Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 29(3): 1174-1185, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33352107

RESUMO

Self-amplifying RNA (saRNA) is a cutting-edge platform for both nucleic acid vaccines and therapeutics. saRNA is self-adjuvanting, as it activates types I and III interferon (IFN), which enhances the immunogenicity of RNA vaccines but can also lead to inhibition of translation. In this study, we screened a library of saRNA constructs with cis-encoded innate inhibiting proteins (IIPs) and determined the effect on protein expression and immunogenicity. We observed that the PIV-5 V and Middle East respiratory syndrome coronavirus (MERS-CoV) ORF4a proteins enhance protein expression 100- to 500-fold in vitro in IFN-competent HeLa and MRC5 cells. We found that the MERS-CoV ORF4a protein partially abates dose nonlinearity in vivo, and that ruxolitinib, a potent Janus kinase (JAK)/signal transducer and activator of transcription (STAT) inhibitor, but not the IIPs, enhances protein expression of saRNA in vivo. Both the PIV-5 V and MERS-CoV ORF4a proteins were found to enhance the percentage of resident cells in human skin explants expressing saRNA and completely rescued dose nonlinearity of saRNA. Finally, we observed that the MERS-CoV ORF4a increased the rabies virus (RABV)-specific immunoglobulin G (IgG) titer and neutralization half-maximal inhibitory concentration (IC50) by ∼10-fold in rabbits, but not in mice or rats. These experiments provide a proof of concept that IIPs can be directly encoded into saRNA vectors and effectively abate the nonlinear dose dependency and enhance immunogenicity.


Assuntos
Imunidade Inata/efeitos dos fármacos , Imunogenicidade da Vacina , Biossíntese de Proteínas/efeitos dos fármacos , Vacinas Sintéticas/farmacologia , Proteínas do Envelope Viral/administração & dosagem , Animais , Linhagem Celular , Vírus da Encefalite Equina Venezuelana/efeitos dos fármacos , Vírus da Encefalite Equina Venezuelana/imunologia , Vírus da Encefalite Equina Venezuelana/patogenicidade , Fibroblastos , Regulação da Expressão Gênica , Células HeLa , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoglobulina G/biossíntese , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Janus Quinases/antagonistas & inibidores , Janus Quinases/genética , Janus Quinases/imunologia , Camundongos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , NF-kappa B/genética , NF-kappa B/imunologia , Nitrilas , Vírus da Parainfluenza 5/efeitos dos fármacos , Vírus da Parainfluenza 5/imunologia , Vírus da Parainfluenza 5/patogenicidade , Pirazóis/farmacologia , Pirimidinas , Coelhos , Vírus da Raiva/efeitos dos fármacos , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Ratos , Fatores de Transcrição STAT/antagonistas & inibidores , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Vacinas Sintéticas/biossíntese , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas de mRNA
2.
PLoS Pathog ; 15(2): e1007561, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30742688

RESUMO

Paramyxoviruses can establish persistent infections both in vitro and in vivo, some of which lead to chronic disease. However, little is known about the molecular events that contribute to the establishment of persistent infections by RNA viruses. Using parainfluenza virus type 5 (PIV5) as a model we show that phosphorylation of the P protein, which is a key component of the viral RNA polymerase complex, determines whether or not viral transcription and replication becomes repressed at late times after infection. If the virus becomes repressed, persistence is established, but if not, the infected cells die. We found that single amino acid changes at various positions within the P protein switched the infection phenotype from lytic to persistent. Lytic variants replicated to higher titres in mice than persistent variants and caused greater infiltration of immune cells into infected lungs but were cleared more rapidly. We propose that during the acute phases of viral infection in vivo, lytic variants of PIV5 will be selected but, as the adaptive immune response develops, variants in which viral replication can be repressed will be selected, leading to the establishment of prolonged, persistent infections. We suggest that similar selection processes may operate for other RNA viruses.


Assuntos
Infecções por Paramyxoviridae/genética , Paramyxoviridae/genética , Fosfoproteínas/genética , Proteínas Virais/genética , Células A549 , Substituição de Aminoácidos/genética , Animais , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/patogenicidade , Paramyxoviridae/patogenicidade , Infecções por Paramyxoviridae/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Fosforilação , RNA Viral , Proteínas Virais/metabolismo , Proteínas Virais/fisiologia , Replicação Viral
4.
Virus Res ; 178(2): 423-9, 2013 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-24050998

RESUMO

A novel porcine parainfluenza 5 (pPIV5), KNU-11, in the genus Rubulavirus of the subfamily Paramyxovirinae, was isolated from the lung of a piglet in Korea in 2011. To understand the importance of this virus as an infectious agent, in vitro and in vivo characteristics of KNU-11 virus was investigated. KNU-11 was remarkably cytopathogenic, showing distinct cell rounding and clumping evident in porcine alveolar macrophage (PAM), porcine kidney (PK-15), and swine testicle (ST) cells within 12h postinfection and capable of hemagglutinating guinea pig red blood cells. Interestingly, this cytopathology was found to be absent in cell lines from other mammalian species. To evaluate the in vitro immunity of the pPIV5 isolate, we sought to explore alteration of inflammatory cytokine and chemokine expression in PAM cells infected with KNU-11 by using quantitative real-time RT-PCR. Most cytokine and chemokine genes including type 1 interferons (IFN-α/ß) and IFN-related antiviral genes were found to be significantly elevated in KNU-11 virus-infected PAM cells. A serum neutralization test-based serosurvey demonstrated that neutralizing antibodies against KNU-11 are readily detected in domestic swine populations, suggesting high prevalence of pPIV5 in Korean pig farms. Animal studies showed that KNU-11 fails to establish an acute respiratory illness, indicating that pPIV5 is non- or very mildly pathogenic to pigs.


Assuntos
Vírus da Parainfluenza 5/isolamento & purificação , Vírus da Parainfluenza 5/fisiologia , Infecções por Rubulavirus/veterinária , Doenças dos Suínos/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linhagem Celular , Citocinas/biossíntese , Citocinas/genética , Efeito Citopatogênico Viral , Perfilação da Expressão Gênica , Cobaias , Hemaglutinação , Pulmão/virologia , Testes de Neutralização , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/patogenicidade , Reação em Cadeia da Polimerase em Tempo Real , República da Coreia/epidemiologia , Infecções por Rubulavirus/epidemiologia , Infecções por Rubulavirus/virologia , Estudos Soroepidemiológicos , Suínos , Doenças dos Suínos/epidemiologia
5.
Med Mol Morphol ; 44(4): 213-20, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22179184

RESUMO

A recent report has indicated that proteins and genes of simian virus 5 (SV5) are detected in a human gastric adenocarcinoma (AGS) cell line, which is widely provided for oncology, immunology, and microbiology research. However, the production of infective virions has not been determined in this cell line. In this study, the morphology and infectivity of the virus particles of the AGS cell line were studied by light and electron microscopy and virus transmission assay. The virus particles were approximately 176.0 ± 41.1 nm in diameter. The particles possessed projections 8-12 nm long on the surface and contained a nucleocapsid determined to be 13-18 nm in width and less than 1,000 nm in length. The virus was transmissible to the Vero cell line, induced multinuclear giant cell formation, and reproduced the same shape of antigenic virions. In this study, the persistently infected virus in the AGS cell line was determined to be infective and form reproducible virions, and a new morphological feature of SV5 was determined.


Assuntos
Linhagem Celular/virologia , Vírus da Parainfluenza 5/patogenicidade , Vírus da Parainfluenza 5/ultraestrutura , Infecções por Rubulavirus/virologia , Vírion/patogenicidade , Vírion/ultraestrutura , Animais , Anticorpos Antivirais/farmacologia , Forma Celular/efeitos dos fármacos , Helicobacter pylori/fisiologia , Humanos , Corpos de Inclusão Viral/metabolismo , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Vírus da Parainfluenza 5/imunologia , Coelhos , Infecções por Rubulavirus/transmissão , Vírion/imunologia
6.
Virology ; 405(2): 383-9, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20605567

RESUMO

The paramyxovirus Simian Virus 5 (SV5) is a poor inducer of interferon (IFN) secretion in all cell types tested so far, including primary epithelial cells and primary human myeloid dendritic cells. SV5 is hypothesized to limit induction of antiviral responses through control of viral gene expression and production of the V protein antagonist. Plasmacytoid dendritic cells (pDCs) are known to uniquely express toll-like receptor (TLR)-7 and are a main producer of IFN-alpha among peripheral blood mononuclear cells in response to many viruses. Here, we tested whether SV5 would remain a poor inducer of IFN in primary human pDCs. The efficiency of SV5 infection of pDCs could be increased by an increasing multiplicity of infection. pDCs infected by both live and UV-inactivated SV5 induced large amounts of IFN-alpha secretion and resulted in upregulation of maturation markers CD80 and CD86. However, IL-6 secretion was not induced by SV5 infection. When TLR7 signaling was inhibited, SV5 induced less IFN secretion and CD80 expression, and there was a corresponding increase in number of infected cells. Similar effects were seen with inhibitors of cellular autophagy pathways, suggesting that the SV5 activation of pDC requires access to the cytoplasm and autophagic sampling of cytoplasmic contents. These results have implications for control of SV5 infections in vivo and for development of SV5 as a vaccine vector.


Assuntos
Autofagia , Células Dendríticas/imunologia , Interferon-alfa/metabolismo , Vírus da Parainfluenza 5/patogenicidade , Receptor 7 Toll-Like/metabolismo , Animais , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Células Cultivadas , Células Dendríticas/virologia , Humanos , Interferon-alfa/imunologia , Vírus da Parainfluenza 5/imunologia , Vírus da Parainfluenza 5/efeitos da radiação
7.
J Virol ; 83(18): 9057-67, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19587032

RESUMO

The parainfluenza virus simian virus 5 (SV5) is a poor inducer of innate immune responses. In contrast, the naturally occurring SV5 variant Wake Forest parainfluenza virus (WF-PIV) activates the synthesis of proinflammatory cytokines and beta interferon (IFN-beta). Comparison of SV5 and WF-PIV genome sequences revealed nine nucleotide differences within the viral genomic promoter, including two substitutions (U5C and A14G) in the most highly conserved 3'-end promoter element. To test the consequences of these promoter variations, a recombinant SV5 mutant [Le-(U5C, A14G)] was engineered to harbor the two WF-PIV genomic promoter substitutions in an otherwise wild-type (WT) SV5 background. Human lung epithelial cells infected with the Le-(U5C, A14G) mutant had higher rates of viral protein synthesis and levels of mRNA than cells infected with WT SV5, but levels of genomic RNA were not changed. Unlike WT SV5, the Le-(U5C, A14G) mutant was a potent inducer of interleukin-6 and IFN-beta synthesis, despite expressing a functional V protein antagonist. Cytokine responses to Le-(U5C, A14G) infection were reduced either by small interfering RNA-mediated knockdown of retinoic acid-inducible gene I (RIG-I) or after infection of cells that were engineered to express the reovirus sigma3 double-stranded RNA-binding protein. Le-(U5C, A14G) induced cytopathic effects not seen with WT SV5, and the extent of cell killing correlated with elevated levels of viral F protein and cell-cell fusion. Our results support a model whereby the SV5 promoter has evolved to function at an attenuated level in order to limit (i) synthesis of aberrant RNAs which induce RIG-I-mediated responses and (ii) overproduction of mRNA for potentially toxic gene products, such as the F protein. Control of genomic promoter activity may be particularly important for viruses such as SV5, that express a V protein targeting mda-5 but do not encode antagonists such as the paramyxovirus C proteins, that specifically target RIG-I.


Assuntos
Genoma Viral/imunologia , Imunidade Inata , Vírus da Parainfluenza 5/patogenicidade , Regiões Promotoras Genéticas/genética , Células Cultivadas , Variação Genética , Humanos , Vírus da Parainfluenza 5/imunologia , Paramyxovirinae/imunologia , Paramyxovirinae/patogenicidade , Mutação Puntual , Biossíntese de Proteínas , RNA Viral/genética , Proteínas Virais
8.
Virology ; 368(2): 262-72, 2007 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-17692882

RESUMO

The V protein of parainfluenza virus 5 (PIV5) plays an important role in the evasion of host immune responses. The V protein blocks interferon (IFN) signaling in human cells by causing degradation of the STAT1 protein, a key component of IFN signaling, and blocks IFN-beta production by preventing nuclear translocation of IRF3, a key transcription factor for activating IFN-beta promoter. Interleukin-6 (IL-6), along with tumor necrosis factor (TNF)-alpha and IL-1beta, is a major proinflammatory cytokine that plays important roles in clearing virus infection through inflammatory responses. Many viruses have developed strategies to block IL-6 expression. Wild-type PIV5 infection induces little, if any, expression of cytokines such as IL-6 or TNF-alpha, whereas infection by a mutant PIV5 lacking the conserved C-terminal cysteine rich domain (rPIV5VDeltaC) induced high levels of IL-6 expression. Examination of mRNA levels of IL-6 indicated that the transcription activation of IL-6 played an important role in the increased IL-6 expression. Co-infection with wild-type PIV5 prevented the activation of IL-6 transcription by rPIV5VDeltaC, and a plasmid encoding the full-length PIV5 V protein prevented the activation of IL-6 promoter-driven reporter gene expression by rPIV5VDeltaC, indicating that the V protein played a role in inhibiting IL-6 transcription. The activation of IL-6 was independent of IFN-beta even though rPIV5VDeltaC-infected cells produced IFN-beta. Using reporter gene assays and chromatin immunoprecipitation (ChIP), it was found that NF-kappaB played an important role in activating expression of IL-6. We have proposed a model of activating and inhibiting IL-6 transcription by PIV5.


Assuntos
Regulação da Expressão Gênica , Interleucina-6/metabolismo , Vírus da Parainfluenza 5/patogenicidade , Proteínas Estruturais Virais/farmacologia , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Genes Reporter , Células HeLa , Humanos , Interleucina-6/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/metabolismo , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo
9.
J Gen Virol ; 87(Pt 12): 3643-3648, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17098980

RESUMO

The paramyxovirus Simian virus 5 (SV5) is largely non-cytopathic in human epithelial and fibroblast cells. WF-PIV has been described previously as a naturally occurring SV5 variant that encodes P and V proteins differing from the wild-type (WT) SV5 proteins in eight and five amino acid positions, respectively. In this study, it is shown that WF-PIV is like WT SV5 by being largely non-cytopathic in A549 lung epithelial cells. However, substitution of the WF-PIV P/V gene into the background of WT SV5 resulted in a hybrid virus (P/V-WF) that induced apoptotic cell death not seen with either of the parental viruses. The kinetics of HeLa cell killing and induction of apoptosis by the P/V-WF chimera differed from those of the previously described P/V-CPI- chimera by being slower and less extensive. HeLa cell killing by the P/V-WF chimera was effectively reduced by inhibitors of caspase-9, but not of caspase-8. These results demonstrate that an exchange of P/V genes from two non-cytopathic SV5 variants can produce apoptosis-inducing chimeras, and that the role of the SV5 P/V gene products in limiting apoptosis can be dependent on expression in the context of a native viral genome.


Assuntos
Inibidores de Caspase , Sobrevivência Celular , Células Epiteliais/virologia , Vírus da Parainfluenza 5/patogenicidade , Fosfoproteínas/genética , Recombinação Genética , Proteínas Virais/genética , Proteínas Estruturais Virais/genética , Apoptose , Linhagem Celular , Linhagem Celular Tumoral , Efeito Citopatogênico Viral/genética , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Vírus da Parainfluenza 5/genética , Proteínas de Ligação a RNA
10.
Biochem Biophys Res Commun ; 348(3): 916-22, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16904649

RESUMO

The fusion proteins of enveloped viruses mediating the fusion between the viral and cellular membranes comprise two discontinuous heptad repeat (HR) domains located at the ectodomain of the enveloped glycoproteins. The crystal structure of the fusion protein core of Mumps virus (MuV) was determined at 2.2 A resolution. The complex is a six-helix bundle in which three HR1 peptides form a central highly hydrophobic coiled-coil and three HR2 peptides pack against the hydrophobic grooves on the surface of central coiled-coil in an oblique antiparallel manner. Fusion core of MuV, like those of simian virus 5 and human respiratory syncytium virus, forms typical 3-4-4-4-3 spacing. The similar characterization in HR1 regions, as well as the existence of O-X-O motif in extended regions of HR2 helix, suggests a basic rule for the formation of the fusion core of viral fusion proteins.


Assuntos
Fusão de Membrana/fisiologia , Vírus da Caxumba/química , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Sequência Conservada , Cristalografia por Raios X , Dados de Sequência Molecular , Vírus da Caxumba/patogenicidade , Vírus da Parainfluenza 5/química , Vírus da Parainfluenza 5/patogenicidade , Fragmentos de Peptídeos/química , Conformação Proteica , Estrutura Secundária de Proteína , Sequências Repetitivas de Aminoácidos , Proteínas Virais de Fusão/fisiologia
11.
J Virol ; 80(7): 3416-27, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16537609

RESUMO

Human epithelial cells infected with the parainfluenza virus simian virus 5 (SV5) show minimal activation of host cell interferon (IFN), cytokine, and cell death pathways. In contrast, a recombinant SV5 P/V gene mutant (rSV5-P/V-CPI-) overexpresses viral gene products and is a potent inducer of IFN, proinflammatory cytokines, and apoptosis in these cells. In this study, we have compared the outcomes of wild-type (WT) SV5 and rSV5-P/V-CPI- infections of primary human dendritic cells (DC), important antigen-presenting cells for initiating adaptive immune responses. We have tested the hypothesis that a P/V mutant which activates host antiviral responses will be a more potent inducer of DC maturation and function than WT rSV5, which suppresses host cell responses. Infection of peripheral blood mononuclear cell-derived immature DC with WT rSV5 resulted in high levels of viral protein and progeny virus but very little increase in cell surface costimulatory molecules or secretion of IFN and proinflammatory cytokines. In contrast, immature DC infected with the rSV5-P/V-CPI- mutant produced only low levels of viral protein and progeny virus, but these infected cells were induced to secrete IFN-alpha and other cytokines and showed elevated levels of maturation markers. Unexpectedly, DC infected with WT rSV5 showed extensive cytopathic effects and increased levels of active caspase-3, while infection of DC with the P/V mutant was largely noncytopathic. In mixed-culture assays, WT rSV5-infected DC were impaired in the ability to stimulate proliferation of autologous CD4+ T cells, whereas DC infected with the P/V mutant were very effective at activating T-cell proliferation. The addition of a pancaspase inhibitor to DC infected with WT rSV5 reduced cytopathic effects and resulted in higher surface expression levels of maturation markers. Our finding that the SV5 P/V mutant has both a reduced cytopathic effect in human DC compared to WT SV5 and an enhanced ability to induce DC function has implications for the rational design of novel recombinant paramyxovirus vectors based on engineered mutations in the viral P/V gene.


Assuntos
Células Dendríticas/virologia , Mutação , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/patogenicidade , Fosfoproteínas/fisiologia , Proteínas Virais/fisiologia , Proteínas Estruturais Virais/fisiologia , Apoptose , Western Blotting , Caspase 3 , Caspases/análise , Proliferação de Células , Células Cultivadas , Efeito Citopatogênico Viral , Células Dendríticas/citologia , Células Dendríticas/enzimologia , Ativação Enzimática , Corantes Fluorescentes , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Indóis , Interferon Tipo I/metabolismo , Microscopia de Fluorescência , Vírus da Parainfluenza 5/crescimento & desenvolvimento , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Proteínas de Ligação a RNA , Recombinação Genética , Fator de Transcrição STAT1/metabolismo , Linfócitos T/fisiologia , Proteínas Virais/biossíntese , Proteínas Virais/genética , Proteínas Estruturais Virais/biossíntese , Proteínas Estruturais Virais/genética
12.
Virology ; 347(1): 11-27, 2006 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-16375939

RESUMO

The fusion (F) protein of simian virus 5 strain W3A induces syncytium formation independently of coexpression of the hemagglutinin-neuraminidase protein. This property can be transferred to the F protein of strain WR by replacing the leucine at position 22 with the W3A F counterpart, proline. The resulting mutant L22P has a conformation that is distinct from that of the WR F protein. Se-L22P is a cleavage site mutant of L22P that is cleavable only by addition of exogenous trypsin. We showed here that the cell surface-localized L22P was internalized with a t1/2 of 25 min and degraded in the cell, while the WR F protein was not. The cell surface-localized Se-L22P underwent a significant conformational change upon cleavage. Intriguingly, it disappeared from the cell surface due to its internalization, while inducing extensive syncytium formation. These results indicate that L22P may display an internalization signal during the course of fusion induction.


Assuntos
Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/fisiologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/fisiologia , Substituição de Aminoácidos , Transporte Biológico Ativo , Células Gigantes/virologia , Células HeLa , Hemaglutininas Virais/fisiologia , Humanos , Peso Molecular , Neuraminidase/fisiologia , Vírus da Parainfluenza 5/patogenicidade , Mutação Puntual , Conformação Proteica , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Proteínas Virais de Fusão/química
13.
J Virol ; 79(12): 7544-57, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15919909

RESUMO

Dendritic cells are the most potent antigen-presenting cell for priming naive T cells. Optimal activation of T cells requires that dendritic cells undergo a process of maturation resulting in the increased expression of costimulatory molecules, such as CD40, CD86, and CD80, and the production of cytokines. In this study we analyzed the effect of infection of dendritic cells obtained from two strains of mice, BALB/c and C57BL/6, with the paramyxovirus simian virus 5 (SV5). Our results show that C57BL/6 bone marrow-derived dendritic cells (BMDC) are much more permissive to infection with SV5 at a multiplicity of infection (MOI) of 10 PFU/cell compared to BALB/c BMDC, as determined by the production of viral proteins and progeny. However, infection of BALB/c BMDC with a higher MOI of 50 PFU/cell resulted in a productive infection with the production of significant amounts of viral proteins and progeny. Regardless of the permissivity to infection, both BALB/c and C57BL/6 BMDC efficiently upregulated CD40 and CD86. However, CD80 upregulation correlated with the level of expression of viral proteins and the production of viral progeny. While secreted alpha/beta interferon was required for increased expression of all three molecules, optimal CD80 expression was dependent on an additional signal provided by a productive viral infection. These findings provide evidence that the signals controlling the expression of costimulatory molecules following viral infection are distinct. Further, they suggest that the amount of virus encountered and/or the permissivity of a dendritic cell to infection can alter the resulting maturation phenotype and functional capacity of the infected dendritic cell.


Assuntos
Antígenos CD/metabolismo , Antígeno B7-1/metabolismo , Antígenos CD40/metabolismo , Células Dendríticas/citologia , Células Dendríticas/virologia , Glicoproteínas de Membrana/metabolismo , Vírus da Parainfluenza 5/patogenicidade , Regulação para Cima , Animais , Antígeno B7-2 , Células da Medula Óssea/citologia , Diferenciação Celular , Células Dendríticas/imunologia , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Interferon beta/imunologia , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus da Parainfluenza 5/genética , Infecções por Rubulavirus/imunologia , Infecções por Rubulavirus/virologia , Especificidade da Espécie
14.
Virology ; 335(1): 131-44, 2005 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-15823612

RESUMO

A paramyxovirus SV5 mutant (rSV5-P/V-CPI-) that encodes 6 naturally-occurring P/V gene substitutions is a potent inducer of type I interferon (IFN) and is restricted for low moi growth, two phenotypes not seen with WT SV5. In this study, we have compared the IFN sensitivity of WT SV5 and the rSV5-P/V-CPI- mutant in tumor cell lines and in cultures of normal primary cells. We have tested the hypothesis that differences in IFN induction elicited by WT rSV5 and rSV5-P/V-CPI- are responsible for differences in low moi growth and spread. In contrast to WT SV5, low moi infection of A549 lung carcinoma cells with rSV5-P/V-CPI- resulted in a plateau of virus production by 24-48 h pi when secreted IFN levels were between approximately 100 and 1000 U/ml. Gene microarray and RT-PCR analyses identified IFN genes and IFN-stimulated genes whose expression were increased by infection of A549 cells with WT and P/V mutant viruses. Restricted low moi growth and spread of rSV5-P/V-CPI- in A549 cells was relieved in the presence of neutralizing antibodies to IFN-beta but not TNF-alpha. When A549 or MDA-MB-435 breast tumor cells were pretreated with IFN, both WT and P/V mutant viruses showed delayed spread and approximately 10-fold reduction in virus yield, but infections were not eliminated. Using normal primary human epithelial cells that have undergone limited passage in culture, WT rSV5 and rSV5-P/V-CPI- displayed high moi growth properties that were similar to that seen in A549 cells. However, IFN pretreatment of these primary cells as well as normal human lung cells eliminated low moi spread of both mutant and WT rSV5 infections. Together, these data demonstrate that SV5 growth in normal primary human cells is highly sensitive to IFN compared to growth in some tumor cell lines, regardless of whether the P/V gene is WT or mutant. These results suggest a model in which spread of WT SV5 in normal human cells is dependent on the ability of the virus to prevent IFN synthesis. The implications of these results for the use of recombinant paramyxoviruses as vectors are discussed.


Assuntos
Interferon Tipo I/farmacologia , Mutação , Vírus da Parainfluenza 5/crescimento & desenvolvimento , Fosfoproteínas/genética , Recombinação Genética , Proteínas Virais/genética , Proteínas Estruturais Virais/genética , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Interferon Tipo I/metabolismo , Vírus da Parainfluenza 5/efeitos dos fármacos , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/patogenicidade , Proteínas de Ligação a RNA
15.
J Virol ; 79(3): 1543-51, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15650180

RESUMO

Formation of a six-helix bundle comprised of three C-terminal heptad repeat regions in antiparallel orientation in the grooves of an N-terminal coiled-coil is critical for promotion of membrane fusion by paramyxovirus fusion (F) proteins. We have examined the effect of mutations in four residues of the N-terminal heptad repeat in the simian virus 5 (SV5) F protein on protein folding, transport, and fusogenic activity. The residues chosen have previously been shown from study of isolated peptides to have differing effects on stability of the N-terminal coiled-coil and six-helix bundle (R. E. Dutch, G. P. Leser, and R. A. Lamb, Virology 254:147-159, 1999). The mutant V154M showed reduced proteolytic cleavage and surface expression, indicating a defect in intracellular transport, though this mutation had no effect when studied in isolated peptides. The mutation I137M, previously shown to lower thermostability of the six-helix bundle, resulted in an F protein which was properly processed and transported to the cell surface but which had reduced fusogenic activity. Finally, mutations at L140M and L161M, previously shown to disrupt alpha-helix formation of isolated N-1 peptides but not to affect six-helix bundle formation, resulted in F proteins that were properly processed. Interestingly, the L161M mutant showed increased syncytium formation and promoted fusion at lower temperatures than the wild-type F protein. These results indicate that interactions separate from formation of an N-terminal coiled-coil or six-helix bundle are important in the initial folding and transport of the SV5 F protein and that mutations that destabilize the N-terminal coiled-coil can result in stimulation of membrane fusion.


Assuntos
Fusão de Membrana , Vírus da Parainfluenza 5/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células Gigantes , Células HeLa , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 5/patogenicidade , Conformação Proteica , Dobramento de Proteína , Proteínas Recombinantes de Fusão , Células Vero , Proteínas Virais de Fusão/genética
16.
J Virol ; 78(16): 8513-23, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15280460

RESUMO

SER virus, a paramyxovirus that is closely related to simian virus 5 (SV5), is unusual in that it fails to induce syncytium formation. The SER virus F protein has an unusually long cytoplasmic tail (CT), and it was previously observed that truncations or specific mutations of this domain result in enhanced syncytium formation. In addition to the long CT, the SER F protein has nine amino acid differences from the F protein of SV5. We previously observed only a partial suppression of fusion in a chimeric SV5 F protein with a CT derived from SER virus, indicating that these other amino acid differences between the SER and SV5 F proteins also play a role in regulating the fusion phenotype. To examine the effects of individual amino acid differences, we mutated the nine SER residues individually to the respective residues of the SV5 F protein. We found that most of the mutants were expressed well and were transported to the cell surface at levels comparable to that of the wild-type SER F protein. Many of the mutants showed enhanced lipid mixing, calcein transfer, and syncytium formation even in the presence of the long SER F protein CT. Some mutants, such as the I310 M, T438S, M489I, T516V, and N529K mutants, also showed fusion at lower temperatures of 32, 25, and 18 degrees C. The residue Asn529 plays a critical role in the suppression of fusion activity, as the mutation of this residue to lysine caused a marked enhancement of fusion. The effect of the N529K mutation on the enhancement of fusion by a previously described mutant, L539,548A, as well as by chimeric SV5/SER F proteins was also dramatic. These results indicate that activation to a fusogenic conformation is dependent on the interplay of residues in the ectodomain, the transmembrane domain, and the CT domain of paramyxovirus F proteins.


Assuntos
Fusão de Membrana/fisiologia , Mutação , Paramyxovirinae/patogenicidade , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Fusão Celular , Regulação Viral da Expressão Gênica , Células Gigantes/fisiologia , Cobaias , Células HeLa , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/metabolismo , Vírus da Parainfluenza 5/patogenicidade , Paramyxovirinae/genética , Paramyxovirinae/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais de Fusão/genética
17.
Virology ; 316(1): 41-54, 2003 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-14599789

RESUMO

Infection of human cells with the paramyxovirus simian virus 5 (SV5) results in minimal cytopathic effect, and host interferon (IFN) and apoptotic pathways are not activated. We have previously shown that an rSV5 containing six naturally occurring P/V gene substitutions (rSV5-P/V-CPI-) displays premature and elevated expression of viral RNA and protein. In addition, cells infected with rSV5-P/V-CPI- show induction of the IFN-beta promoter as well as activation of IFN signaling and apoptotic pathways. In this article, we have tested the hypothesis that rSV5-WT can supply trans-acting factors that prevent host cell antiviral responses induced by rSV5-P/V-CPI-. During coinfection of human A549 cells, rSV5-WT blocked cell rounding, loss of cell volume, and DNA fragmentation induced by rSV5-P/V-CPI-, three later events in the apoptotic pathway, but was not able to block the loss of mitochondrial membrane potential (DeltaPsi(m)), an early event in the cell death process. As expected, IFN signaling was blocked during coinfections, and this was attributed to the loss of STAT1 induced by the rSV5-WT V protein. Surprisingly, simultaneous infection with rSV5-WT could not suppress the activation of the IFN-beta promoter by rSV5-P/V-CPI- infection. However, the IFN-beta promoter was not activated in cells that were first preinfected for 1 h with rSV5-WT and then subsequently infected with rSV5-P/V-CPI-. A model is proposed for activation of host responses to infection with the rSV5-P/V-CPI- mutant and the steps that are blocked by rSV5-WT.


Assuntos
Apoptose , Interferon beta/metabolismo , Mutação , Vírus da Parainfluenza 5/patogenicidade , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Proteínas Virais/genética , Proteínas Estruturais Virais/genética , Linhagem Celular , Efeito Citopatogênico Viral , Humanos , Interferon-alfa/metabolismo , Interferon beta/genética , Potenciais da Membrana , Mitocôndrias , Vírus da Parainfluenza 5/genética , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA , Transdução de Sinais , Ativação Transcricional , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...