Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 100(1): 259-68, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23994498

ABSTRACT

We have examined the role that hydroxymethylglutaryl coenzyme A reductase (HMGCR) plays during respiratory syncytial virus (RSV) maturation. Imaging analysis indicated that virus-induced changes in F-actin structure correlated with the formation of virus filaments, and that these virus filaments played a direct role in virus cell-to-cell transmission. Treatment with cytochalasin D (CYD) prevented virus filament formation and virus transmission, but this could be reversed by removal of CYD. This observation, together with the presence of F-actin within the virus filaments suggested that newly polymerised F-actin was required for virus transmission. The virus-induced change in F-actin was inhibited by the HMGCR inhibitor lovastatin, and this correlated with the inhibition of both virus filament formation and the incorporation of F-actin in these virus structures. Furthermore, this inhibitory effect on virus filament formation correlated with a significant reduction in RSV transmission. Collectively these data suggested that HMGCR-mediated changes in F-actin structure play an important role in the inter-cellular transmission of mature RSV particles. These data also highlighted the interplay between cellular metabolism and RSV transmission, and demonstrate that this interaction can be targeted using anti-virus strategies.


Subject(s)
Actins/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Respiratory Syncytial Virus Infections/enzymology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Antiviral Agents/pharmacology , Cytochalasin D/pharmacology , Hep G2 Cells , Humans , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/drug effects
2.
J Virol ; 86(15): 7818-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22593157

ABSTRACT

The maturation of the hepatitis C virus (HCV) core protein requires proteolytic processing by two host proteases: signal peptidase (SP) and the intramembrane-cleaving protease signal peptide peptidase (SPP). Previous work on HCV genotype 1a (GT1a) and GT2a has identified crucial residues required for efficient signal peptide processing by SPP, which in turn has an effect on the production of infectious virus particles. Here we demonstrate that the JFH1 GT2a core-E1 signal peptide can be adapted to the GT3a sequence without affecting the production of infectious HCV. Through mutagenesis studies, we identified crucial residues required for core-E1 signal peptide processing, including a GT3a sequence-specific histidine (His) at position 187. In addition, the stable knockdown of intracellular SPP levels in HuH-7 cells significantly affects HCV virus titers, further demonstrating the requirement for SPP for the maturation of core and the production of infectious HCV particles. Finally, our nuclear magnetic resonance (NMR) structural analysis of a synthetic HCV JFH1 GT2a core-E1 signal peptide provides an essential structural template for a further understanding of core processing as well as the first model for an SPP substrate within its membrane environment. Our findings give deeper insights into the mechanisms of intramembrane-cleaving proteases and the impact on viral infections.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Hepacivirus/metabolism , Hepatitis C/metabolism , Protein Sorting Signals , Viral Core Proteins/metabolism , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/genetics , HEK293 Cells , Hepacivirus/chemistry , Hepacivirus/genetics , Hepatitis C/genetics , Humans , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Viral Core Proteins/chemistry , Viral Core Proteins/genetics
3.
Mol Cell Proteomics ; 9(9): 1829-48, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20530633

ABSTRACT

In this study, we used imaging and proteomics to identify the presence of virus-associated cellular proteins that may play a role in respiratory syncytial virus (RSV) maturation. Fluorescence microscopy of virus-infected cells revealed the presence of virus-induced cytoplasmic inclusion bodies and mature virus particles, the latter appearing as virus filaments. In situ electron tomography suggested that the virus filaments were complex structures that were able to package multiple copies of the virus genome. The virus particles were purified, and the protein content was analyzed by one-dimensional nano-LC MS/MS. In addition to all the major virus structural proteins, 25 cellular proteins were also detected, including proteins associated with the cortical actin network, energy pathways, and heat shock proteins (HSP70, HSC70, and HSP90). Representative actin-associated proteins, HSC70, and HSP90 were selected for further biological validation. The presence of beta-actin, filamin-1, cofilin-1, HSC70, and HSP90 in the virus preparation was confirmed by immunoblotting using relevant antibodies. Immunofluorescence microscopy of infected cells stained with antibodies against relevant virus and cellular proteins confirmed the presence of these cellular proteins in the virus filaments and inclusion bodies. The relevance of HSP90 to virus infection was examined using the specific inhibitors 17-N-Allylamino-17-demethoxygeldanamycin. Although virus protein expression was largely unaffected by these drugs, we noted that the formation of virus particles was inhibited, and virus transmission was impaired, suggesting an important role for HSP90 in virus maturation. This study highlights the utility of proteomics in facilitating both our understanding of the role that cellular proteins play during RSV maturation and, by extrapolation, the identification of new potential targets for antiviral therapy.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , Respiratory Syncytial Viruses/physiology , Virion/physiology , Virus Assembly/physiology , Blotting, Western , Electrophoresis, Polyacrylamide Gel , HSP90 Heat-Shock Proteins/physiology , Immunoprecipitation , RNA, Small Interfering , Virion/chemistry
4.
Virology ; 386(1): 168-82, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19178924

ABSTRACT

We examined the structure of lipid-raft membranes in respiratory syncytial virus infected cells. Cholesterol depletion studies using methyl-beta-cyclodextrin suggested that membrane cholesterol was required for virus filament formation, but not inclusion bodies. In addition, virus filament formation coincided with elevated 3-hydroxy-3-methylglutaryl-coenzyme A reductase expression, suggesting an increase in requirement for endogenous cholesterol synthesis during virus assembly. Lipid raft membranes were examined by mass spectrometry, which suggested that virus infection induced subtle changes in the lipid composition of these membrane structures. This analysis revealed increased levels of raft-associated phosphatidylinositol (PI) and phosphorylated PI during RSV infection, which correlated with the appearance of phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol 3,4,5-triphosphate (PIP(3)) within virus inclusion bodies, and inhibiting the synthesis of PIP(3) impaired the formation of progeny virus. Collectively, our analysis suggests that RSV infection induces specific changes in the composition of raft-associated lipids, and that these changes play an important role in virus maturation.


Subject(s)
Lipid Metabolism , Lipids/analysis , Membrane Microdomains/chemistry , Respiratory Syncytial Viruses/physiology , Virus Assembly , Cell Line , Humans , Hydroxymethylglutaryl-CoA-Reductases, NADP-dependent/metabolism , Inclusion Bodies, Viral/chemistry , Mass Spectrometry
5.
Virology ; 369(2): 309-23, 2007 Dec 20.
Article in English | MEDLINE | ID: mdl-17825340

ABSTRACT

During respiratory syncytial virus (RSV) infection there is a close physical interaction between the filamentous actin (F-actin) and the virus, involving both inclusion bodies and the virus filaments. This interaction appears to occur relatively early in the replication cycle, and can be detected from 8 h post-infection. Furthermore, during virus assembly we obtained evidence for the participation of an F-actin-associated signalling pathway involving phosphatidyl-3-kinase (PI3K). Treatment with the PI3K inhibitor LY294002 prevented the formation of virus filaments, although no effect was observed either on virus protein expression, or on trafficking of the virus glycoproteins to the cell surface. Inhibition of the activity of Rac GTPase, a down-stream effector of PI3K, by treatment with the Rac-specific inhibitor NSC23766 gave similar results. These data suggest that an intimate interaction occurs between actin and RSV, and that actin-associated signalling pathway, involving PI3K and Rac GTPase, may play an important role during virus assembly.


Subject(s)
Actins/physiology , Respiratory Syncytial Virus, Human/physiology , Respiratory Syncytial Virus, Human/ultrastructure , Virus Assembly/physiology , Aminoquinolines/pharmacology , Cell Line , Chromones/pharmacology , Cytoskeleton/ultrastructure , Cytoskeleton/virology , Enzyme Inhibitors/pharmacology , Humans , Inclusion Bodies, Viral/physiology , Inclusion Bodies, Viral/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Pyrimidines/pharmacology , Signal Transduction , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/physiology
6.
Methods Mol Biol ; 379: 69-83, 2007.
Article in English | MEDLINE | ID: mdl-17502671

ABSTRACT

The respiratory syncytial virus fusion (F) protein is initially expressed as a single polypeptide chain (F0). The F0 subsequently undergoes posttranslational cleavage-by-cell protease activity to produce the F1 and F2 subunits. Each of the two subunits within the mature F protein is modified by the addition of N-linked glycans. The individual N-linked glycans on the F protein were selectively removed by using site-directed mutagenesis to mutate the individual glycan-acceptor sites. In this way the role of these individual glycans in targeting of the F protein to the cell surface, and on the ability of the F protein to induce membrane fusion, was examined.


Subject(s)
Membrane Fusion/physiology , Polysaccharides/metabolism , Protein Processing, Post-Translational/physiology , Protein Subunits/metabolism , Respiratory Syncytial Viruses/metabolism , Viral Fusion Proteins/metabolism , Animals , Chlorocebus aethiops , HeLa Cells , Humans , Protein Subunits/genetics , Respiratory Syncytial Viruses/genetics , Vero Cells , Viral Fusion Proteins/genetics
7.
Methods Mol Biol ; 379: 149-61, 2007.
Article in English | MEDLINE | ID: mdl-17502677

ABSTRACT

Sequences derived from the respiratory syncytial virus (RSV) fusion (F) protein were expressed in insect cells as recombinant glutathione-S-transferase (GST)-tagged proteins. The sequence covering the F2 subunit (GST-F2), and a truncated form of the F protein in which the transmembrane domain was removed (GST-F2/F1), were cloned into the baculovirus pAcSecG2T secretory vector. These virus sequences also had the endogenous virus signal sequence removed and replaced with a signal sequence derived from the baculovirus gp67 glycoprotein, which was present in pAcSecG2T. The recombinant RSV glycoproteins were successfully detected in expressing cells by immunofluorescence assay and in the tissue culture medium by western blot analysis. The secreted recombinant GST-F2/F1 protein was further analysed using glycosidases. Our results showed that the GST-F2/F1 protein were sensitive to peptide:N-glycosidase F (PNGase F) treatment, but not to Endoglycosidase H (EndoH) treatment. This indicates that the secreted recombinant proteins were modified by the addition of mature N-linked glycan chains.


Subject(s)
Respiratory Syncytial Viruses , Viral Fusion Proteins/metabolism , Animals , Baculoviridae/genetics , Cell Line , Gene Expression , Genetic Vectors/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Respiratory Syncytial Viruses/genetics , Spodoptera , Viral Fusion Proteins/biosynthesis , Viral Fusion Proteins/genetics
8.
Virology ; 350(2): 289-301, 2006 Jul 05.
Article in English | MEDLINE | ID: mdl-16513154

ABSTRACT

Glycan heterogeneity of the respiratory syncytial virus (RSV) fusion (F) protein was demonstrated by proteomics. The effect of maturation of the virus glycoproteins-associated glycans on virus infectivity was therefore examined using the alpha-mannosidase inhibitors deoxymannojirimycin (DMJ) and swainsonine (SW). In the presence of SW the N-linked glycans on the F protein appeared in a partially mature form, whereas in the presence of DMJ no maturation of the glycans was observed. Neither inhibitor had a significant effect on G protein processing or on the formation of progeny virus. Although the level of infectious virus and syncytia formation was not significantly affected by SW-treatment, DMJ-treatment correlated with a one hundred-fold reduction in virus infectivity. Our data suggest that glycan maturation of the RSV glycoproteins, in particular those on the F protein, is an important step in virus maturation and is required for virus infectivity.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycoproteins/metabolism , Polysaccharides/metabolism , Respiratory Syncytial Virus, Human/physiology , Viral Proteins/metabolism , alpha-Mannosidase/antagonists & inhibitors , Cell Fusion , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Glycoside Hydrolases , Humans , Microscopy, Electron, Scanning , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/pathogenicity , Viral Proteins/genetics , Viral Proteins/isolation & purification
9.
Virology ; 338(1): 69-80, 2005 Jul 20.
Article in English | MEDLINE | ID: mdl-15936795

ABSTRACT

In this report, the interaction between respiratory syncytial virus (RSV) and heat shock protein 70 (HSP70) was examined. Although no significant increase in total HSP70 protein levels was observed during virus infection, analysis of the HSP70 content in lipid-raft membranes from mock- and virus-infected cells revealed an increase in the levels of raft-associated HSP70 during virus infection. Fluorescence microscopy demonstrated that this transport of HSP70 into lipid-raft membranes correlated with the appearance of HSP70 within virus-induced inclusion bodies. Furthermore, co-localisation of HSP70 with the virus N protein and the raft lipid GM1 was observed within these structures. Immunoprecipitation experiments demonstrated the ability of HSP70 to interact with the virus polymerase complex in lipid-rafts in an ATP-dependent manner. Collectively, these data suggest that RSV may induce cellular changes which allow the recruitment of specific host-cell factors, via lipid-raft membranes, to the polymerase complex.


Subject(s)
HSP70 Heat-Shock Proteins/physiology , Membrane Microdomains/physiology , Membrane Microdomains/virology , Respiratory Syncytial Viruses/physiology , Respiratory Syncytial Viruses/pathogenicity , Viral Proteins/physiology , Amino Acid Sequence , Cell Line , HSP70 Heat-Shock Proteins/genetics , Humans , Inclusion Bodies/physiology , Inclusion Bodies/virology , Microscopy, Fluorescence , Molecular Sequence Data , Multiprotein Complexes , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Respiratory Syncytial Viruses/genetics , Viral Proteins/genetics , Virus Replication
10.
Virology ; 330(1): 147-57, 2004 Dec 05.
Article in English | MEDLINE | ID: mdl-15527841

ABSTRACT

The interaction between the respiratory syncytial virus (RSV) polymerase complex and lipid rafts was examined in HEp2 cells. Lipid-raft membranes were prepared from virus-infected cells and their protein content was analysed by Western blotting and mass spectrometry. This analysis revealed the presence of the N, P, L, M2-1 and M proteins. However, these proteins appeared to differ from one another in their association with these structures, with the M2-1 protein showing a greater partitioning into raft membranes compared to that of the N, P or M proteins. Determination of the polymerase activity profile of the gradient fractions revealed that 95% of the detectable viral enzyme activity was associated with lipid-raft membranes. Furthermore, analysis of virus-infected cells by confocal microscopy suggested an association between these proteins and the raft-lipid, GM1. Together, these results provide evidence that the RSV polymerase complex is able to associate with lipid rafts in virus-infected cells.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Membrane Microdomains/virology , Respiratory Syncytial Virus, Human/enzymology , Amino Acid Sequence , Cell Line, Tumor , DNA-Directed RNA Polymerases/chemistry , Humans , Mass Spectrometry , Molecular Sequence Data , Peptide Fragments/chemistry
11.
Virology ; 327(2): 175-85, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15351205

ABSTRACT

The assembly of respiratory syncytial virus (RSV) in lipid-rafts was examined in Hep2 cells. Confocal and electron microscopy showed that during RSV assembly, the cellular distribution of the complement regulatory proteins, decay accelerating factor (CD55) and CD59, changes and high levels of these cellular proteins are incorporated into mature virus filaments. The detergent-solubility properties of CD55, CD59, and the RSV fusion (F) protein were found to be consistent with each protein being located predominantly within lipid-raft structures. The levels of these proteins in cell-released virus were examined by immunoelectronmicroscopy and found to account for between 5% and 15% of the virus attachment (G) glycoprotein levels. Collectively, our findings suggest that an intimate association exists between RSV and lipid-raft membranes and that significant levels of these host-derived raft proteins, such as those regulating complement activation, are subsequently incorporated into the envelope of mature virus particles.


Subject(s)
CD55 Antigens/metabolism , CD59 Antigens/metabolism , Membrane Microdomains/metabolism , Respiratory Syncytial Virus, Human/pathogenicity , Virus Assembly , Animals , Cell Line, Tumor , Chlorocebus aethiops , Humans , Membrane Microdomains/chemistry , Microscopy, Confocal , Microscopy, Electron , Respiratory Syncytial Virus, Human/metabolism , Vero Cells , Virion/metabolism
12.
J Gen Virol ; 85(Pt 5): 1153-1165, 2004 May.
Article in English | MEDLINE | ID: mdl-15105532

ABSTRACT

The cellular distribution of the small hydrophobic (SH) protein in respiratory syncytial virus (RSV)-infected cells was examined. Although the SH protein was distributed throughout the cytoplasm, it appeared to accumulate in the Golgi complex within membrane structures that were enriched in the raft lipid, GM1. The ability of the SH protein to interact with lipid-raft membranes was further confirmed by examining its detergent-solubility properties in Triton X-100 at 4 degrees C. This analysis showed that a large proportion of the SH protein exhibited detergent-solubility characteristics that were consistent with an association with lipid-raft membranes. Analysis of virus-infected cells by immuno-transmission electron microscopy revealed SH protein clusters on the cell surface, but only very low levels of the protein appeared to be associated with mature virus filaments and inclusion bodies. These data suggest that during virus infection, the compartments in the secretory pathway, such as the endoplasmic reticulum (ER) and Golgi complex, are major sites of accumulation of the SH protein. Furthermore, although a significant amount of this protein interacts with lipid-raft membranes within the Golgi complex, its presence within mature virus filaments is minimal.


Subject(s)
Golgi Apparatus/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , G(M1) Ganglioside/metabolism , Microscopy, Fluorescence , Vero Cells , Viral Envelope Proteins/analysis , Viral Proteins/analysis
13.
Virology ; 306(2): 254-67, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12642099

ABSTRACT

Field emission scanning electron microscopy (FE SEM) was used to visualize the distribution of virus-associated components, the virus-attachment (G) protein, and the host-cell-derived lipid, GM1, in respiratory syncytial virus (RSV) filaments. RSV-infected cells were labeled in situ with a G protein antibody (MAb30) whose presence was detected using a second antibody conjugated to colloidal gold. No bound MAb30 was detected in mock-infected cells, whereas significant quantities bound to viral filaments revealing G protein clusters throughout the filaments. GM1 was detected using cholera toxin B subunit conjugated to colloidal gold. Mock-infected cells revealed numerous GM1 clusters on the cell surface. In RSV-infected cells, these gold clusters were detected on the filaments in low, but significant, amounts, indicating the incorporation of GM1 within the viral envelope. This report describes the first use of FE SEM to map the distribution of specific structural components within the envelope of a Paramyxovirus.


Subject(s)
G(M1) Ganglioside/metabolism , Respiratory Syncytial Viruses/metabolism , Viral Proteins/metabolism , Animals , Antibodies, Monoclonal , Antibodies, Viral , Cell Membrane/metabolism , Cell Membrane/virology , Chlorocebus aethiops , Microscopy, Electron, Scanning/methods , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/ultrastructure , Vero Cells
14.
J Gen Virol ; 83(Pt 8): 1841-1850, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12124448

ABSTRACT

We have previously shown that respiratory syncytial virus (RSV) assembly occurs within regions of the host-cell surface membrane that are enriched in the protein caveolin-1 (cav-1). In this report, we have employed immunofluorescence microscopy to further examine the RSV assembly process. Our results show that RSV matures at regions of the cell surface that, in addition to cav-1, are enriched in the lipid-raft ganglioside GM1. Furthermore, a comparison of mock-infected and RSV-infected cells by confocal microscopy revealed a significant change in the cellular distribution of phosphocaveolin-1 (pcav-1). In mock-infected cells, pcav-1 was located at regions of the cell that interact with the extracellular matrix, termed focal adhesions (FA). In contrast, RSV-infected cells showed both a decrease in the levels of pcav-1 associated with FA and the appearance of pcav-1-containing cytoplasmic vesicles, the latter being absent in mock-infected cells. These cytoplasmic vesicles were clearly visible between 9 and 18 h post-infection and coincided with the formation of RSV filaments, although we did not observe a direct association of pcav-1 with mature virus. In addition, we noted a strong colocalization between pcav-1 and growth hormone receptor binding protein-7 (Grb7), within these cytoplasmic vesicles, which was not observed in mock-infected cells. Collectively, these findings show that the RSV assembly process occurs within specialized lipid-raft structures on the host-cell plasma membrane, induces the cellular redistribution of pcav-1 and results in the formation of cytoplasmic vesicles that contain both pcav-1 and Grb7.


Subject(s)
Caveolins/metabolism , G(M1) Ganglioside/metabolism , Membrane Microdomains/virology , Respiratory Syncytial Virus, Human/growth & development , Respiratory Syncytial Virus, Human/metabolism , Virus Assembly , Animals , Caveolin 1 , Chlorocebus aethiops , Cytoplasmic Vesicles/metabolism , GRB7 Adaptor Protein , Humans , Membrane Microdomains/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Phosphorylation , Proteins/metabolism , Tyrosine/metabolism , Vero Cells
15.
J Gen Virol ; 83(Pt 3): 611-621, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11842256

ABSTRACT

We have employed immunofluorescence microscopy and transmission electron microscopy to examine the assembly and maturation of respiratory syncytial virus (RSV) in the Vero cell line C1008. RSV matures at the apical cell surface in a filamentous form that extends from the plasma membrane. We observed that inclusion bodies containing viral ribonucleoprotein (RNP) cores predominantly appeared immediately below the plasma membrane, from where RSV filaments form during maturation at the cell surface. A comparison of mock-infected and RSV-infected cells by confocal microscopy revealed a significant change in the pattern of caveolin-1 (cav-1) fluorescence staining. Analysis by immuno-electron microscopy showed that RSV filaments formed in close proximity to cav-1 clusters at the cell surface membrane. In addition, immuno-electron microscopy showed that cav-1 was closely associated with early budding RSV. Further analysis by confocal microscopy showed that cav-1 was subsequently incorporated into the envelope of RSV filaments maturing on the host cell membrane, but was not associated with other virus structures such as the viral RNPs. Although cav-1 was incorporated into the mature virus, it was localized in clusters rather than being uniformly distributed along the length of the viral filaments. Furthermore, when RSV particles in the tissue culture medium from infected cells were examined by immuno-negative staining, the presence of cav-1 on the viral envelope was clearly demonstrated. Collectively, these findings show that cav-1 is incorporated into the envelope of mature RSV particles during egress.


Subject(s)
Caveolins/metabolism , Cell Membrane/metabolism , Cell Membrane/virology , Respiratory Syncytial Virus, Human/growth & development , Respiratory Syncytial Virus, Human/metabolism , Virus Assembly , Animals , Caveolin 1 , Cell Membrane/ultrastructure , Cell Polarity , Chlorocebus aethiops/virology , Fluorescent Antibody Technique , Immunohistochemistry , Inclusion Bodies, Viral/metabolism , Inclusion Bodies, Viral/ultrastructure , Microscopy, Electron , Microscopy, Immunoelectron , Protein Transport , Respiratory Syncytial Virus, Human/ultrastructure , Ribonucleoproteins/metabolism , Vero Cells , Viral Proteins/metabolism
16.
J Gen Virol ; 83(Pt 1): 61-66, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752701

ABSTRACT

Analysis of the respiratory syncytial virus (RSV) fusion (F) protein in RSV-infected Vero cells showed the presence of a single F1 subunit and at least two different forms of the F2 subunit, designated F2a (21 kDa) and F2b (16 kDa), which were collectively referred to as [F2](a/b). Enzymatic deglycosylation of [F2](a/b) produced a single 10 kDa product suggesting that [F2](a/b) arises from differences in the glycosylation pattern of F2a and F2b. The detection of [F2](a/b) was dependent upon the post-translational cleavage of the F protein by furin, since its appearance was prevented in RSV-infected Vero cells treated with the furin inhibitor dec-RVKR-cmk. Analysis by protein cross-linking revealed that the F1 subunit interacted with [F2](a/b), via disulphide bonding, to produce equivalent F protein trimers, which were expressed on the surface of infected cells. Collectively, these data show that multiple F protein species are expressed in RSV-infected cells.


Subject(s)
Respiratory Syncytial Virus, Human/metabolism , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/metabolism , Viral Proteins/metabolism , Amidohydrolases/metabolism , Animals , Chlorocebus aethiops , Fucose/metabolism , Glucosamine/metabolism , Glycosylation , HeLa Cells , Humans , Mannose/metabolism , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Tumor Cells, Cultured , Vero Cells , Viral Envelope Proteins/biosynthesis , Viral Fusion Proteins/biosynthesis
17.
J Gen Virol ; 82(Pt 6): 1375-1386, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11369882

ABSTRACT

The intracellular cleavage of respiratory syncytial virus (RSV) fusion (F) protein by furin was examined. In RSV-infected LoVo cells, which express an inactive form of furin, and in RSV-infected Vero cells treated with the furin inhibitor decanoyl-Arg-Val-Lys-Arg-chloromethyl ketone (dec-RVKR-cmk), the F protein was expressed as a non-cleaved 73 kDa species. In both cases the F protein was initially expressed as an endoglycosidase H (Endo H)-sensitive precursor (F0(EHs)) which was modified approximately 40 min post-synthesis by the addition of complex carbohydrates to produce the Endo H-resistant form (F0(EHr)). The size and glycosylation state of F0(EHr) were identical to a transient intermediate form of non-cleaved F protein which was detected in RSV-infected Vero cells in the absence of inhibitor. Cell surface biotinylation and surface immunofluorescence staining showed that F0(EHr) was present on the surface of RSV-infected cells. RSV filaments have been shown to be the predominant form of the budding virus that is detected during virus replication. Analysis of the RSV-infected cells using scanning electron microscopy (SEM) showed that, in the presence of dec-RVKR-cmk, virus budding was impaired, producing fewer and much smaller viral filaments than in untreated cells. A comparison of immunofluorescence and SEM data showed that F0(EHr) was routed to the surface of virus-infected cells but not located in these smaller structures. Our findings suggest that activation of the F protein is required for the efficient formation of RSV filaments.


Subject(s)
Cell Membrane/metabolism , Protein Processing, Post-Translational , Respiratory Syncytial Viruses/metabolism , Subtilisins/metabolism , Viral Fusion Proteins/metabolism , Animals , Biotinylation , Cell Line , Cell Membrane/ultrastructure , Cell Membrane/virology , Chlorocebus aethiops , Fluorescent Antibody Technique , Furin , Glycosylation , Kinetics , Microscopy, Electron, Scanning , Molecular Weight , Mutation/genetics , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/ultrastructure , Serine Proteinase Inhibitors/pharmacology , Subtilisins/antagonists & inhibitors , Subtilisins/genetics , Vero Cells , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/ultrastructure , Virus Assembly/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...