may play important roles in the different biological properties of the viruses.
Collapse
Affiliation(s)
- Brewster F Kingham
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19717, USA1
| | - Vladimır Zelnık
- Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 842 45 Bratislava, Slovak Republic2
| | - Juraj Kopáček
- Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 842 45 Bratislava, Slovak Republic2
| | - Vladimır Majerčiak
- Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 842 45 Bratislava, Slovak Republic2
| | - Erik Ney
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19717, USA1
| | - Carl J Schmidt
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19717, USA1
| |
Collapse
|
31
|
Foster TP, Chouljenko VN, Kousoulas KG. Functional characterization of the HveA homolog specified by African green monkey kidney cells with a herpes simplex virus expressing the green fluorescence protein. Virology 1999; 258:365-74. [PMID: 10366573 DOI: 10.1006/viro.1999.9743] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We cloned the gene specified by African monkey kidney cells (Vero) that codes for the homolog of the herpes virus entry mediator (HveA) specified by HeLa cells. The primary sequence of the monkey HveA (HveAs) differed significantly from HveA. Single amino acid differences were distributed throughout the amino and carboxyl terminal portions of the HveAs in comparison with the HveA, whereas certain regions were highly conserved. The predicted membrane spanning domains of the two receptors differed substantially due to insertions and deletions of short amino acid sequences. The ability of HveAs to mediate HSV virus entry was tested in a series of experiments using the recombinant virus KOS/EGFP, which constitutively expressed the enhanced green fluorescence protein (EGFP) and Chinese hamster ovary cells (CHO) transformed with the HveAs gene. The KOS/EGFP virus was constructed by inserting an EGFP gene cassette within the intergenic region between the UL53 (gK) and UL54 (ICP27) genes. The KOS/EGFP virus formed viral plaques and replicated as well as the wild-type KOS virus. HveAs-transformed CHO cells constitutively expressing HveAs mediated herpesvirus entry efficiently, whereas cells transformed with the HveAs gene in the noncoding orientation did not mediate virus entry. A genetically engineered protein composed of the amino-terminal portion of the HveAs protein fused to the heavy chain of mouse IgG immunoglobulin as well as mouse antibodies raised against HveAs blocked virus entry into HveAs-transformed CHO cells. Thus, HveAs is the functional homolog of HveA.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- CHO Cells
- Cell Line, Transformed
- Chlorocebus aethiops
- Cloning, Molecular
- Cricetinae
- Gene Expression
- Green Fluorescent Proteins
- HeLa Cells
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/metabolism
- Herpesvirus 1, Human/physiology
- Humans
- Kidney/cytology
- Luminescent Proteins/genetics
- Mice
- Molecular Sequence Data
- Receptors, Tumor Necrosis Factor
- Receptors, Tumor Necrosis Factor, Member 14
- Receptors, Virus/metabolism
- Recombination, Genetic
- Vero Cells
- Viral Plaque Assay
Collapse
Affiliation(s)
- T P Foster
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, 70803, USA
| | | | | |
Collapse
|
32
|
Matis J, Kúdelová M, Rajcáni J. Interference of the low-pH inactivated herpes simplex virus type 1 (HSV-1) strain HSZP with the early shutoff function of superinfecting HSV-1 strain KOS. Virus Res 1999; 60:81-6. [PMID: 10225276 DOI: 10.1016/s0168-1702(99)00004-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In former studies, we described that the HSZP strain of herpes simplex virus type 1 (HSV-1) was defective with respect to the early shutoff of host protein synthesis but was effective at interfering with the early shutoff function of the HSV-1 strain KOS, even when heat-inactivated or neutralized by antibody. However, the HSZP strain failed to interfere when inactivated with zinc ions or purified from cells treated with 2-deoxy-D-glucose. In this study, we provide evidence that the ability of the purified low-pH inactivated (citrate buffer, pH 3.0) and gel-filtered (Sephadex G-25) HSZP virions to adsorb host cells was not significantly affected. However, their ability to induce interference with the early shutoff function of the superinfecting HSV-1 strain KOS was restricted. In comparison with native virus, up to eight times more low-pH inactivated HSZP virions were needed to interfere with the shutoff by strain KOS. The interference was not due to exclusion of strain KOS by HSZP at the level of adsorption and/or penetration. The restriction was partially overcome by treatment of the cells with polyethylene glycol after adsorption of the low-pH inactivated HSZP virions. This observation indicates that the direct fusion of the virion envelope of low-pH inactivated HSZP with the plasma cell membrane was predominantly hampered.
Collapse
Affiliation(s)
- J Matis
- Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | | | | |
Collapse
|
33
|
Karger A, Schmidt J, Mettenleiter TC. Infectivity of a pseudorabies virus mutant lacking attachment glycoproteins C and D. J Virol 1998; 72:7341-8. [PMID: 9696830 PMCID: PMC109958 DOI: 10.1128/jvi.72.9.7341-7348.1998] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Initiation of herpesvirus infection requires attachment of virions to the host cell followed by fusion of virion envelope and cellular cytoplasmic membrane during penetration. In several alphaherpesviruses, glycoprotein C (gC) is the primary attachment protein, interacting with cell-surface heparan sulfate proteoglycans. Secondary binding is mediated by gD, which, normally, is also required for penetration. Recently, we described the isolation of a gD-negative infectious pseudorabies virus (PrV) mutant, PrV gD- Pass (J. Schmidt, B. G. Klupp, A. Karger, and T. C. Mettenleiter, J. Virol. 71:17-24, 1997). In PrV gD- Pass, attachment and penetration occur in the absence of gD. To assess the importance of specific attachment for infectivity of PrV gD- Pass, the gene encoding gC was deleted, resulting in mutant PrV gCD- Pass. Deletion of both known attachment proteins reduced specific infectivity compared to wild-type PrV by more than 10,000-fold. Surprisingly, the virus mutant still retained significant infectivity and could be propagated on normal noncomplementing cells, indicating the presence of another receptor-binding virion protein. Selection of bovine kidney (MDBK) cells resistant to infection by PrV gCD- Pass resulted in the isolation of a cell clone, designated NB, which was susceptible to infection by wild-type PrV but refractory to infection by either PrV gCD- Pass or PrV gD- Pass, a defect which could partially be overcome by polyethylene glycol (PEG)-induced membrane fusion. However, even after PEG-induced infection plaque formation of PrV gCD- Pass or PrV gD- Pass did not ensue in NB cells. Also, phenotypic gD complementation of PrV gCD- Pass or PrV gD- Pass rescued the defect in infection of NB cells but did not restore plaque formation. Glycosaminoglycan analyses of MDBK and NB cells yielded identical results, and NB cells were normally susceptible to infection by other alphaherpesviruses as well as vesicular stomatitis virus. Infectious center assays after PEG-induced infection of NB cells with PrV gD- Pass on MDBK cells indicated efficient exit of virions from infected NB cells. Together, our data suggest the presence of another receptor and receptor-binding virion protein which can mediate PrV entry and cell-to-cell spread in MDBK cells.
Collapse
Affiliation(s)
- A Karger
- Institute of Molecular and Cellular Virology, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, D-17498 Insel Riems, Germany
| | | | | |
Collapse
|
34
|
Norton DD, Dwyer DS, Muggeridge MI. Use of a neural network secondary structure prediction to define targets for mutagenesis of herpes simplex virus glycoprotein B. Virus Res 1998; 55:37-48. [PMID: 9712510 DOI: 10.1016/s0168-1702(98)00030-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Herpes simplex virus glycoprotein B (HSV gB) is essential for penetration of virus into cells, for cell-to-cell spread of virus, and for cell-cell fusion. Every member of the family Herpesviridae has a gB homolog, underlining its importance. The antigenic structure of gB has been studied extensively, but little is known about which regions of the protein are important for its roles in virus entry and spread. In contrast to successes with other HSV glycoproteins, attempts to map functional domains of gB by insertion mutagenesis have been largely frustrated by the misfolding of most mutants. The present study shows that this problem can be overcome by targeting mutations to the loop regions that connect alpha-helices and beta-strands, avoiding the helices and strands themselves. The positions of loops in the primary sequence were predicted by the PHD neural network procedure, using a multiple sequence alignment of 19 alphaherpesvirus gB sequences as input. Comparison of the prediction with a panel of insertion mutants showed that all mutants with insertions in predicted alpha-helices or beta-strands failed to fold correctly and consequently had no activity in virus entry; in contrast, half the mutants with insertions in predicted loops were able to fold correctly. There are 27 predicted loops of four or more residues in gB; targeting of mutations to these regions will minimize the number of misfolded mutants and maximize the likelihood of identifying functional domains of the protein.
Collapse
Affiliation(s)
- D D Norton
- Department of Microbiology and Immunology, Louisiana State University School of Medicine, Shreveport 71130, USA
| | | | | |
Collapse
|
35
|
Roller RJ, Rauch D. Herpesvirus entry mediator HVEM mediates cell-cell spread in BHK(TK-) cell clones. J Virol 1998; 72:1411-7. [PMID: 9445042 PMCID: PMC124620 DOI: 10.1128/jvi.72.2.1411-1417.1998] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/1997] [Accepted: 11/03/1997] [Indexed: 02/05/2023] Open
Abstract
95-19 and U(S)11c119.3 are BHK(TK-)-derived cell lines that are highly resistant to postattachment entry of herpes simplex virus type 1 (HSV-1) and HSV-2 but not to later steps in single-step replication. The resistance properties of these two cell types are not identical. U(S)11c119.3 cells are fully susceptible to pseudorabies virus (PRV), as shown by single-step growth experiments, whereas 95-19 cells are resistant to entry of free PRV but not to entry by cell-cell spread. We have tested the ability of HVEM to overcome the block to infection in both cell lines following transient and stable transfection. HVEM was able to mediate entry of free HSV-1 into both cell lines, as shown by an increase in the number of beta-galactosidase-expressing cells in cultures transiently transfected with an HVEM expression plasmid and infected with lacZ-expressing HSV-1. In stably transfected 95-19 cells, HVEM enhanced infection by free HSV-1, as shown by an increase in the number of infectious centers obtained following infection. In both cell types, HVEM strongly enhanced entry of HSV-1 and HSV-2 by cell-cell spread, suggesting that HVEM can function as an entry mediator both in entry of free virus and in entry by cell-cell spread.
Collapse
Affiliation(s)
- R J Roller
- Department of Microbiology, University of Iowa, Iowa City 52242, USA.
| | | |
Collapse
|
36
|
Lee SK, Compton T, Longnecker R. Failure to complement infectivity of EBV and HSV-1 glycoprotein B (gB) deletion mutants with gBs from different human herpesvirus subfamilies. Virology 1997; 237:170-81. [PMID: 9344919 DOI: 10.1006/viro.1997.8765] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Glycoprotein B (gB) is conserved among the herpesvirus family which infects a broad range of species. To investigate the functional homology of human alpha-herpesviruses, beta-herpesviruses, and gamma-herpesviruses gB proteins, complementation studies were performed with gB genes from each subfamily member using EBV gp110 (EBV gB homologue) and HSV-1 gB null mutants. Neither the alpha-herpesvirus HSV-1 gB gene nor the beta-herpesvirus HCMV gB gene were able to complement the gp110 null mutant. Conversely, neither the beta-herpesvirus HCMV gB or the gamma-herpesvirus EBV gp110 gene were able to complement HSV-1 gB null mutants. To further investigate functional domains of EBV gp110 and HSV-1 gB, gB-gp110 chimeric proteins were constructed. Surprisingly, none of the chimeric proteins were able to complement either HSV-1 gB null mutants or EBV gp110 null mutants. These results demonstrate that there is not sufficient functional homology between the different gBs to allow complementation in other subfamily members of the herpesvirus family.
Collapse
Affiliation(s)
- S K Lee
- Microbiology-Immunology, Northwestern University Medical School, 303 East Chicago Avenue, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
37
|
Pertel PE, Spear PG. Partial resistance to gD-mediated interference conferred by mutations affecting herpes simplex virus type 1 gC and gK. J Virol 1997; 71:8024-8. [PMID: 9311899 PMCID: PMC192166 DOI: 10.1128/jvi.71.10.8024-8028.1997] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cells expressing herpes simplex virus (HSV) gD can be resistant to HSV entry as a result of gD-mediated interference. HSV strains differ in sensitivity to this interference, which blocks viral penetration but not binding. Previous studies have shown that mutations or variations in virion-associated gD can confer resistance to gD-mediated interference. Here we show that HSV-1 mutants selected for enhanced ability to bind and penetrate in the presence of inhibitory concentrations of heparin were partially resistant to gD-mediated interference. The resistance was largely due to the presence of two mutations: one in gC (the major heparin-binding glycoprotein) resulting in the absence of gC expression and the other in gK resulting in a syncytial phenotype. The results imply that heparin selected for mutants with altered postbinding requirements for entry. Resistance to gD-mediated interference conferred by mutations affecting gC and gK has not been previously described.
Collapse
Affiliation(s)
- P E Pertel
- Department of Medicine, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | |
Collapse
|
38
|
Cohen JI, Nguyen H. Varicella-zoster virus glycoprotein I is essential for growth of virus in Vero cells. J Virol 1997; 71:6913-20. [PMID: 9261418 PMCID: PMC191974 DOI: 10.1128/jvi.71.9.6913-6920.1997] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Varicella-zoster virus (VZV) encodes at least six glycoproteins. Glycoprotein I (gI), the product of open reading frame 67, is a 58- to 62-kDa glycoprotein found in VZV-infected cells. We constructed two VZV gI deletion mutants. Immunoprecipitation of VZV gE from infected cells indicated that cells infected with VZV deleted for gI expressed a gE that was larger (100 kDa) than that expressed in cells infected with the parental virus (98 kDa). Cell-associated or cell-free VZV deleted for gI grew to lower titers in melanoma cells than did parental VZV. While VZV deleted for gI replicated in other human cells, the mutant virus replicated to very low titers in primary guinea pig and monkey cells and did not replicate in Vero cells. When compared with the parental virus, rescued viruses, in which the gI deletion was restored with a wild-type allele, showed a similarly sized gE and comparable growth patterns in melanoma and Vero cells. VZV deleted for gI entered Vero cells; however, viral DNA synthesis was impaired in these cells. The VZV gI mutant was slightly impaired for adsorption to human cells. Thus, VZV gI is required for replication of the virus in Vero cells, for efficient replication of the virus in nonhuman cells, and for normal processing of gE.
Collapse
Affiliation(s)
- J I Cohen
- Medical Virology Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
39
|
Roller RJ, Herold BC. Characterization of a BHK(TK-) cell clone resistant to postattachment entry by herpes simplex virus types 1 and 2. J Virol 1997; 71:5805-13. [PMID: 9223469 PMCID: PMC191835 DOI: 10.1128/jvi.71.8.5805-5813.1997] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BHK(TK-) cells selected for resistance to polyethylene glycol-mediated fusion give rise to clones that are resistant to herpes simplex virus (HSV) infection. We have characterized one such clone, designated 95-19, and found that it is resistant to entry of HSV type 1 (HSV-1), HSV-2, and the related alphaherpesvirus pseudorabies virus (PRV). Single-step growth experiments show no detectable replication of multiple strains of HSV-1 and HSV-2 on 95-19 cells. Three lines of evidence suggest that these cells are resistant to postattachment entry. (i) Measurements of binding of radiolabeled virus show that heparin-sensitive binding of HSV-1 and HSV-2 to 95-19 cells is identical to binding to BHK(TK-) cells, suggesting that the block to replication occurs after attachment to heparan sulfate proteoglycan. (ii) 95-19 cells exposed to HSV-1 or HSV-2 at high multiplicity show no detectable immediate-early (IE) mRNA expression. (iii) Exposure of attached virus and cells to polyethylene glycol results in partial recovery of both IE gene expression and virus yield in single-step growth. The degrees of recovery of single-step yield and IE gene expression are similar, suggesting that the only block to single-step replication is at the point of virus entry and that these cells are deficient in some cellular factor required for efficient postattachment entry of free virus. 95-19 cells are also highly resistant to entry by cell-to-cell spread, suggesting that the same cellular factor participates in both types of entry.
Collapse
Affiliation(s)
- R J Roller
- Department of Microbiology, University of Iowa, Iowa City 52242, USA
| | | |
Collapse
|
40
|
Hata A, Mukai T, Isegawa Y, Yamanishi K. Identification and analyses of glycoprotein B of human herpesvirus 7. Virus Res 1996; 46:125-37. [PMID: 9029785 DOI: 10.1016/s0168-1702(96)01395-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The gene for the human herpes virus 7 (HHV-7) glycoprotein B (gB) has been identified by sequencing a molecularly cloned HHV-7 DNA fragment. A 2.5-kb open reading frame (ORF) encoded a protein of 822 amino acids with characteristics of a transmembrane glycoprotein, and showed the strongest similarity (56.5%) with the human herpesvirus 6 (HHV-6) gB. The genes for the transport/capsid assembly protein (tp/cap) and the DNA polymerase (pol) existed upstream and downstream of the gB gene, respectively. This arrangement was the same as that of HHV-6. Antisera were generated by immunizing mice with a glutathione S-transferase-carboxy terminal gB fusion protein. Immunofluorescent tests demonstrated that the antisera reacted specifically with HHV-7 antigens in cytoplasm of infected cells. The antisera immunoprecipitated proteins with apparent molecular masses of 51, 63 and 112 kDa from HHV-7 infected cells by pulse-chase analysis. In the presence of tunicamycin, the protein with a molecular mass of 112 kDa was replaced by a protein with a molecular mass of 88 kDa, and this size was consistent with the predicted size of the primary translation product of the HHV-7 gB gene. These results suggested that the protein with a molecular mass of 112 kDa was a glycoprotein synthesized by addition of N-linked oligosaccharides to a non-glycosylated precursor of the protein with a molecular mass of 88 kDa and then cleaved into the proteins with molecular masses of 51 and 63 kDa in HHV-7 infected cells.
Collapse
Affiliation(s)
- A Hata
- Department of Virology, Osaka University, Japan
| | | | | | | |
Collapse
|
41
|
Norais N, Tang D, Kaur S, Chamberlain SH, Masiarz FR, Burke RL, Marcus F. Disulfide bonds of herpes simplex virus type 2 glycoprotein gB. J Virol 1996; 70:7379-87. [PMID: 8892856 PMCID: PMC190805 DOI: 10.1128/jvi.70.11.7379-7387.1996] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Glycoprotein B (gB) is the most highly conserved envelope glycoprotein of herpesviruses. The gB protein is required for virus infectivity and cell penetration. Recombinant forms of gB being used for the development of subunit vaccines are able to induce virus-neutralizing antibodies and protective efficacy in animal models. To gain structural information about the protein, we have determined the location of the disulfide bonds of a 696-amino-acid residue truncated, recombinant form of herpes simplex virus type 2 glycoprotein gB (HSV gB2t) produced by expression in Chinese hamster ovary cells. The purified protein, which contains virtually the entire extracellular domain of herpes simplex virus type 2 gB, was digested with trypsin under nonreducing conditions, and peptides were isolated by reversed-phase high-performance liquid chromatography (HPLC). The peptides were characterized by using mass spectrometry and amino acid sequence analysis. The conditions of cleavage (4 M urea, pH 7) induced partial carbamylation of the N termini of the peptides, and each disulfide peptide was found with two or three different HPLC retention times (peptides with and without carbamylation of either one or both N termini). The 10 cysteines of the molecule were found to be involved in disulfide bridges. These bonds were located between Cys-89 (C1) and Cys-548 (C8), Cys-106 (C2) and Cys-504 (C7), Cys-180 (C3) and Cys-244 (C4), Cys-337 (C5) and Cys-385 (C6), and Cys-571 (C9) and Cys-608 (C10). These disulfide bonds are anticipated to be similar in the corresponding gBs from other herpesviruses because the 10 cysteines listed above are always conserved in the corresponding protein sequences.
Collapse
Affiliation(s)
- N Norais
- Chiron Corporation, Emeryville, California 94608, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Montgomery RI, Warner MS, Lum BJ, Spear PG. Herpes simplex virus-1 entry into cells mediated by a novel member of the TNF/NGF receptor family. Cell 1996; 87:427-36. [PMID: 8898196 DOI: 10.1016/s0092-8674(00)81363-x] [Citation(s) in RCA: 951] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We identified and cloned a cellular mediator of herpes simplex virus (HSV) entry. Hamster and swine cells resistant to viral entry became susceptible upon expression of a human cDNA encoding this protein, designated HVEM (for herpesvirus entry mediator). HVEM was shown to mediate the entry of several wild-type HSV strains of both serotypes. Anti-HVEM antibodies and a soluble hybrid protein containing the HVEM ectodomain inhibited HVEM-dependent infection but not virus binding to cells. Mutations in the HSV envelope glycoprotein gD significantly reduced HVEM-mediated entry. The contribution of HVEM to HSV entry into human cells was demonstrable in activated T cells. HVEM, the first identified mediator of HSV entry, is a new member of the TNF/NGF receptor family.
Collapse
Affiliation(s)
- R I Montgomery
- Northwestern University Medical School, Department of Microbiology-Immunology, Chicago, Illinois 60611, USA
| | | | | | | |
Collapse
|
43
|
Lin XH, Ali MA, Openshaw H, Cantin EM. Deletion of the carboxy-terminus of herpes simplex virus type 1 (HSV-1) glycoprotein B does not affect oligomerization, heparin-binding activity, or its ability to protect against HSV challenge. Arch Virol 1996; 141:1153-65. [PMID: 8712932 DOI: 10.1007/bf01718618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A recombinant vaccinia virus designated VgBt which expresses a truncated secreted herpes simplex virus gB (gBt) was constructed and compared to V11gB, a vaccinia recombinant previously studied which expresses gB exclusively on the surface of infected cells. Indirect immunofluorescence assay (IFA) revealed that gBt was strongly associated with the surface of infected cells despite being released slowly into the cell culture medium. Both gB and gBt existed as oligomers, and both membrane bound and secreted forms of gBt exhibited heparin-binding activity. In protection studies VgBt and V11gB conferred equivalent protection against both homologous (HSV-1) and heterologous (HSV-2) challenge with HSV.
Collapse
MESH Headings
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Disease Models, Animal
- Female
- Heparin/metabolism
- Herpes Simplex/immunology
- Herpes Simplex/prevention & control
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/immunology
- Humans
- Mice
- Mice, Inbred BALB C
- Sequence Deletion
- Structure-Activity Relationship
- Vaccination
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/metabolism
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/metabolism
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Viral Vaccines/metabolism
Collapse
Affiliation(s)
- X H Lin
- Department of Neurology, City of Hope National Medical Center, Deuarte, California, USA
| | | | | | | |
Collapse
|
44
|
Goade DE, Bell R, Yamada T, Mertz GJ, Jenison S. Locations of herpes simplex virus type 2 glycoprotein B epitopes recognized by human serum immunoglobulin G antibodies. J Virol 1996; 70:2950-6. [PMID: 8627770 PMCID: PMC190153 DOI: 10.1128/jvi.70.5.2950-2956.1996] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Herpes simplex virus type 2 (HSV-2) glycoprotein B (gB-2) gene segments were expressed as recombinant proteins in Escherichia coli. gB-2 recombinant proteins were reacted with human serum immunoglobulin G (IgG) antibodies in Western immunoblot assays. Initially, samples were tested for the presence of HSV-1-specific antibodies and HSV-2-specific antibodies by using HSV-infected cell lysates as antigen targets in Western blot assays. Serum samples that contained HSV-2-specific IgG (n = 58), HSV-1-specific IgG (n = 33), or no detectable HSV antibodies (n = 31) were tested for reactivities with the gB-2 recombinant proteins. In 58 of 58 samples that contained HSV-2-specific IgG, antibodies were present that reacted strongly with a gB-2 amino-proximal segment between amino acids (aa) 18 and 75. Three of 33 serum samples that contained HSV-1- and not HSV-2-specific IgG (as defined by the HSV lysate Western blot assay) reacted with this segment. Both HSV-2 antibodies and HSV-1 antibodies reacted strongly with a carboxy-terminal gB-2 segment between aa 819 and 904; a second minor cross-reactive region was mapped to a gB-2 segment between aa 564 and 626. The gB-2 segment from aa 18 to 75 may constitute a useful reagent for the virus type-specific serodiagnosis of HSV-2 infections. Further studies will be required to determine the relative sensitivities and specificities of the assay for gB-2 aa 18 to 75, HSV gG assays, and HSV lysate Western blot assays for detecting virus type-specific antibody responses in acute and chronic HSV-2 infections.
Collapse
Affiliation(s)
- D E Goade
- Department of Medicine, University of New Mexico School of Medicine, Albuquerque 87131-5271, USA
| | | | | | | | | |
Collapse
|
45
|
Herrold RE, Marchini A, Fruehling S, Longnecker R. Glycoprotein 110, the Epstein-Barr virus homolog of herpes simplex virus glycoprotein B, is essential for Epstein-Barr virus replication in vivo. J Virol 1996; 70:2049-54. [PMID: 8627735 PMCID: PMC190038 DOI: 10.1128/jvi.70.3.2049-2054.1996] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The Epstein-Barr virus (EBV) glycoprotein gp110 has substantial amino acid homology to gB of herpes simplex virus but localizes differently within infected cells and is essentially undetectable in virions. To investigate whether gp110, like gB, is essential for EBV infection, a selectable marker was inserted within the gp110 reading frame, BALF4, and the resulting null mutant EBV stain, B95-110HYG, was recovered in lymphoblastoid cell lines (LCLs). While LCLs infected with the parental virus B95-8 expressed the gp110 protein product following productive cycle induction, neither full-length gp110 nor the predicted gp110 truncation product was detectable in B95-110HYG LCLs. Infectious virus could not be recovered from B95-110HYG LCLs unless gp110 was provided in trans. Rescued B95-110HYG virus latently infected and growth transformed primary B lymphocytes. Thus, gp110 is required for the production of transforming virus but not for the maintenance of transformation of primary B lymphocytes by EBV.
Collapse
Affiliation(s)
- R E Herrold
- Department of Microbiology--Immunology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | | | |
Collapse
|
46
|
Sawitzky D, Voigt A, Zeichhardt H, Habermehl KO. Glycoprotein B (gB) of pseudorabies virus interacts specifically with the glycosaminoglycan heparin. Virus Res 1996; 41:101-8. [PMID: 8725106 DOI: 10.1016/0168-1702(95)01277-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We have previously shown that the pseudorabies virus (PrV) glycoproteins gB and gC (former PrV-gII and PrV-gIII) exhibit heparin-binding properties. While PrV-gC functions as the major adsorption protein, the biological role of the heparin-binding properties of PrV-gB are not understood. We used a gC-deleted PrV-mutant, PrV (dlg92/dltk), to analyse the heparin-binding properties of PrV-gB and the biological role of the PrV-gB-protein in adsorption. PrV-gB was the only glycoprotein of this vaccine strain binding to immobilised heparin in in vitro assays. Presence of the gC-protein was not necessary for the interaction of gB with heparin. Soluble heparin also interfered with adsorption of this mutant virus to a similar extent as it blocked adsorption of wild-type PrV (Ka), but it had only a minor inhibitory effect on infectivity of the mutant strain. These results show that PrV-gB interacts specifically with immobilized heparin and heparin-like structures on the cell surface, but this interaction is not required for a productive infection.
Collapse
Affiliation(s)
- D Sawitzky
- Institute for Clinical and Experimental Virology, Free University Berlin, Germany.
| | | | | | | |
Collapse
|
47
|
Lingen M, Seck T, Weise K, Falke D. Two mutations in gB-1 and gD-1 of herpes simplex virus type 1 are involved in the "fusion from without" phenotype in different cell types. Virus Genes 1996; 13:221-8. [PMID: 9035366 DOI: 10.1007/bf00366982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Previous studies have shown that certain strains of herpes simplex viruses type 1 (HSV-1) are able to induce "fusion from without" (FFWO) which means no transcription or translation of the viral genome happens. The main determinants for FFWO in BHK cells are mutations in the C-terminal part of gB-1. But single mutations in this part of the genome are not sufficient to transfer the FFWO phenotype also to Vero cells. Here, we report that FFWO of HSV strains indeed need additional mutations in the N-terminal part of gD in order to produce the FFWO phenotype in BHK and Vero cells. By marker transfer we are able to show that loss of mutations in the N-terminal part of gD influences the ability to induce FFWO in Vero cells but not in BHK cells. We assume that a mutated gD allows the entrance of a multiple number of virus particles into the cell and enhances therefore the fusion activity of the mutated gB. Mutations in gD alone are not sufficient for fusion activity of HSV.
Collapse
Affiliation(s)
- M Lingen
- Institut für Virologie J. Gutenburg Universität Mainz, Germany
| | | | | | | |
Collapse
|
48
|
Oxford JS, al-Jabri AA, Stein CA, Levantis P. Analysis of resistance mutants of viral polymerases. Methods Enzymol 1996; 275:555-600. [PMID: 9026659 DOI: 10.1016/s0076-6879(96)75031-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- J S Oxford
- Academic Virology and Retroscreen Ltd., The London Hospital Medical College, Whitechapel, England
| | | | | | | |
Collapse
|
49
|
Billstrom MA, Britt WJ. Postoligomerization folding of human cytomegalovirus glycoprotein B: identification of folding intermediates and importance of disulfide bonding. J Virol 1995; 69:7015-22. [PMID: 7474121 PMCID: PMC189621 DOI: 10.1128/jvi.69.11.7015-7022.1995] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Human cytomegalovirus glycoprotein B (gB or UL55) has been demonstrated to be a disulfide-linked homodimer within the envelope of mature virions. Previously, it has been shown that gB undergoes a rapid dimerization nearly coincident with its synthesis. Following dimerization, the molecule slowly folds into a form which can be transported from the endoplasmic reticulum. In this study we have examined the prolonged folding of gB by using a set of defined gB-reactive murine monoclonal antibodies and gB expressed as a recombinant protein in the absence of other human cytomegalovirus proteins. Our results have documented a folding pathway consistent with the relatively rapid dimerization of the translation product followed by delayed conversion into a fully folded molecule. Assembly of the dominant antigenic domain of gB, AD-1, preceded dimerization and folding of the molecule. The fully folded dimer was heat stable, but its conformation was altered by treatment with 2% sodium dodecyl sulfate (SDS), whereas an oligomeric folding intermediate was both heat and SDS stable. Postoligomerization disulfide bond formation could be demonstrated during folding of gB, suggesting that the formation of these covalent bonds could contribute to the prolonged folding of this glycoprotein.
Collapse
Affiliation(s)
- M A Billstrom
- Department of Pediatrics, University of Alabama at Birmingham, School of Medicine 35233, USA
| | | |
Collapse
|
50
|
Maeda K, Hirasawa K, Kawaguchi Y, Ono M, Mori T, Gemma T, Yokoyama N, Doi K, Mikami T. Expression and identification of the feline herpesvirus type 1 glycoprotein B (gp143/108). Virus Res 1995; 39:55-61. [PMID: 8607284 DOI: 10.1016/s0168-1702(95)00059-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gene for feline herpesvirus type 1 (FHV-1) glycoprotein B (gB) has been cloned into an expression vector, pRVSVneo, containing the long terminal repeat of Rous sarcoma virus and polyadenylation signal of SV40. This expression vector containing FHV-1 gB gene, pRVSVgBneo, was transfected into Crandell feline kidney (CRFK) cells which are susceptible to FHV-1 infection. By indirect immunofluorescence analysis, the expressed gB was recognized with a panel of monoclonal antibodies (MAbs) against FHV-1 gp143/108. Immunoprecipitation analysis using a MAb 34H12 showed that molecular weights of the gB were 143 and 108 kDa under non-denaturing conditions that 108, 70, 64, and 58 kDa under denaturing conditions. The molecular weights were similar to those of the gB expressed in FHV-1-infected CRFK cells. In addition, when plasmid DNAs were injected into mice to obtain gB-monospecific serum, the pooled serum from mice inoculated with pRVSVgBneo, but not with pRVSVgDneo or pRVSVneo, recognized the FHV-1 gB polypeptides.
Collapse
Affiliation(s)
- K Maeda
- Department of Veterinary Microbiology, Faculty of Agriculture, University of Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|