Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Virology ; 591: 109985, 2024 03.
Article in English | MEDLINE | ID: mdl-38227992

ABSTRACT

Evidence for a stable interaction between the respiratory syncytial virus (RSV) F and G proteins on the surface of virus filaments was provided using antibody immunoprecipitation studies on purified RSV particles, and by the in situ analysis on the surface of RSV-infected cells using the proximity ligation assay. Imaging of the F and G protein distribution on virus filaments suggested that this protein complex was localised at the distal ends of the virus filaments, and suggested that this protein complex played a direct role in mediating efficient localised cell-to-cell virus transmission. G protein expression was required for efficient localised cell-to-cell transmission of RSV in cell monolayers which provided evidence that this protein complex mediates efficient multiple cycle infection. Collectively, these data provide evidence that F and G proteins form a complex on the surface of RSV particles, and that a role for this protein complex in promoting virus transmission is suggested.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Humans , Viral Fusion Proteins/genetics , Cytoskeleton , Protein Processing, Post-Translational , GTP-Binding Proteins/metabolism , Antibodies, Viral
2.
Subcell Biochem ; 106: 227-249, 2023.
Article in English | MEDLINE | ID: mdl-38159230

ABSTRACT

During respiratory syncytial virus (RSV) particle assembly, the mature RSV particles form as filamentous projections on the surface of RSV-infected cells. The RSV assembly process occurs at the / on the cell surface that is modified by a virus infection, involving a combination of several different host cell factors and cellular processes. This induces changes in the lipid composition and properties of these lipid microdomains, and the virus-induced activation of associated Rho GTPase signaling networks drives the remodeling of the underlying filamentous actin (F-actin) cytoskeleton network. The modified sites that form on the surface of the infected cells form the nexus point for RSV assembly, and in this review chapter, they are referred to as the RSV assembleome. This is to distinguish these unique membrane microdomains that are formed during virus infection from the corresponding membrane microdomains that are present at the cell surface prior to infection. In this article, an overview of the current understanding of the processes that drive the formation of the assembleome during RSV particle assembly is given.


Subject(s)
Respiratory Syncytial Virus, Human , Virus Diseases , Humans , Virus Assembly/physiology , Respiratory Syncytial Virus, Human/physiology , Cell Membrane/metabolism , Virus Diseases/metabolism , Lipids
3.
Virology ; 580: 28-40, 2023 03.
Article in English | MEDLINE | ID: mdl-36746062

ABSTRACT

The association of the SH protein with respiratory syncytial virus (RSV) particles was examined in HEp2 cells and human ciliated nasal epithelial cells. Imaging of infected cells demonstrated the presence of the SH protein in virus filaments, and analysis of purified RSV particles revealed a SH protein species whose size was consistent with the glycosylated SH protein. Although the SH protein was detected in virus filaments it was not required for virus filament formation. Analysis of RSV-infected ciliated cells also revealed that the SH protein was trafficked into the cilia, and this correlated with reduced cilia density on these cells. Reduced cilia loss was not observed on ciliated cells infected with a RSV isolate that failed to express the SH protein. These data provide direct evidence that the SH protein is trafficked into virus particles, and suggests that the SH protein may also promote cilia dysfunction on nasal epithelial cells.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus, Human/physiology , Epithelial Cells , Cytoskeleton , Virion
4.
J Virol ; 96(13): e0045522, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35727030

ABSTRACT

A human monoclonal antibody panel (PD4, PD5, PD7, SC23, and SC29) was isolated from the B cells of convalescent patients and used to examine the S protein in SARS-CoV-2-infected cells. While all five antibodies bound conformational-specific epitopes within SARS-CoV-2 spike (S) protein, only PD5, PD7, and SC23 were able to bind to the receptor binding domain (RBD). Immunofluorescence microscopy was used to examine the S protein RBD in cells infected with the Singapore isolates SARS-CoV-2/0334 and SARS-CoV-2/1302. The RBD-binders exhibited a distinct cytoplasmic staining pattern that was primarily localized within the Golgi complex and was distinct from the diffuse cytoplasmic staining pattern exhibited by the non-RBD-binders (PD4 and SC29). These data indicated that the S protein adopted a conformation in the Golgi complex that enabled the RBD recognition by the RBD-binders. The RBD-binders also recognized the uncleaved S protein, indicating that S protein cleavage was not required for RBD recognition. Electron microscopy indicated high levels of cell-associated virus particles, and multiple cycle virus infection using RBD-binder staining provided evidence for direct cell-to-cell transmission for both isolates. Although similar levels of RBD-binder staining were demonstrated for each isolate, SARS-CoV-2/1302 exhibited slower rates of cell-to-cell transmission. These data suggest that a conformational change in the S protein occurs during its transit through the Golgi complex that enables RBD recognition by the RBD-binders and suggests that these antibodies can be used to monitor S protein RBD formation during the early stages of infection. IMPORTANCE The SARS-CoV-2 spike (S) protein receptor binding domain (RBD) mediates the attachment of SARS-CoV-2 to the host cell. This interaction plays an essential role in initiating virus infection, and the S protein RBD is therefore a focus of therapeutic and vaccine interventions. However, new virus variants have emerged with altered biological properties in the RBD that can potentially negate these interventions. Therefore, an improved understanding of the biological properties of the RBD in virus-infected cells may offer future therapeutic strategies to mitigate SARS- CoV-2 infection. We used physiologically relevant antibodies that were isolated from the B cells of convalescent COVID-19 patients to monitor the RBD in cells infected with SARS-CoV-2 clinical isolates. These immunological reagents specifically recognize the correctly folded RBD and were used to monitor the appearance of the RBD in SARS-CoV-2-infected cells and identified the site where the RBD first appears.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , COVID-19 , Spike Glycoprotein, Coronavirus , Antibodies, Monoclonal/metabolism , Antibodies, Viral/metabolism , Humans , Protein Binding , Protein Domains , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemical synthesis , Spike Glycoprotein, Coronavirus/metabolism
5.
Virology ; 557: 86-99, 2021 05.
Article in English | MEDLINE | ID: mdl-33677389

ABSTRACT

The distributions of the rac1, rhoA and cdc42 proteins in respiratory syncytial virus (RSV) infected cells was examined. All three rhoGTPases were detected within inclusion bodies, and while the rhoA and rac1 proteins were associated with virus filaments, only the rac1 protein was localised throughout the virus filaments. RSV infection led to increased rac1 protein activation, and using the rac1 protein inhibitor NS23766 we provided evidence that the increased rac1 activation occurred at the site of RSV assembly and facilitated F-actin remodeling during virus morphogenesis. A non-infectious virus-like particle (VLP) assay showed that the RSV VLPs formed in lipid-raft microdomains containing the rac1 protein, together with F-actin and filamin-1 (cell proteins associated with virus filaments). This provided evidence that the virus envelope proteins are trafficked to membrane microdomains containing the rac1 protein. Collectively, these data provide evidence that the rac1 protein plays a direct role in the RSV assembly process.


Subject(s)
Respiratory Syncytial Virus, Human/physiology , Virus Assembly , rac1 GTP-Binding Protein/genetics , Actins/metabolism , Cell Line , Humans , Respiratory Syncytial Virus, Human/genetics , rac1 GTP-Binding Protein/metabolism
6.
Cells ; 9(2)2020 02 15.
Article in English | MEDLINE | ID: mdl-32075271

ABSTRACT

Although the influenza A virus H7N9 subtype circulates within several avian species, it can also infect humans with a severe disease outcome. To better understand the biology of the H7N9 virus we examined the host response to infection in avian and human cells. In this study we used the A/Anhui/1/2013 strain, which was isolated during the first wave of the H7N9 epidemic. The H7N9 virus-infected both human (Airway Epithelial cells) and avian (Chick Embryo Fibroblast) cells, and each infected host transcriptome was examined with bioinformatic tools and compared with other representative avian and human influenza A virus subtypes. The H7N9 virus induced higher expression changes (differentially regulated genes) in both cell lines, with more prominent changes observed in avian cells. Ortholog mapping of differentially expression genes identified significant enriched common and cell-type pathways during H7N9 infections. This data confirmed our previous findings that different influenza A virus subtypes have virus-specific replication characteristics and anti-virus signaling in human and avian cells. In addition, we reported for the first time, the new HIPPO signaling pathway in avian cells, which we hypothesized to play a vital role to maintain the antiviral state of H7N9 virus-infected avian cells. This could explain the absence of disease symptoms in avian species that tested positive for the presence of H7N9 virus.


Subject(s)
Influenza A Virus, H7N9 Subtype/physiology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , A549 Cells , Animals , Chick Embryo , Chickens , Dogs , Gene Expression , Humans , Influenza in Birds/genetics , Influenza in Birds/metabolism , Influenza in Birds/virology , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/metabolism , Signal Transduction
7.
Cells ; 9(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-32028682

ABSTRACT

In humans, (A549) cells impaired H9N2 virus nuclear export of the ribonucleoprotein (RNP) complex contrasted with the early and efficient nuclear export of the H1N1/WSN and pH1N1 virus RNP complexes. Although nuclear export of the RNP complex occurred via the nuclear pore complex, H9N2 virus infection also induced modifications in the nuclear envelope and induced cell cytotoxicity. Reduced PA protein levels in H9N2 virus-infected A549 cells occurred, and this phenomenon was independent of virus infection. Silencing the H1N1/WSN PA protein expression leads to impaired nuclear export of RNP complexes, suggesting that the impaired nuclear export of the H9N2 virus RNP complex may be one of the consequences of reduced PA protein levels. Early and efficient export of the RNP complex occurred in H9N2 virus-infected avian (CEF) cells, although structural changes in the nuclear envelope also occurred. Collectively our data suggest that a combination of delayed nuclear export and virus-induced cell cytotoxicity restricts H9N2 virus transmission in A549 cells. However, the early and efficient export of the RNP complex mitigated the effects of virus-induced cytotoxicity on H9N2 virus transmission in CEF cells. Our findings highlight the multi-factorial nature of host-adaptation of the polymerase proteins of avian influenza viruses in non-avian cell environments.


Subject(s)
Cell Nucleus/metabolism , Ducks/virology , Influenza A Virus, H9N2 Subtype/physiology , Lung/pathology , Lung/virology , Ribonucleoproteins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Death , Cell Line , Chickens , Humans , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Viral Proteins/metabolism , Virus Replication
8.
Viruses ; 10(12)2018 12 05.
Article in English | MEDLINE | ID: mdl-30563103

ABSTRACT

The poxviruses are large, linear, double-stranded DNA viruses about 130 to 230 kbp, that have an animal origin and evolved to infect a wide host range. Variola virus (VARV), the causative agent of smallpox, is a poxvirus that infects only humans, but other poxviruses such as monkey poxvirus and cowpox virus (CPXV) have crossed over from animals to infect humans. Therefore understanding the biology of poxviruses can devise antiviral strategies to prevent these human infections. In this study we used a system-based approach to examine the host responses to three orthopoxviruses, CPXV, vaccinia virus (VACV), and ectromelia virus (ECTV) in the murine macrophage RAW 264.7 cell line. Overall, we observed a significant down-regulation of gene expressions for pro-inflammatory cytokines, chemokines, and related receptors. There were also common and virus-specific changes in the immune-regulated gene expressions for each poxvirus-infected RAW cells. Collectively our results showed that the murine macrophage RAW 264.7 cell line is a suitable cell-based model system to study poxvirus host response.


Subject(s)
Cowpox virus/immunology , Cytokines/immunology , Ectromelia virus/immunology , Macrophages/immunology , Vaccinia virus/immunology , Animals , Chemokines/genetics , Chemokines/immunology , Cytokines/genetics , Down-Regulation , Gene Expression , Macrophages/virology , Mice , Microarray Analysis , Polymerase Chain Reaction , RAW 264.7 Cells , Up-Regulation
9.
Virology ; 521: 20-32, 2018 08.
Article in English | MEDLINE | ID: mdl-29870884

ABSTRACT

Respiratory syncytial virus infection was examined using a human nasal epithelial cell model. Maximum levels of shed-virus were produced at between 3 and 5 days post-infection (dpi), and the infectivity of the shed-virus was stable up to 10 dpi. The highest levels of interferon signalling were recorded at 2dpi, and infection induced a widespread antivirus response in the nasal epithelium, involving both infected cells and non-infected cells. Although these cellular responses were associated with reduced levels of progeny virus production and restricted virus spread, they did not inhibit the infectivity virus that is shed early in infection. In the clinical context these data suggest that although the host cell response in the nasal epithelium may restrict the levels of progeny virus particles produced, the stability of the shed-virus in the nasal mucosa may be an important factor in both disease progression and virus transmission.


Subject(s)
Interferon-beta/immunology , Interleukins/immunology , Nasal Mucosa/immunology , Respiratory Syncytial Virus, Human/physiology , Virus Replication , Cells, Cultured , Humans , Interferons , Nasal Mucosa/virology , Oxidoreductases Acting on CH-CH Group Donors , Polymerase Chain Reaction , Proteins/metabolism , Respiratory Syncytial Virus, Human/genetics , Signal Transduction , Virus Shedding
10.
Int J Mol Sci ; 18(11)2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29104227

ABSTRACT

Low pathogenic avian influenza (LPAI) viruses are a source of sporadic human infections and could also contribute to future pandemic outbreaks but little is known about inter-species differences in the host responses to these viruses. Here, we studied host gene expression signatures of cell lines from three species (human, chicken, and canine) in response to six different viruses (H1N1/WSN, H5N2/F59, H5N2/F118, H5N2/F189, H5N3 and H9N2). Comprehensive microarray probe set re-annotation and ortholog mapping of the host genes was necessary to allow comparison over extended functionally annotated gene sets and orthologous pathways. The annotations are made available to the community for commonly used microarray chips. We observe a strong tendency of the response being cell type- rather than virus-specific. In chicken cells, we found up-regulation of host factors inducing virus infectivity (e.g., oxysterol binding protein like 1A (OSBPL1A) and Rho GTPase activating protein 21 (ARHGAP21)) while reducing apoptosis (e.g., mitochondrial ribosomal protein S27 (MRPS27)) and increasing cell proliferation (e.g., COP9 signalosome subunit 2 (COPS2)). On the other hand, increased antiviral, pro-apoptotic and inflammatory signatures have been identified in human cells while cell cycle and metabolic pathways were down-regulated. This signature describes how low pathogenic avian influenza (LPAI) viruses are being tolerated and shed from chicken but potentially causing cellular disruption in mammalian cells.


Subject(s)
Influenza A virus/physiology , Orthomyxoviridae Infections/genetics , Transcriptome , Animals , Apoptosis , Cell Line , Chickens , Dogs , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N2 Subtype/physiology , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/genetics , Influenza in Birds/metabolism , Influenza in Birds/virology , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Metabolic Networks and Pathways , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Species Specificity
11.
Sci Rep ; 7(1): 5805, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724991

ABSTRACT

Several available online tools provide network growing functions where an algorithm utilizing different data sources suggests additional genes/proteins that should connect an input gene set into functionally meaningful networks. Using the well-studied system of influenza host interactions, we compare the network growing function of two free tools GeneMANIA and STRING and the commercial IPA for their performance of recovering known influenza A virus host factors previously identified from siRNA screens. The result showed that given small (~30 genes) or medium (~150 genes) input sets all three network growing tools detect significantly more known host factors than random human genes with STRING overall performing strongest. Extending the networks with all the three tools significantly improved the detection of GO biological processes of known host factors compared to not growing networks. Interestingly, the rate of identification of true host factors using computational network growing is equal or better to doing another experimental siRNA screening study which could also be true and applied to other biological pathways/processes.


Subject(s)
Benchmarking , Computational Biology/methods , Gene Regulatory Networks , Host-Pathogen Interactions/genetics , Viruses/metabolism , Algorithms , Gene Ontology , Humans , RNA, Small Interfering/metabolism , Signal Transduction
12.
BMC Genomics ; 18(1): 420, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28558796

ABSTRACT

BACKGROUND: Influenza A virus (IAV) is a major public health concern, being responsible for the death of approximately half a million people each year. Zoonotic transmissions of the virus from swine and avian origin have occurred in the past, and can potentially lead to the emgergence of new IAV stains in future pandemics. Pulmonary macrophages have been implicated in disease severity in the lower airway, and understanding the host response of macrophages infected with avian influenza viruses should provide new therapeutic strategies. RESULTS: We used a systems-based approach to investigate the transcriptome response of primary murine lung macrophages (PMФ) infected with the mouse-adapted H1N1/WSN virus and low pathogenic avian influenza (LPAI) viruses H5N2 and H5N3. The results showed that the LPAI viruses H5N2 and H5N3 can infect PMФ with similar efficiency to the H1N1/WSN virus. While all viruses induced antiviral responses, the H5N3 virus infection resulted in higher expression levels of cytokines and chemokines associated with inflammatory responses. CONCLUSIONS: The LPAI H5N2 and H5N3 viruses are able to infect murine lung macrophages. However, the H5N3 virus was associated with increased expression of pro-inflammatory mediators. Although the H5N3 virus it is capable of inducing high levels of cytokines that are associated with inflammation, this property is distinct from its inability to efficiently replicate in a mammalian host.


Subject(s)
Cytokines/genetics , Gene Expression Profiling , Host-Pathogen Interactions/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N2 Subtype/physiology , Lung/immunology , Macrophages/metabolism , Animals , Asia , Cell Death/drug effects , Cell Death/genetics , Host-Pathogen Interactions/immunology , Interferons/pharmacology , Lung/virology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/immunology , Mice , Molecular Sequence Annotation
13.
J Cell Sci ; 130(6): 1037-1050, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28154158

ABSTRACT

Respiratory syncytial virus (RSV) is an enveloped virus that assembles into filamentous virus particles on the surface of infected cells. Morphogenesis of RSV is dependent upon cholesterol-rich (lipid raft) membrane microdomains, but the specific role of individual raft molecules in RSV assembly is not well defined. Here, we show that RSV morphogenesis occurs within caveolar membranes and that both caveolin-1 and cavin-1 (also known as PTRF), the two major structural and functional components of caveolae, are actively recruited to and incorporated into the RSV envelope. The recruitment of caveolae occurred just prior to the initiation of RSV filament assembly, and was dependent upon an intact actin network as well as a direct physical interaction between caveolin-1 and the viral G protein. Moreover, cavin-1 protein levels were significantly increased in RSV-infected cells, leading to a virus-induced change in the stoichiometry and biophysical properties of the caveolar coat complex. Our data indicate that RSV exploits caveolae for its assembly, and we propose that the incorporation of caveolae into the virus contributes to defining the biological properties of the RSV envelope.


Subject(s)
Caveolae/metabolism , Cell Membrane/metabolism , Respiratory Syncytial Virus, Human/physiology , Virus Assembly/physiology , Actins/metabolism , Caveolae/ultrastructure , Caveolin 1/metabolism , HeLa Cells , Humans , Models, Biological , Morphogenesis , Protein Binding , Protein Stability , RNA-Binding Proteins/metabolism , Respiratory Syncytial Virus, Human/ultrastructure , Viral Proteins/metabolism
14.
J Gen Virol ; 98(5): 906-921, 2017 May.
Article in English | MEDLINE | ID: mdl-28141511

ABSTRACT

We have examined the expression profile of the influenza virus PA protein in pH1N1/2009 virus-infected cells. Immunoblotting analysis of virus-infected MDCK cells revealed the presence of full-length PA protein from 8 h post-infection, together with the simultaneous appearance of PA protein species of approximately 50, 35/39 and 20/25 kDa (collectively referred to as PA*). PA* was also detected in H1N1/WSN-virus-infected cells, indicating that its presence was not virus-specific, and it was also observed in virus-infected A549 and chick embryo fibroblast (CEF) cells, indicating that its presence was not cell-type-specific. PA* was detected in cells expressing the recombinant PA protein, indicating that the PA* formation occurred in the absence of virus infection. These data collectively indicated that PA* formation is an intrinsic property of PA gene expression. The association of PA* with purified influenza virus particles was demonstrated by immunoblotting, and a protease protection assay provided evidence that PA* was packaged into virus particles. The ribonucleoprotein (RNP) complex was isolated from purified influenza virus particles using glycerol gradient centrifugation, which demonstrated that PA* was associated with the RNP complex. To the best of our knowledge, this is the first report to demonstrate that PA protein species containing only segments of the C-terminal domain form during influenza virus infection. Furthermore, these truncated PA protein species are subsequently packaged into virus particles as part of the functional RNP complex.

15.
Methods Mol Biol ; 1442: 175-94, 2016.
Article in English | MEDLINE | ID: mdl-27464695

ABSTRACT

The identification of cellular factors that play a role in respiratory syncytial virus (RSV) replication is an alternative strategy in the identification of druggable cellular protein that are essential for RSV replication. In this regard experimental strategies that are able to screen relevant proteins from the vast array of proteins in the cellular milieu will facilitate the identification of potential drug targets. In this chapter we describe a procedure where RSV particles are purified from cells that are permissive for RSV infection, and the protein composition of the purified virus particles characterized using a proteomics-based strategy. This procedure revealed that actin, several actin-binding proteins, and the chaperones HSP70 and HSP90 also co-purified with the virus particles. The relevance of the HSP90 protein to virus replication was then further validated using imaging, gene silencing and by using an established small molecule HSP90 inhibitor.


Subject(s)
Proteomics/methods , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/physiology , Virion/physiology , Actins/isolation & purification , Cell Line , Chromatography, Liquid , HSP70 Heat-Shock Proteins/isolation & purification , HSP90 Heat-Shock Proteins/isolation & purification , Humans , Microfilament Proteins/isolation & purification , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/metabolism , Tandem Mass Spectrometry , Virion/metabolism , Virus Replication
16.
Virol J ; 13: 12, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26790623

ABSTRACT

BACKGROUND: During respiratory syncytial virus (RSV) infection filamentous virus particles are formed on the cell surface. Although the virus infectivity remains cell-associated, low levels of cell-free virus is detected during advanced infection. It is currently unclear if this cell-free virus infectivity is due to a low-efficiency specific cell-release mechanism, or if it arises due to mechanical breakage following virus-induced cell damage at the advanced stage of infection. Understanding the origin of this cell-free virus is a prerequisite for understanding the mechanism of RSV transmission in permissive cells. In this study we describe a detailed examination of RSV transmission in permissive HEp2 cell monolayers. METHODS: HEp2 cell monolayers were infected with RSV using a multiplicity of infection of 0.0002, and the course of infection monitored over 5 days. The progression of the virus infection within the cell monolayers was performed using bright-field microscopy to visualise the cell monolayer and immunofluorescence microscopy to detect virus-infected cells. The cell-associated and cell-free virus infectivity were determined by virus plaque assay, and the virus-induced cell cytotoxicity determined by measuring cell membrane permeability and cellular DNA fragmentation. RESULTS: At 2 days-post infection (dpi), large clusters of virus-infected cells could be detected indicating localised transmission in the cell monolayer, and during this stage we failed to detect either cell-free virus or cell cytotoxicity. At 3 dpi the presence of much larger infected cell clusters correlated with the begining of virus-induced changes in cell permeability. The presence of cell-free virus correlated with continued increase in cell permeability and cytotoxicity at 4 and 5 dpi. At 5 dpi extensive cell damage, syncytial formation, and increased cellular DNA fragmentation was noted. However, even at 5 dpi the cell-free virus constituted less than 1 % of the total virus infectivity. CONCLUSIONS: Our data supports a model of RSV transmission that initially involves the localised cell-to-cell spread of virus particles within the HEp2 cell monolayer. However, low levels of cell free-virus infectivity was observed at the advanced stages of infection, which correlated with a general loss in cell monolayer integrity due to virus-induced cytotoxicity.


Subject(s)
Respiratory Syncytial Virus, Human/physiology , Virus Replication , Actins/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane Permeability , Cells, Cultured , DNA Fragmentation , Humans , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/transmission , Respiratory Syncytial Virus Infections/virology , Signal Transduction
18.
Microb Genom ; 2(8): e000082, 2016 08.
Article in English | MEDLINE | ID: mdl-28348873

ABSTRACT

In this study we compared the NS1 protein of Influenza B/Lee/40 and several non-cultured Influenza B virus clinical strains detected in Singapore. In B/Lee/40 virus-infected cells and in cells expressing the recombinant B/Lee/40 NS1 protein a full-length 35 kDa NS1 protein and a 23 kDa NS1 protein species (p23) were detected. Mutational analysis of the NS1 gene indicated that p23 was generated by a novel cleavage event within the linker domain between an aspartic acid and proline at amino acid residues at positions 92 and 93 respectively (DP92-93), and that p23 contained the first 92 amino acids of the NS1 protein. Sequence analysis of the Singapore strains indicated the presence of either DP92-93 or NP92-93 in the NS1 protein, but protein expression analysis showed that p23 was only detected in NS1 proteins with DP92-93.. An additional adjacent proline residue at position 94 (P94) was present in some strains and correlated with increased p23 levels, suggesting that P94 has a synergistic effect on the cleavage of the NS1 protein. The first 145 amino acids of the NS1 protein are required for inhibition of ISG15-mediated ubiquitination, and our analysis showed that Influenza B viruses circulating in Singapore with DP92-93 expressed truncated NS1 proteins and may differ in their capacity to inhibit ISG15 activity. Thus, DP92-93 in the NS1 protein may confer a disadvantage to Influenza B viruses circulating in the human population and interestingly the low frequency of DP92-93detection in the NS1 protein since 2004 is consistent with this suggestion.


Subject(s)
Influenza B virus/genetics , Influenza, Human/virology , Viral Nonstructural Proteins/genetics , Humans , Male , Singapore , Viral Nonstructural Proteins/metabolism
19.
Virology ; 484: 395-411, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26231613

ABSTRACT

The distribution of cilia and the respiratory syncytial virus (RSV) nucleocapsid (N) protein, fusion (F) protein, attachment (G) protein, and M2-1 protein in human ciliated nasal epithelial cells was examined at between 1 and 5 days post-infection (dpi). All virus structural proteins were localized at cell surface projections that were distinct from cilia. The F protein was also trafficked into the cilia, and while its presence increased as the infection proceeded, the N protein was not detected in the cilia at any time of infection. The presence of the F protein in the cilia correlated with cellular changes in the cilia and reduced cilia function. At 5dpi extensive cilia loss and further reduced cilia function was noted. These data suggested that although RSV morphogenesis occurs at non-cilia locations on ciliated nasal epithelial cells, RSV infection induces changes in the cilia body that leads to extensive cilia loss.


Subject(s)
Epithelial Cells/virology , Membrane Microdomains/virology , Respiratory Syncytial Viruses/physiology , Virus Assembly , Adult , Cells, Cultured , Cilia/chemistry , Cilia/virology , Epithelial Cells/chemistry , Humans , Membrane Microdomains/chemistry , Respiratory Syncytial Viruses/chemistry , Viral Structural Proteins/analysis
20.
Antiviral Res ; 114: 11-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25433308

ABSTRACT

We examined the effect of respiratory syncytial virus (RSV) infection on viperin protein expression in the permissive HEp2 and non-permissive RAW 264.7 macrophage cell lines. In RSV-infected HEp2 cells low levels of the viperin protein was localized to the virus-induced inclusion bodies and did not impair virus transmission in these cells. In contrast, RSV-infected RAW 264.7 cells increased expression of the STAT1 protein occurred at between 6 and 12h post-infection, which coincided with the appearance of P-STAT1. A relatively high level of viperin protein expression was detected in infected RAW 264.7 cells, and it was extensively localized throughout the cytoplasm of infected cells. The effect of early viperin protein expression on RSV infection in cells that are normally permissive to RSV cultivation was examined by using either transient transfected HEp2 cells or stable transfected HeLa cells that expressed the viperin protein. The early expression of viperin in HeLa cells did not prevent virus infection, and no significant inhibitory effect on either virus protein expression or targeting of virus proteins to the cell surface was noted. However, while inclusion body formation was not inhibited, early viperin protein expression was associated with the inhibition of virus filament formation and reduced cell-to-cell virus transmission. Inhibition of virus filament formation was also observed in HEp2 cells expressing viperin. Collectively our data suggested that viperin impaired RSV transmission by inhibiting virus filament formation, providing a basis for its anti-virus activity in RSV-infected cells.


Subject(s)
Macrophages/virology , Proteins/metabolism , Respiratory Syncytial Virus, Human/physiology , Animals , Cell Line , Cell Line, Tumor , Fluorescent Antibody Technique , Gene Expression Regulation , HeLa Cells , Humans , Inclusion Bodies, Viral/metabolism , Inclusion Bodies, Viral/ultrastructure , Macrophages/metabolism , Mice , Morphogenesis , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Real-Time Polymerase Chain Reaction , Respiratory Syncytial Virus, Human/genetics , STAT1 Transcription Factor/metabolism , Viral Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...