Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
2.
PLoS Pathog ; 17(8): e1009824, 2021 08.
Article in English | MEDLINE | ID: mdl-34398933

ABSTRACT

The herpes simplex virus (HSV)-1 protein pUL21 is essential for efficient virus replication and dissemination. While pUL21 has been shown to promote multiple steps of virus assembly and spread, the molecular basis of its function remained unclear. Here we identify that pUL21 is a virus-encoded adaptor of protein phosphatase 1 (PP1). pUL21 directs the dephosphorylation of cellular and virus proteins, including components of the viral nuclear egress complex, and we define a conserved non-canonical linear motif in pUL21 that is essential for PP1 recruitment. In vitro evolution experiments reveal that pUL21 antagonises the activity of the virus-encoded kinase pUS3, with growth and spread of pUL21 PP1-binding mutant viruses being restored in adapted strains where pUS3 activity is disrupted. This study shows that virus-directed phosphatase activity is essential for efficient herpesvirus assembly and spread, highlighting the fine balance between kinase and phosphatase activity required for optimal virus replication.


Subject(s)
Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Phosphoric Monoester Hydrolases/metabolism , Viral Proteins/metabolism , Virus Assembly , Virus Replication , Animals , Chlorocebus aethiops , HEK293 Cells , Herpesvirus 1, Human/enzymology , Humans , Phosphoric Monoester Hydrolases/genetics , Vero Cells , Viral Proteins/genetics , Virus Release
3.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27852850

ABSTRACT

The tegument of herpesviruses is a highly complex structural layer between the nucleocapsid and the envelope of virions. Tegument proteins play both structural and regulatory functions during replication and spread, but the interactions and functions of many of these proteins are poorly understood. Here we focus on two tegument proteins from herpes simplex virus 1 (HSV-1), pUL7 and pUL51, which have homologues in all other herpesviruses. We have now identified that HSV-1 pUL7 and pUL51 form a stable and direct protein-protein interaction, their expression levels rely on the presence of each other, and they function as a complex in infected cells. We demonstrate that expression of the pUL7-pUL51 complex is important for efficient HSV-1 assembly and plaque formation. Furthermore, we also discovered that the pUL7-pUL51 complex localizes to focal adhesions at the plasma membrane in both infected cells and in the absence of other viral proteins. The expression of pUL7-pUL51 is important to stabilize focal adhesions and maintain cell morphology in infected cells and cells infected with viruses lacking pUL7 and/or pUL51 round up more rapidly than cells infected with wild-type HSV-1. Our data suggest that, in addition to the previously reported functions in virus assembly and spread for pUL51, the pUL7-pUL51 complex is important for maintaining the attachment of infected cells to their surroundings through modulating the activity of focal adhesion complexes. IMPORTANCE: Herpesviridae is a large family of highly successful human and animal pathogens. Virions of these viruses are composed of many different proteins, most of which are contained within the tegument, a complex structural layer between the nucleocapsid and the envelope within virus particles. Tegument proteins have important roles in assembling virus particles as well as modifying host cells to promote virus replication and spread. However, little is known about the function of many tegument proteins during virus replication. Our study focuses on two tegument proteins from herpes simplex virus 1 that are conserved in all herpesviruses: pUL7 and pUL51. We demonstrate that these proteins directly interact and form a functional complex that is important for both virus assembly and modulation of host cell morphology. Further, we identify for the first time that these conserved herpesvirus tegument proteins localize to focal adhesions in addition to cytoplasmic juxtanuclear membranes within infected cells.


Subject(s)
DNA Helicases/metabolism , DNA Primase/metabolism , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Multiprotein Complexes/metabolism , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , DNA Helicases/genetics , DNA Primase/genetics , Gene Expression Regulation, Viral , HEK293 Cells , Herpesvirus 1, Human/ultrastructure , Humans , Protein Binding , Protein Transport , Vero Cells , Viral Matrix Proteins/genetics , Viral Proteins/genetics , Virus Assembly
4.
J Gen Virol ; 92(Pt 7): 1550-1560, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21471322

ABSTRACT

The difficulty of eliminating herpesvirus carriage makes host entry a key target for infection control. However, its viral requirements are poorly defined. Murid herpesvirus-4 (MuHV-4) can potentially provide insights into gammaherpesvirus host entry. Upper respiratory tract infection requires the MuHV-4 thymidine kinase (TK) and ribonucleotide reductase large subunit (RNR-L), suggesting a need for increased nucleotide production. However, both TK and RNR-L are likely to be multifunctional. We therefore tested further the importance of nucleotide production by disrupting the MuHV-4 ribonucleotide reductase small subunit (RNR-S). This caused a similar attenuation to RNR-L disruption: despite reduced intra-host spread, invasive inoculations still established infection, whereas a non-invasive upper respiratory tract inoculation did so only at high dose. Histological analysis showed that RNR-S(-), RNR-L(-) and TK(-) viruses all infected cells in the olfactory neuroepithelium but unlike wild-type virus then failed to spread. Thus captured host nucleotide metabolism enzymes, up to now defined mainly as important for alphaherpesvirus reactivation in neurons, also have a key role in gammaherpesvirus host entry. This seemed to reflect a requirement for lytic replication to occur in a terminally differentiated cell before a viable pool of latent genomes could be established.


Subject(s)
Rhadinovirus/enzymology , Ribonucleotide Reductases/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Female , Herpesviridae Infections/virology , Humans , Mice , Mice, Inbred BALB C , Protein Subunits/genetics , Protein Subunits/metabolism , Rhadinovirus/genetics , Rhadinovirus/physiology , Ribonucleotide Reductases/genetics , Viral Proteins/genetics , Virus Replication
5.
J Virol ; 84(20): 10937-42, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20668075

ABSTRACT

Viral enzymes that process small molecules provide potential chemotherapeutic targets. A key constraint-the replicative potential of spontaneous enzyme mutants-has been hard to define with human gammaherpesviruses because of their narrow species tropisms. Here, we disrupted the murid herpesvirus 4 (MuHV-4) ORF61, which encodes its ribonucleotide reductase (RNR) large subunit. Mutant viruses showed delayed in vitro lytic replication, failed to establish infection via the upper respiratory tract, and replicated to only a very limited extent in the lower respiratory tract without reaching lymphoid tissue. RNR could therefore provide a good target for gammaherpesvirus chemotherapy.


Subject(s)
Rhadinovirus/enzymology , Rhadinovirus/pathogenicity , Ribonucleotide Reductases/physiology , Animals , Base Sequence , Cell Line , Cricetinae , DNA, Viral/genetics , Genes, Viral , Herpesviridae Infections/virology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutagenesis, Insertional , Respiratory System/virology , Rhadinovirus/genetics , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/genetics , Tumor Virus Infections/virology , Virulence/genetics , Virulence/physiology , Virus Replication/genetics , Virus Replication/physiology
6.
J Gen Virol ; 90(Pt 11): 2592-2603, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19625459

ABSTRACT

Antibody is an important antiviral defence. However, it is considered to do little against human gamma-herpesviruses, which establish predominantly latent infections regulated by T cells. One limitation on analysing these infections has been that latency is already well-established at clinical presentation; early infection may still be accessible to antibody. Here, using murid herpesvirus-4 (MuHV-4), we tested the impact of adoptively transferred antibody on early gamma-herpesvirus infection. Immune sera and neutralizing and non-neutralizing monoclonal antibodies (mAbs) all reduced acute lytic MuHV-4 replication. The reductions, even by neutralizing mAbs, were largely or completely dependent on host IgG Fc receptors. Therefore, passive antibody can blunt acute gamma-herpesvirus lytic infection, and does this principally by IgG Fc-dependent functions rather than by neutralization.


Subject(s)
Antibodies, Viral/immunology , Immunoglobulin G/immunology , Receptors, Fc/immunology , Rhadinovirus/immunology , Rhadinovirus/physiology , Virus Replication , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Serum/virology , Viral Plaque Assay , Whole Body Imaging
7.
J Gen Virol ; 90(Pt 5): 1202-1214, 2009 May.
Article in English | MEDLINE | ID: mdl-19264603

ABSTRACT

Antibodies readily neutralize acute, epidemic viruses, but are less effective against more indolent pathogens such as herpesviruses. Murid herpesvirus 4 (MuHV-4) provides an accessible model for tracking the fate of antibody-exposed gammaherpesvirus virions. Glycoprotein L (gL) plays a central role in MuHV-4 entry: it allows gH to bind heparan sulfate and regulates fusion-associated conformation changes in gH and gB. However, gL is non-essential: heparan sulfate binding can also occur via gp70, and the gB-gH complex alone seems to be sufficient for membrane fusion. Here, we investigated how gL affects the susceptibility of MuHV-4 to neutralization. Immune sera neutralized gL(-) virions more readily than gL(+) virions, chiefly because heparan sulfate binding now depended on gp70 and was therefore easier to block. However, there were also post-binding effects. First, the downstream, gL-independent conformation of gH became a neutralization target; gL normally prevents this by holding gH in an antigenically distinct heterodimer until after endocytosis. Second, gL(-) virions were more vulnerable to gB-directed neutralization. This covered multiple epitopes and thus seemed to reflect a general opening up of the gH-gB entry complex, which gL again normally restricts to late endosomes. gL therefore limits MuHV-4 neutralization by providing redundancy in cell binding and by keeping key elements of the virion fusion machinery hidden until after endocytosis.


Subject(s)
Antibodies, Viral/immunology , Glycoproteins/immunology , Rhadinovirus/immunology , Rhadinovirus/metabolism , Viral Envelope Proteins/immunology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Epithelial Cells , Female , Fibroblasts , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Binding
8.
PLoS One ; 3(2): e1669, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18301747

ABSTRACT

The first contact a virus makes with cells is an important determinant of its tropism. Murid Herpesvirus-4 (MuHV-4) is highly dependent on glycosaminoglycans (GAGs) for cell binding. Its first contact is therefore likely to involve a GAG-binding virion glycoprotein. We have previously identified two such proteins, gp70 and gp150. Gp70 binds strongly to GAGs. However, deleting it makes little difference to MuHV-4 cell binding or GAG-dependence. Deleting gp150, by contrast, frees MuHV-4 from GAG dependence. This implies that GAGs normally displace gp150 to allow GAG-independent cell binding. But the gp150 GAG interaction is weak, and so would seem unlikely to make an effective first contact. Since neither gp70 nor gp150 matches the expected profile of a first contact glycoprotein, our understanding of MuHV-4 GAG interactions must be incomplete. Here we relate the seemingly disconnected gp70 and gp150 GAG interactions by showing that the MuHV-4 gH/gL also binds to GAGs. gH/gL-blocking and gp70-blocking antibodies individually had little effect on cell binding, but together were strongly inhibitory. Thus, there was redundancy in GAG binding between gp70 and gH/gL. Gp150-deficient MuHV-4 largely resisted blocks to gp70 and gH/gL binding, consistent with its GAG independence. The failure of wild-type MuHV-4 to do the same argues that gp150 is normally engaged only down-stream of gp70 or gH/gL. MuHV-4 GAG dependence is consequently two-fold: gp70 or gH/gL binding provides virions with a vital first foothold, and gp150 is then engaged to reveal GAG-independent binding.


Subject(s)
Glycosaminoglycans/metabolism , Receptors, Virus , Rhadinovirus/chemistry , Viral Envelope Proteins/metabolism , Glycoproteins/metabolism , Protein Binding
9.
J Gen Virol ; 89(Pt 6): 1352-1363, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18474550

ABSTRACT

Herpesviruses are ancient pathogens that infect all vertebrates. The most conserved component of their entry machinery is glycoprotein B (gB), yet how gB functions is unclear. A striking feature of the murid herpesvirus 4 (MuHV-4) gB is its resistance to neutralization. Here, we show by direct visualization of infected cells that the MuHV-4 gB changes its conformation between extracellular virions and those in late endosomes, where capsids are released. Specifically, epitopes on its N-terminal cell-binding domain become inaccessible, whilst non-N-terminal epitopes are revealed, consistent with structural changes reported for the vesicular stomatitis virus glycoprotein G. Inhibitors of endosomal acidification blocked the gB conformation switch. They also blocked capsid release and the establishment of infection, implying that the gB switch is a key step in entry. Neutralizing antibodies could only partially inhibit the switch. Their need to engage a less vulnerable, upstream form of gB, because its fusion form is revealed only in endosomes, helps to explain why gB-directed MuHV-4 neutralization is so difficult.


Subject(s)
Glycoproteins/metabolism , Herpesviridae Infections/virology , Rhadinovirus/physiology , Tumor Virus Infections/virology , Viral Proteins/metabolism , Animals , Antibodies, Viral/immunology , Cell Line , Cricetinae , Cricetulus , Humans , Mice , Neutralization Tests , Protein Conformation , Rhadinovirus/chemistry , Virus Attachment
10.
PLoS One ; 3(7): e2811, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18665235

ABSTRACT

The glycoprotein H (gH)/gL heterodimer is crucial for herpesvirus membrane fusion. Yet how it functions is not well understood. The Murid Herpesvirus-4 gH, like that of other herpesviruses, adopts its normal virion conformation by associating with gL. However, gH switched back to a gL-independent conformation after virion endocytosis. This switch coincided with a conformation switch in gB and with capsid release. Virions lacking gL constitutively expressed the down-stream form of gH, prematurely switched gB to its down-stream form, and showed premature capsid release with poor infectivity. These data argue that gL plays a key role in regulating a gH and gB functional switch from cell binding to membrane fusion.


Subject(s)
Membrane Glycoproteins/physiology , Molecular Chaperones/physiology , Rhadinovirus/metabolism , Viral Envelope Proteins/physiology , Viral Proteins/physiology , Animals , Cricetinae , Dimerization , Endocytosis , Endosomes/metabolism , Glycoproteins/chemistry , Hydrogen-Ion Concentration , Membrane Fusion , Membrane Glycoproteins/metabolism , Mice , Molecular Chaperones/metabolism , NIH 3T3 Cells , Protein Conformation , Viral Envelope Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism
11.
PLoS One ; 2(8): e705, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17684552

ABSTRACT

Herpesviruses maintain long-term infectivity without marked antigenic variation. They must therefore evade neutralization by other means. Immune sera block murine gammaherpesvirus-68 (MHV-68) infection of fibroblasts, but fail to block and even enhance its infection of IgG Fc receptor-bearing cells, suggesting that the antibody response to infection is actually poor at ablating virion infectivity completely. Here we analyzed this effect further by quantitating the glycoprotein-specific antibody response of MHV-68 carrier mice. Gp150 was much the commonest glycoprotein target and played a predominant role in driving Fc receptor-dependent infection: when gp150-specific antibodies were boosted, Fc receptor-dependent infection increased; and when gp150-specific antibodies were removed, Fc receptor-dependent infection was largely lost. Neither gp150-specific monoclonal antibodies nor gp150-specific polyclonal sera gave significant virion neutralization. Gp150 therefore acts as an immunogenic decoy, distorting the MHV-68-specific antibody response to promote Fc receptor-dependent infection and so compromise virion neutralization. This immune evasion mechanism may be common to many non-essential herpesvirus glycoproteins.


Subject(s)
Antibodies, Neutralizing/immunology , Glycoproteins/immunology , Rhadinovirus/immunology , Viral Proteins/immunology , Virion/immunology , Animals , Antibodies, Monoclonal/immunology , Epitope Mapping , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Fc/immunology
12.
J Virol ; 81(1): 280-91, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050601

ABSTRACT

The herpesvirus glycoprotein H (gH) and gL associate to form a heterodimer that plays a central role in virus-driven membrane fusion. When archetypal alpha- or betaherpesviruses lack gL, gH misfolds and progeny virions are noninfectious. In order to define the role that gL plays in gamma-2 herpesvirus infections, we disrupted its coding sequence in murine gammaherpesvirus-68 (MHV-68). MHV-68 lacking gL folded gH into a conformation antigenically distinct from the form that normally predominates on infected cells. gL-deficient virions bound less well than the wild type to epithelial cells and fibroblasts. However, they still incorporated gH and remained infectious. The cell-to-cell spread of gL-deficient viruses was remarkably normal, as was infection, dissemination, and latency establishment in vivo. Viral membrane fusion was therefore gL independent. The major function of gL appeared to be allowing gH to participate in cell binding prior to membrane fusion. This function was most important for the entry of MHV-68 virions into fibroblasts and epithelial cells.


Subject(s)
Membrane Glycoproteins/metabolism , Rhadinovirus/physiology , Viral Fusion Proteins/metabolism , Animals , Antibodies, Monoclonal , Cell Line , Cricetinae , Female , Kinetics , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mutation , Protein Folding , Protein Structure, Tertiary , Rhadinovirus/genetics , Rhadinovirus/pathogenicity , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Virion/genetics , Virion/metabolism , Virion/physiology , Virus Latency/genetics
13.
J Gen Virol ; 87(Pt 12): 3515-3527, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17098966

ABSTRACT

Persistent viruses disseminate from immune hosts. They must therefore resist neutralization by antibody. Murine gammaherpesvirus-68 (MHV-68) represents an accessible model with which to address how resistance to neutralization is achieved and how overcoming it might improve infection control. The MHV-68 glycoprotein B (gB), like that of other herpesviruses, is a virion protein that is essential for infectivity. As such, it presents a potential neutralization target. In order to test whether virus-induced antibodies reduce virion infectivity by binding to gB, monoclonal antibodies (mAbs) were derived from MHV-68-infected mice. gB-specific mAbs were common, but only an IgM specific for the gB N terminus reduced virion infectivity significantly. It inhibited MHV-68 entry into BHK-21 cells at a post-binding step that was linked closely to membrane fusion. Reducing the mAb to IgM monomers compromised neutralization severely, suggesting that a pentameric structure was crucial to its function. Antibody treatment never blocked BHK-21 cell infection completely and blocked the infection of NMuMG epithelial cells hardly at all. Virions saturated with antibody also remained infectious to mice. Thus, the MHV-68 gB presents at best a very difficult target for antibody-mediated neutralization.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Rhadinovirus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Antigens, Viral/immunology , Cell Line , Cricetinae , Disease Models, Animal , Female , Herpesviridae Infections/immunology , Immunoglobulin M/immunology , Immunoglobulin M/isolation & purification , Lung/virology , Membrane Fusion , Mice , Mice, Inbred BALB C , Neutralization Tests , Rhadinovirus/physiology , Viral Plaque Assay , Virus Internalization
14.
J Gen Virol ; 87(Pt 6): 1465-1475, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16690911

ABSTRACT

Herpesviruses characteristically persist in immune hosts as latent genomes, but to transmit infection they must reactivate and replicate lytically. The interaction between newly formed virions and pre-existing antibody is therefore likely to be a crucial determinant of viral fitness. Murine gammaherpesvirus-68 (MHV-68) behaves as a natural pathogen of conventional, inbred mice and consequently allows such interactions to be analysed experimentally in a relatively realistic setting. Here, monoclonal antibodies (mAbs) were derived from MHV-68-infected mice and all those recognizing infected-cell surfaces were tested for their capacity to neutralize MHV-68 virions. All of the neutralizing mAbs identified were specific for the viral glycoprotein H (gH)-gL heterodimer and required both gH and gL to reproduce their cognate epitopes. Based on antibody interference, there appeared to be two major neutralization epitopes on gH-gL. Analysis of a representative mAb indicated that it blocked infection at a post-binding step--either virion endocytosis or membrane fusion.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibody Specificity , Rhadinovirus/immunology , Viral Envelope Proteins/immunology , Animals , Cell Line , Cricetinae , Epitopes/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Mice , Neutralization Tests , Rhadinovirus/metabolism , Rhadinovirus/pathogenicity , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Virion/immunology
15.
J Virol ; 79(6): 3459-67, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731240

ABSTRACT

All herpesviruses encode a homolog of glycoprotein M (gM), which appears to function in virion morphogenesis. Despite its conservation, gM is inessential for the lytic replication of alphaherpesviruses. In order to address the importance of gM in gammaherpesviruses, we disrupted it in the murine gammaherpesvirus 68 (MHV-68). The mutant virus completely failed to propagate in normally permissive fibroblasts. The defective genome was rescued by either homologous recombination to restore the wild-type gM in situ or the insertion of an ectopic, intergenic expression cassette encoding gM into the viral genome. Thus, gM was essential for the lytic replication of MHV-68.


Subject(s)
Gammaherpesvirinae/physiology , Glycoproteins/physiology , Viral Envelope Proteins/physiology , Virus Replication , Animals , Cell Line , Cricetinae , Genes, Essential , Genes, Viral , Mutagenesis, Insertional , Recombination, Genetic , Viral Plaque Assay
16.
J Virol ; 79(8): 5059-68, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15795291

ABSTRACT

Herpesviruses remain predominantly cell associated within their hosts, implying that they spread between cells by a mechanism distinct from free virion release. We previously identified the efficient release of murine gammaherpesvirus 68 (MHV-68) virions as a function of the viral gp150 protein. Here we show that the MHV-68 ORF27 gene product, gp48, contributes to the direct spread of viruses from lytically infected to uninfected cells. Monoclonal antibodies to gp48 identified it on infected cell surfaces and in virions. gp48-deficient viruses showed no obvious deficit in virion cell binding, single-cycle replication, or virion release but had reduced lytic propagation between cells. After intranasal infection of mice, ORF27-deficient viruses were impaired predominantly in lytic replication in the lungs. There was a small deficit in latency establishment, but long-term latency appeared normal. Since ORF27 has homologs in both Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, it is likely part of a conserved mechanism employed by gammaherpesviruses to disseminate lytically in their hosts.


Subject(s)
Gammaherpesvirinae/genetics , Open Reading Frames/genetics , Amino Acid Sequence , Animals , Female , Gammaherpesvirinae/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid , Virus Replication/genetics
17.
Traffic ; 6(9): 780-93, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16101681

ABSTRACT

The murine gamma-herpesvirus-68 (MHV-68) ORF27 encodes gp48, a type 2 transmembrane glycoprotein that contributes to intercellular viral spread. Gp48 is expressed on the surface of infected cells but is retained intracellularly after transfection. In this study, we show that the multimembrane spanning ORF58 gene product is both necessary and sufficient for gp48 to reach the cell surface. ORF58-deficient MHV-68 expressed ORF27 in normal amounts, but retained it in the endoplasmic reticulum (ER). Transfected ORF27 also remained in ER, whereas green fluorescent protein-tagged ORF58 localized to the ER and trans-Golgi network. When ORF27 and ORF58 were co-transfected, they formed a protein complex and reached the cell surface. Surprisingly, ORF58 rather than ORF27 mediated cell binding via a small extracellular loop. The heavily glycosylated ORF27 component of the complex may, therefore, act mainly to protect this loop against antibody. The interdependent transport of ORF27 and ORF58 transport ensures that such protection is always present.


Subject(s)
Glycoproteins/physiology , Intracellular Membranes/metabolism , Rhadinovirus/genetics , Viral Proteins/physiology , Virus Replication , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Cell Line , Cricetinae , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Fibroblasts/ultrastructure , Fibroblasts/virology , Fluorescent Antibody Technique, Indirect , Glycoproteins/metabolism , Humans , Immunoblotting , Immunohistochemistry , Intracellular Membranes/ultrastructure , Kidney/cytology , Kidney/embryology , Mice , Microscopy, Confocal , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Precipitin Tests , Protein Structure, Tertiary , Protein Transport , Rhadinovirus/physiology , Viral Proteins/chemistry
18.
J Virol ; 78(23): 13370-5, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542690

ABSTRACT

Murine gammaherpesvirus 68 (MHV-68) glycoprotein B (gB) was identified in purified virions by immunoblotting, immunoprecipitation, and immunoelectron microscopy. It was synthesized as a 120-kDa precursor in infected cells and cleaved into 65-kDa and 55-kDa disulfide-linked subunits close to the time of virion release. The N-linked glycans on the cleaved, virion gB remained partially endoglycosidase H sensitive. The processing of MHV-68 gB therefore appears similar to that of Kaposi's sarcoma-associated herpesvirus gB and human cytomegalovirus gB.


Subject(s)
Viral Envelope Proteins/chemistry , Amino Acid Sequence , Animals , Disulfides/chemistry , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Rhadinovirus/chemistry , Rhadinovirus/physiology , Viral Envelope Proteins/physiology , Virion/physiology
SELECTION OF CITATIONS
SEARCH DETAIL