Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
J Gen Virol ; 92(Pt 7): 1493-1499, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21471320

ABSTRACT

The prophylactic use of topical antiviral agents has recently been validated by the reduction in human immunodeficiency virus (HIV) type 1 infection incidence seen using tonofovir-containing microbicides. In order to develop a wide-spectrum microbicide to prevent infection with a wide range of sexually transmitted viruses, we have previously reported the development of HIV-neutralizing aptamers and here report the isolation and characterization of aptamers that neutralize herpes simplex virus type 2 (HSV-2). These aptamers bind the envelope glycoprotein (gD), are potent (IC(50) of 20-50 nM) and are able to block infection pathways dependent on both major entry receptors, Nectin1 and HVEM. Structural analysis and mutagenesis of these aptamers reveal a core specificity element that could provide the basis for pharmaceutical development. As HSV-2 is a major risk factor for the acquisition of HIV-1, a microbicide capable of preventing HSV-2 infection would not only reduce the morbidity associated with HSV-2, but also that derived from HIV-1.


Subject(s)
Antiviral Agents/pharmacology , Aptamers, Nucleotide/pharmacology , Herpes Simplex/virology , Herpesvirus 2, Human/drug effects , Animals , Antiviral Agents/chemistry , Aptamers, Nucleotide/chemistry , Base Sequence , Cell Adhesion Molecules/metabolism , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/physiology , Humans , Molecular Sequence Data , Nectins
2.
J Virol ; 81(19): 10742-57, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17652377

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.


Subject(s)
Herpesvirus 1, Suid/physiology , Neurons/virology , Peripheral Nervous System/virology , Pseudorabies/virology , Viral Envelope Proteins/physiology , Virus Internalization , Animals , Axons/virology , Cell Adhesion Molecules/genetics , Cells, Cultured , Fibroblasts/virology , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Herpesvirus 1, Suid/genetics , Humans , Mice , Nectins , Peripheral Nervous System/cytology , Viral Envelope Proteins/genetics
3.
J Virol ; 75(22): 11185-95, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602758

ABSTRACT

One step in the process of herpes simplex virus (HSV) entry into cells is the binding of viral glycoprotein D (gD) to a cellular receptor. Human nectin-2 (also known as HveB and Prr2), a member of the immunoglobulin (Ig) superfamily, serves as a gD receptor for the entry of HSV-2, variant forms of HSV-1 that have amino acid substitutions at position 25 or 27 of gD (for example, HSV-1/Rid), and porcine pseudorabies virus (PRV). The gD binding region of nectin-2 is believed to be localized to the N-terminal variable-like (V) Ig domain. In order to identify specific amino acid sequences in nectin-2 that are important for HSV entry activity, chimeric molecules were constructed by exchange of sequences between human nectin-2 and its mouse homolog, mouse nectin-2, which mediates entry of PRV but not HSV-1 or HSV-2. The nectin-2 chimeric molecules were expressed in Chinese hamster ovary cells, which normally lack a gD receptor, and tested for cell surface expression and viral entry activity. As expected, chimeric molecules containing the V domain of human nectin-2 exhibited HSV entry activity. Replacement of either of two small regions in the V domain of mouse nectin-2 with amino acids from the equivalent positions in human nectin-2 (amino acids 75 to 81 or 89) transferred HSV-1/Rid entry activity to mouse nectin-2. The resulting chimeras also exhibited enhanced HSV-2 entry activity and gained the ability to mediate wild-type HSV-1 entry. Replacement of amino acid 89 of human nectin-2 with the corresponding mouse amino acid (M89F) eliminated HSV entry activity. These results identify two different amino acid sequences, predicted to lie adjacent to the C' and C" beta-strands of the V domain, that are critical for HSV entry activity. This region is homologous to the human immunodeficiency virus binding region of CD4 and to the poliovirus binding region of CD155.


Subject(s)
Cell Adhesion Molecules/chemistry , Simplexvirus/physiology , Amino Acid Sequence , Animals , Cell Adhesion Molecules/physiology , Humans , Mice , Molecular Sequence Data , Nectins , Receptors, Virus/chemistry , Receptors, Virus/physiology , Species Specificity , Structure-Activity Relationship
4.
Virology ; 287(2): 301-9, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11531408

ABSTRACT

Both human and murine forms of nectin-1 (HveC, Prr1) can serve as entry receptors for several neurotropic herpesviruses, including herpes simplex viruses 1 and 2 (HSV-1, HSV-2), porcine pseudorabies virus (PRV), and bovine herpesvirus 1. HSV-1, HSV-2, and PRV can cause lethal neurological disease in mice whether inoculation is directly into the central nervous system or by peripheral routes. Expression of nectin-1 transcripts in cells of the adult mouse nervous system was assessed by in situ hybridization. Specific hybridization signals were detected in neurons in sensory, sympathetic, and parasympathetic ganglia of the peripheral nervous system. In addition, specific signals were observed in neurons of the ventral and dorsal horns of the spinal cord and of the brain stem, cerebellum, cerebral cortex, hippocampus, dentate gyrus, and olfactory bulb. These results show that the nectin-1 gene is widely transcribed in neurons in adult mouse. Nectin-1 is the only known receptor capable of mediating the entry of all three viruses, HSV-1, HSV-2, and PRV. Its pattern of expression in the nervous system suggests a key role in neurological disease caused by these viruses.


Subject(s)
Cell Adhesion Molecules/genetics , Neurons/metabolism , Animals , Cell Adhesion Molecules/metabolism , Central Nervous System/metabolism , Female , Ganglia/metabolism , Gene Expression , Herpesviridae/physiology , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred BALB C , Nectins , RNA/metabolism , Spinal Cord/metabolism , Transcription, Genetic
5.
Biochem J ; 359(Pt 1): 235-41, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11563988

ABSTRACT

3-O-Sulphates are the rarest substituent of heparan sulphate and are therefore ideally suited to the selective regulation of biological activities. Individual isoforms of heparan sulphate D-glucosaminyl 3-O-sulphotransferase (3-OST) exhibit sequence-specific action, which creates heparan sulphate structures with distinct biological functions. For example, 3-OST-1 preferentially generates binding sites for anti-thrombin, whereas 3-OST-3 isoforms create binding sites for the gD envelope protein of herpes simplex virus 1 (HSV-1), which enables viral entry. 3-OST enzymes comprise a presumptive sulphotransferase domain and a divergent N-terminal region. To localize determinants of sequence specificity, we conducted domain swaps between cDNA species. The N-terminal region of 3-OST-1 was fused with the sulphotransferase domain of 3-OST-3(A) to generate N1-ST3(A). Similarly, the N-terminal region of 3-OST-3(A) was fused to the sulphotransferase domain of 3-OST-1 to generate N3(A)-ST1. Wild-type and chimaeric enzymes were transiently expressed in COS-7 cells and extracts were analysed for selective generation of binding sites for anti-thrombin. 3-OST-1 was 270-fold more efficient at forming anti-thrombin-binding sites than 3-OST-3(A), indicating its significantly greater selectivity for substrates that can be 3-O-sulphated to yield such sites. N3(A)-ST1 was as active as 3-OST-1, whereas the activity of N1-ST3(A) was as low as that of 3-OST-3(A). Analysis of Chinese hamster ovary cell transfectants revealed that only 3-OST-3(A) and N1-ST3(A) generated gD-binding sites and conveyed susceptibility to infection by HSV-1. Thus sequence-specific properties of 3-OSTs are defined by a self-contained sulphotransferase domain and are not directly influenced by the divergent N-terminal region.


Subject(s)
Antithrombins/metabolism , Herpesvirus 1, Human/physiology , Sulfotransferases/metabolism , Amino Acid Sequence , Animals , Binding Sites , CHO Cells/enzymology , COS Cells/enzymology , Chimera , Cricetinae , DNA Primers/chemistry , DNA, Complementary/genetics , Female , Heparitin Sulfate , Herpes Simplex/genetics , Humans , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Plasmids , Polymerase Chain Reaction , Protein Isoforms , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sulfotransferases/genetics , Transfection
6.
Mol Cell ; 8(1): 2-4, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11511354

ABSTRACT

In this issue of Molecular Cell, Carfí et al. present the X-ray structure of the N-terminal domains of HveA, a TNF receptor family member, in complex with herpes simplex virus gD, providing a first step to understanding the herpesvirus mode of membrane fusion.


Subject(s)
Membrane Fusion , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Virus/chemistry , Viral Envelope Proteins/chemistry , Animals , Crystallography, X-Ray , Humans , Ligands , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Member 14 , Receptors, Virus/metabolism , Simplexvirus , Viral Envelope Proteins/metabolism
8.
Virology ; 285(2): 366-75, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11437670

ABSTRACT

Human nectin-1 (HveC, Prr1), a member of the immunoglobulin superfamily and a receptor for the entry of herpes simplex viruses 1 and 2 (HSV-1, HSV-2), pseudorabies virus (PRV), and bovine herpesvirus 1 (BHV-1), binds to viral gD. For HSV-1, HSV-2, and PRV, the gD-binding region of nectin-1 has been localized to the N-terminal V-like domain. To determine whether the two C-like domains of nectin-1 influenced gD binding and entry activity, genes encoding chimeric proteins were constructed. Portions of nectin-1 were replaced with homologous regions from nectin-2 (HveB, Prr2), a related protein with ability to mediate the entry of PRV, HSV-2, and Rid mutants of HSV-1, but not HSV-1 or BHV-1. Also, one or more domains of nectin-1 were fused to the two membrane-proximal Ig domains of CD4, a protein with no herpesvirus entry or gD-binding activity. The chimeric proteins were expressed in Chinese hamster ovary cells, which normally lack alphaherpesvirus entry receptors, and detected on the cell surface by one or more anti-nectin-1 monoclonal antibodies. One chimeric protein (nectin-1 amino acids 1-124 fused to CD4) failed to bind to soluble forms of HSV-1, HSV-2, PRV, and BHV-1 gD and, as expected, also failed to mediate entry of the viruses from which these gDs were derived. The other chimeric receptors bound all forms of gD. Some mediated the entry of all the viruses tested but others mediated entry of some but not all the viruses. We conclude that binding of gD to the nectin-1 V domain is not sufficient for entry activity, that there are structural requirements for entry activity independent of gD binding, and that these requirements are different for the several alphaherpesviruses that can use nectin-1 as a receptor.


Subject(s)
Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cattle , Cell Adhesion Molecules/genetics , Cell Membrane/metabolism , Cricetinae , Gene Expression , Herpesvirus 1, Bovine/metabolism , Herpesvirus 1, Bovine/physiology , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Suid/metabolism , Herpesvirus 1, Suid/physiology , Herpesvirus 2, Human/metabolism , Herpesvirus 2, Human/physiology , Humans , Immunoglobulins/genetics , Molecular Sequence Data , Nectins , Plasmids , Protein Conformation , Receptors, Virus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
Gene ; 265(1-2): 185-94, 2001 Mar 07.
Article in English | MEDLINE | ID: mdl-11255021

ABSTRACT

The Tage4 gene (Tumor-Associated Glycoprotein E4) is a member of the immunoglobulin superfamily overexpressed in rat colon tumors and Min mouse intestinal adenomas. The Tage4 cDNA presents approximately 60% identity with the human CD155, a member of the immunoglobulin superfamily coding for a transmembrane protein capable of serving as an entry receptor for poliovirus, porcine pseudorabies virus and bovine herpesvirus 1. We determined the structure of the Tage4 gene. This gene covers approximately 15 kb and is composed of eight exons and seven introns. We also isolated approximately 2 kb of the 5' flanking region of the Tage4 gene and demonstrated the existence of closely clustered transcription start sites. No splicing variant was identified by RT-PCR indicating that the Tage4 gene is transcribed as a unique mRNA. Finally, the protein encoded by the Tage4 gene was tested for ability to mediate entry of several viruses. These structural and functional features of the rat Tage4 gene were compared to those of the human CD155 gene. The results indicated that the Tage4 gene is probably orthologous to the gene for CD155.


Subject(s)
Genes/genetics , Glycoproteins/genetics , Herpesviridae/metabolism , Membrane Proteins , 3' Untranslated Regions/genetics , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cricetinae , DNA/chemistry , DNA/genetics , Exons , Glycoproteins/metabolism , Herpesviridae/genetics , Humans , Introns , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Rats , Receptors, Virus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
10.
Virus Res ; 74(1-2): 39-45, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11226572

ABSTRACT

Two cell surface proteins (nectin-1/HveC and nectin-2/HveB) shown previously to serve as receptors for the entry of herpes simplex virus 1 (HSV-1) wild-type and/or mutant strains were found to serve also as receptors for HSV-1-induced cell fusion. Transfection with genomic DNA from a syncytial HSV-1 strain encoding wild-type gD resulted in fusion of Chinese hamster ovary (CHO) cells expressing nectin-1 but not of cells expressing nectin-2. In contrast, transfection with DNA from a related HSV-1 strain encoding the mutant Rid1 form of gD resulted in fusion of CHO cells expressing either receptor but not of control cells. These results are consistent with the ability of each receptor to mediate entry of viruses expressing wild-type or Rid1 gD and with results obtained previously with HVEM (HveA), a third HSV-l entry receptor. Undersulfation of GAGs in receptor-expressing cell lines predictably reduced susceptibility to HSV-l infection. In contrast, susceptibility to cell fusion mediated by HVEM or nectin-1 was not reduced. Undersulfation of GAGs partially inhibited cell fusion mediated by nectin-2. We conclude that HSV-1-induced cell fusion requires a gD-binding entry receptor, that ability of an HSV-1 strain to use HVEM, nectin-2 or nectin-1 for cell fusion depends on the allele of gD expressed and that GAGs may influence cell fusion, dependent on the gD-binding receptor used, but are less important for cell fusion mediated by HVEM, nectin-2 or nectin-l than for viral entry.


Subject(s)
Cell Adhesion Molecules/metabolism , Membrane Fusion , Receptors, Virus/metabolism , Simplexvirus/metabolism , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/metabolism , Animals , CHO Cells , Cell Adhesion Molecules/genetics , Cricetinae , DNA, Viral/drug effects , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Mutation , Nectins , Receptors, Virus/genetics , Simplexvirus/genetics , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/metabolism , Transfection/methods , Viral Envelope Proteins/genetics , Viral Fusion Proteins/chemistry
11.
Virology ; 279(1): 313-24, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11145912

ABSTRACT

To characterize cellular factors required for herpes simplex virus type 1 (HSV-1)-induced cell fusion, we used an efficient and quantitative assay relying on expression of HSV-1 glycoproteins in transfected cells. We showed the following: (1) Cell fusion depended not only on expression of four viral glycoproteins (gB, gD, and gH-gL), as previously shown, but also on expression of cell surface entry receptors specific for gD. (2) Cell fusion required expression of all four glycoproteins in the same cell. (3) Heparan sulfate was not required for cell fusion. (4) Coexpression of receptor with the four glycoproteins in the same cell reduced fusion activity, indicating that interaction of gD and receptor can limit polykaryocyte formation. Overall, the viral and cellular determinants of HSV-1-induced cell fusion are similar to those for viral entry, except that HSV-1 entry is significantly enhanced by binding of virus to cell surface heparan sulfate.


Subject(s)
Herpes Simplex/virology , Membrane Fusion , Receptors, Virus/metabolism , Simplexvirus/physiology , Viral Envelope Proteins/metabolism , Animals , CHO Cells , Cell Fusion , Cricetinae , Heparitin Sulfate/metabolism
12.
J Virol ; 74(24): 11773-81, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090177

ABSTRACT

A cDNA encoding the murine homolog of human nectin-1alpha (also known as poliovirus receptor-related protein 1 [Prr1] and herpesvirus entry protein C [HveC]) was isolated. The protein encoded by this cDNA proved to be 95% identical in sequence to the human protein and to have similar herpesvirus entry activity. Upon expression of the murine cDNA in hamster cells resistant to alphaherpesvirus entry, the cells became susceptible to the entry of herpes simplex virus types 1 and 2 (HSV-1 and -2), pseudorabies virus, and bovine herpesvirus 1. HSV envelope glycoprotein D (gD), a viral ligand for human nectin-1alpha, is also a ligand for the murine homolog based on evidence that (i) a soluble hybrid protein composed in part of the murine nectin-1 ectodomain bound specifically to purified soluble forms of HSV-1 and HSV-2 gD as demonstrated by enzyme-linked immunosorbent assay, (ii) a soluble hybrid of HSV-1 gD bound to hamster cells expressing murine nectin-1alpha but not to control cells, and (iii) cells expressing both murine nectin-1alpha and one of the alphaherpesvirus gDs were resistant to entry of HSV-1, indicative of interference with entry resulting from interactions of cell-associated gD with the entry receptor. Northern blot analysis revealed that nectin-1 is expressed in most of the mouse tissues examined and at high levels in the brain, skin, and kidneys. Immunocytochemical localization demonstrated the presence of nectin-1 in epithelial cells of the mouse vagina and also in neuronal cells of the central nervous system, suggesting an expression pattern relevant to both infection at a portal of entry and spread of infection to the brain.


Subject(s)
Alphaherpesvirinae/physiology , Cell Adhesion Molecules/physiology , Receptors, Virus , Viral Envelope Proteins/physiology , Virus Replication , Amino Acid Sequence , Animals , Cattle , Cricetinae , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , Nectins , Sequence Alignment , Sequence Analysis
14.
J Biol Chem ; 275(36): 27733-40, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10864928

ABSTRACT

Heparan sulfate formation occurs by the copolymerization of glucuronic acid (GlcA) and N-acetylglucosamine (GlcNAc) residues. Recent studies have shown that these reactions are catalyzed by a copolymerase encoded by EXT1 and EXT2, members of the exostosin family of putative tumor suppressors linked to hereditary multiple exostoses. Previously, we identified a collection of Chinese hamster ovary cell mutants (pgsD) that failed to make heparan sulfate (Lidholt, K., Weinke, J. L., Kiser, C. S., Lugemwa, F. N., Bame, K. J., Cheifetz, S., Massagué, J., Lindahl, U., and Esko, J. D. (1992) Proc. Natl. Acad. Sci. U. S. A. 89, 2267-2271). Here, we show that pgsD mutants contain mutations that either alter GlcA transferase activity selectively or that affect both GlcNAc and GlcA transferase activities. Expression of EXT1 corrects the deficiencies in the mutants, whereas EXT2 and the related EXT-like cDNAs do not. Analysis of the EXT1 mutant alleles revealed clustered missense mutations in a domain that included a (D/E)X(D/E) motif thought to bind the nucleotide sugar from studies of other transferases. These findings provide insight into the location of the GlcA transferase subdomain of the enzyme and indicate that loss of the GlcA transferase domain may be sufficient to cause hereditary multiple exostoses.


Subject(s)
Glucuronosyltransferase/metabolism , Heparitin Sulfate/genetics , Mutation, Missense , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Acetylglucosamine/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Exostoses, Multiple Hereditary , Humans , Kinetics , Mice , Molecular Sequence Data , N-Acetylglucosaminyltransferases/genetics , Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
15.
Virology ; 268(1): 147-58, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10683337

ABSTRACT

Several human and animal alphaherpesviruses can enter cells via human herpesvirus entry mediator C (HveC), a receptor for viral glycoprotein D (gD). In previous studies with cells expressing unknown entry mediators, cellular expression of alphaherpesvirus gD was shown to inhibit entry of the homologous virus and sometimes also of heterologous alphaherpesviruses. To investigate the mechanism of gD-mediated interference and the basis for cross-interference among alphaherpesviruses, HveC was expressed in cells as the sole entry mediator, in the presence or absence of one of the gDs encoded by herpes simplex virus type 1, pseudorabies virus, or bovine herpesvirus type 1. Cells expressing HveC alone were highly susceptible to entry of all three viruses, whereas cells coexpressing HveC and any one of the gDs were at least partially resistant to infection by each virus. Coexpression of gD with HveC did not cause reduced levels of cell-surface HveC but the HveC had reduced ability to bind to exogenous gD. Coimmunoprecipitation experiments revealed that HveC was complexed with gD in lysates of cells expressing both. Thus, cellular expression of gD can interfere with alphaherpesvirus entry by blocking ligand-binding sites of the gD receptor(s) used for entry and cross-interference can occur because different forms of alphaherpesvirus gD can compete for shared entry receptors.


Subject(s)
Alphaherpesvirinae/genetics , Alphaherpesvirinae/physiology , Receptors, Tumor Necrosis Factor , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Animals , Blotting, Western , CHO Cells , Cricetinae , Flow Cytometry , Fluorescent Antibody Technique , Herpesviridae Infections/virology , Humans , Plasmids/genetics , Precipitin Tests , Receptors, Tumor Necrosis Factor, Member 14 , Transfection , Viral Envelope Proteins/genetics
16.
Cell ; 99(1): 13-22, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10520990

ABSTRACT

Herpes simplex virus type 1 (HSV-1) binds to cells through interactions of viral glycoproteins gB and gC with heparan sulfate chains on cell surface proteoglycans. This binding is not sufficient for viral entry, which requires fusion between the viral envelope and cell membrane. Here, we show that heparan sulfate modified by a subset of the multiple D-glucosaminyl 3-O-sulfotransferase isoforms provides sites for the binding of a third viral glycoprotein, gD, and for initiation of HSV-1 entry. We conclude that susceptibility of cells to HSV-1 entry depends on (1) presence of heparan sulfate chains to which virus can bind and (2) 3-O-sulfation of specific glucosamine residues in heparan sulfate to generate gD-binding sites or the expression of other previously identified gD-binding receptors.


Subject(s)
Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Herpes Simplex/physiopathology , Herpesvirus 1, Human/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Substitution , Animals , CHO Cells/chemistry , CHO Cells/metabolism , CHO Cells/virology , Cloning, Molecular , Cricetinae , Disease Susceptibility , Mice , Molecular Sequence Data , Plasmids , Protein Binding , Sequence Homology, Amino Acid , Transfection
17.
J Virol ; 73(10): 8127-37, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10482562

ABSTRACT

The human herpesvirus entry mediator C (HveC/PRR1) is a member of the immunoglobulin family used as a cellular receptor by the alphaherpesviruses herpes simplex virus (HSV), pseudorabies virus, and bovine herpesvirus type 1. We previously demonstrated direct binding of the purified HveC ectodomain to purified HSV type 1 (HSV-1) and HSV-2 glycoprotein D (gD). Here, using a baculovirus expression system, we constructed and purified truncated forms of the receptor containing one [HveC(143t)], two [HveC(245t)], or all three immunoglobulin-like domains [HveC(346t)] of the extracellular region. All three constructs were equally able to compete with HveC(346t) for gD binding. The variable domain bound to virions and blocked HSV infection as well as HveC(346t). Thus, all of the binding to the receptor occurs within the first immunoglobulin-like domain, or V-domain, of HveC. These data confirm and extend those of Cocchi et al. (F. Cocchi, M. Lopez, L. Menotti, M. Aoubala, P. Dubreuil, and G. Campadelli-Fiume, Proc. Natl. Acad. Sci. USA 95:15700, 1998). Using biosensor analysis, we measured the affinity of binding of gD from HSV strains KOS and rid1 to two forms of HveC. Soluble gDs from the KOS strain of HSV-1 had the same affinity for HveC(346t) and HveC(143t). The mutant gD(rid1t) had an increased affinity for HveC(346t) and HveC(143t) due to a faster rate of complex formation. Interestingly, we found that HveC(346t) was a tetramer in solution, whereas HveC(143t) and HveC(245t) formed dimers, suggesting a role for the third immunoglobulin-like domain of HveC in oligomerization. In addition, the stoichiometry between gD and HveC appeared to be influenced by the level of HveC oligomerization.


Subject(s)
Receptors, Tumor Necrosis Factor , Receptors, Virus/metabolism , Simplexvirus/physiology , Viral Envelope Proteins/metabolism , Animals , Cattle , Cell Line , Dimerization , Humans , Immunoglobulins , Protein Binding , Receptors, Tumor Necrosis Factor, Member 14 , Receptors, Virus/chemistry , Virus Replication
18.
J Virol ; 73(7): 5681-7, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364318

ABSTRACT

The herpesvirus entry mediator A (HveA) is a recently characterized member of the tumor necrosis factor receptor family that mediates the entry of most herpes simplex virus type 1 (HSV-1) strains into mammalian cells. Studies on the interaction of HSV-1 with HveA have shown that of all the viral proteins involved in uptake, only gD has been shown to bind directly to HveA, and this binding mediates viral entry into cells. In addition to gD binding to HveA, the latter has been shown to interact with proteins of tumor necrosis factor receptor-associated factor family, lymphotoxin-alpha (LT-alpha), and a membrane-associated protein referred to as LIGHT. To study the relationship between HveA, its natural ligands, and the viral proteins involved in HSV entry into cells, we have screened two phage-displayed combinatorial peptide libraries for peptide ligands of a recombinant form of HveA. Affinity selection experiments yielded two peptide ligands, BP-1 and BP-2, which could block the interaction between gD and HveA. Of the two peptides, only BP-2 inhibited HSV entry into CHO cells transfected with an HveA-expressing plasmid. When we analyzed these peptides for the ability to interfere with HveA binding to its natural ligand LT-alpha, we found that BP-1 inhibited the interaction of cellular LT-alpha with HveA. Thus, we have dissected the sites of interaction between the cell receptor, its natural ligand LT-alpha and gD, the virus-specific protein involved in HSV entry into cells.


Subject(s)
Carrier Proteins/metabolism , Lymphotoxin-alpha/metabolism , Peptides/metabolism , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Virus/antagonists & inhibitors , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Bacteriophages , Binding, Competitive , CHO Cells , Cell Line , Cricetinae , Ligands , Molecular Sequence Data , Peptides/chemical synthesis , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Member 14 , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spodoptera/cytology
19.
J Virol ; 73(5): 4493-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10196354

ABSTRACT

A mouse member of the immunoglobulin superfamily, originally designated the murine poliovirus receptor homolog (Mph), was found to be a receptor for the porcine alphaherpesvirus pseudorabies virus (PRV). This mouse protein, designated here murine herpesvirus entry protein B (mHveB), is most similar to one of three related human alphaherpesvirus receptors, the one designated HveB and also known as poliovirus receptor-related protein 2. Hamster cells resistant to PRV entry became susceptible upon expression of a cDNA encoding mHveB. Anti-mHveB antibody and a soluble protein composed of the mHveB ectodomain inhibited mHveB-dependent PRV entry. Expression of mHveB mRNA was detected in a variety of mouse cell lines, but anti-mHveB antibody inhibited PRV infection in only a subset of these cell lines, indicating that mHveB is the principal mediator of PRV entry into some mouse cell types but not others. Coexpression of mHveB with PRV gD, but not herpes simplex virus type 1 (HSV-1) gD, inhibited entry activity, suggesting that PRV gD may interact directly with mHveB as a ligand that can cause interference. By analogy with HSV-1, envelope-associated PRV gD probably also interacts directly with mHveB during viral entry.


Subject(s)
Herpesvirus 1, Human/metabolism , Herpesvirus 1, Suid/metabolism , Herpesvirus 2, Human/metabolism , Receptors, Tumor Necrosis Factor , Receptors, Virus/metabolism , 3T3 Cells , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Cell Line , Cricetinae , Gene Expression , Herpesvirus 1, Human/physiology , Herpesvirus 1, Suid/physiology , Herpesvirus 2, Human/physiology , Humans , Melanoma , Mice , Receptors, Tumor Necrosis Factor, Member 14 , Receptors, Virus/genetics , Tumor Cells, Cultured , Viral Envelope Proteins/metabolism
20.
Virology ; 251(2): 402-13, 1998 Nov 25.
Article in English | MEDLINE | ID: mdl-9837804

ABSTRACT

To characterize human herpesvirus 8 (HHV-8) gB, the open reading frame was PCR amplified from the HHV-8-infected cell line BCBL-1 and cloned into an expression vector. To facilitate detection of expressed HHV-8 gB, the cytoplasmic tail of the glycoprotein was tagged with the influenza hemagglutinin (HA) epitope. Expression of tagged HHV-8 gB (gB-HA), as well as the untagged form, was readily detected in CHO-K1 cells and several lymphoblastoid cell lines (LCLs). HHV-8 gB-HA was sensitive to endoglycosidase H treatment, and immunofluorescence revealed that HHV-8 gB-HA was detectable in the perinuclear region of CHO-K1 cells. These observations suggest that HHV-8 gB is not processed in the Golgi and localizes to the endoplasmic reticulum or nuclear membrane. Because both HHV-8 and EBV are gamma-herpesviruses, the ability of HHV-8 gB to interact with and functionally complement EBV gp110 was examined. HHV-8 gB-HA and EBV gp110 co-immunoprecipitated, indicating formation of hetero-oligomers. However, HHV-8 gB-HA and HHV-8 gB failed to restore the infectivity of gp110-negative EBV mutants. These findings indicate that although HHV-8 gB and EBV gp110 have similar patterns of intracellular localization and can interact, there is not sufficient functional homology to allow efficient complementation.


Subject(s)
Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , CHO Cells , Cricetinae , DNA, Viral/genetics , Glycosylation , Herpesvirus 8, Human/genetics , Humans , Molecular Sequence Data , Open Reading Frames , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...