Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
PLoS One ; 8(9): e75346, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058676

RESUMO

Persistent infection of the mouse central nervous system (CNS) with mouse hepatitis virus (MHV) induces a demyelinating disease pathologically similar to multiple sclerosis and is therefore used as a model system. There is little information regarding the host factors that correlate with and contribute to MHV-induced demyelination. Here, we detail the genes and pathways associated with MHV-induced demyelinating disease in the spinal cord. High-throughput sequencing of the host transcriptome revealed that demyelination is accompanied by numerous transcriptional changes indicative of immune infiltration as well as changes in the cytokine milieu and lipid metabolism. We found evidence that a Th1-biased cytokine/chemokine response and eicosanoid-derived inflammation accompany persistent MHV infection and that antigen presentation is ongoing. Interestingly, increased expression of genes involved in lipid transport, processing, and catabolism, including some with known roles in neurodegenerative diseases, coincided with demyelination. Lastly, expression of several genes involved in osteoclast or bone-resident macrophage function, most notably TREM2 and DAP12, was upregulated in persistently infected mouse spinal cord. This study highlights the complexity of the host antiviral response, which accompany MHV-induced demyelination, and further supports previous findings that MHV-induced demyelination is immune-mediated. Interestingly, these data suggest a parallel between bone reabsorption by osteoclasts and myelin debris clearance by microglia in the bone and the CNS, respectively. To our knowledge, this is the first report of using an RNA-seq approach to study the host CNS response to persistent viral infection.


Assuntos
Infecções por Coronavirus/metabolismo , Doenças Desmielinizantes/metabolismo , Vírus da Hepatite Murina/metabolismo , Medula Espinal/metabolismo , Transcriptoma , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Infecções por Coronavirus/patologia , Citocinas/biossíntese , Citocinas/metabolismo , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Feminino , Masculino , Glicoproteínas de Membrana/biossíntese , Camundongos , Proteínas do Tecido Nervoso/biossíntese , Receptores Imunológicos/biossíntese , Medula Espinal/patologia , Células Th1/metabolismo , Células Th1/patologia
2.
J Virol ; 84(18): 9278-91, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20631137

RESUMO

We report here investigation into the genetic basis of mouse hepatitis virus strain 1 (MHV-1) pneumovirulence. Sequencing of the 3' one-third of the MHV-1 genome demonstrated that the genetic organization of MHV-1 was similar to that of other strains of MHV. The hemagglutinin esterase (HE) protein was truncated, and reverse transcription-PCR (RT-PCR) studies confirmed previous work that suggested that the MHV-1 HE is a pseudogene. Targeted recombination was used to select chimeric viruses containing either the MHV-1 S gene or genes encoding all of the MHV-1 structural proteins, on an MHV-A59 background. Challenge studies in mice demonstrated that expression of the MHV-1 S gene within the MHV-A59 background (rA59/S(MHV-1)) increased the pneumovirulence of MHV-A59, and mice infected with this recombinant virus developed pulmonary lesions that were similar to those observed with MHV-1, although rA59/S(MHV-1) was significantly less virulent. Chimeras containing all of the MHV-1 structural genes on an MHV-A59 background were able to reproduce the severe acute respiratory syndrome (SARS)-like pathology observed with MHV-1 and reproducibly increased pneumovirulence relative to rA59/S(MHV-1), but were still much less virulent than MHV-1. These data suggest that important determinants of pneumopathogenicity are contained within the 3' one-third of the MHV-1 genome, but additional important virulence factors must be encoded in the genome upstream of the S gene. The severity of the pulmonary lesions observed correlates better with elevated levels of inflammatory cytokines than with viral replication in the lungs, suggesting that pulmonary disease has an important immunological component.


Assuntos
Pulmão/patologia , Pulmão/virologia , Glicoproteínas de Membrana/fisiologia , Vírus da Hepatite Murina/patogenicidade , Proteínas do Envelope Viral/fisiologia , Fatores de Virulência/fisiologia , Animais , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Feminino , Ordem dos Genes , Genes Virais , Glicoproteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/genética , Pneumonia Viral/patologia , Pneumonia Viral/virologia , RNA Viral/química , RNA Viral/genética , Recombinação Genética , Análise de Sequência de DNA , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/genética , Fatores de Virulência/genética
3.
J Virol ; 83(8): 3743-53, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19176619

RESUMO

The Murine hepatitis virus (MHV) strain A59 ns2 protein is a 30-kDa nonstructural protein that is expressed from a subgenomic mRNA in the cytoplasm of virus-infected cells. Its homologs are also encoded in other closely related group 2a coronaviruses and more distantly related toroviruses. Together, these proteins comprise a subset of a large superfamily of 2H phosphoesterase proteins that are distinguished by a pair of conserved His-x-Thr/Ser motifs encompassing catalytically important residues. We have used a vaccinia virus-based reverse genetic system to produce recombinant viruses encoding ns2 proteins with single-amino-acid substitutions in, or adjacent to, these conserved motifs, namely, inf-ns2 H46A, inf-ns2 S48A, inf-ns2-S120A, and inf-ns2-H126R. All of the mutant viruses replicate in mouse 17 clone 1 fibroblast cells and mouse embryonic cells to the same extent as the parental wild-type recombinant virus, inf-MHV-A59. However, compared to inf-MHV-A59, the inf-ns2 H46A and inf-ns2-H126R mutants are highly attenuated for replication in mouse liver following intrahepatic inoculation. Interestingly, none of the mutant viruses were attenuated for replication in mouse brain following intracranial inoculation. These results show that the ns2 protein of MHV-A59 has an important role in virus pathogenicity and that a substitution of the histidine residues of the MHV-A59 ns2 His-x-Thr/Ser motifs is critical for virus virulence in the liver but not in the brain. This novel phenotype suggests a strategy to investigate the function of the MHV-A59 ns2 protein involving the search for organ-specific proteins or RNAs that react differentially to wild-type and mutant ns2 proteins.


Assuntos
Vírus da Hepatite Murina/enzimologia , Vírus da Hepatite Murina/patogenicidade , Diester Fosfórico Hidrolases/genética , Substituição de Aminoácidos/genética , Animais , Encéfalo/virologia , Células Cultivadas , Infecções por Coronavirus/virologia , Fibroblastos/virologia , Fígado/virologia , Camundongos , Vírus da Hepatite Murina/crescimento & desenvolvimento , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Diester Fosfórico Hidrolases/metabolismo
4.
J Virol ; 82(13): 6150-60, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18417581

RESUMO

Virus-specific CD8(+) T cells are critical for protection against neurotropic coronaviruses; however, central nervous system (CNS) infection with the recombinant JHM (RJHM) strain of mouse hepatitis virus (MHV) elicits a weak CD8(+) T-cell response in the brain and causes lethal encephalomyelitis. An adoptive transfer model was used to elucidate the kinetics of CD8(+) T-cell priming during CNS infection with RJHM as well as with two MHV strains that induce a robust CD8(+) T-cell response (RA59 and SJHM/RA59, a recombinant A59 virus expressing the JHM spike). While RA59 and SJHM/RA59 infections resulted in CD8(+) T-cell priming within the first 2 days postinfection, RJHM infection did not lead to proliferation of naïve CD8(+) T cells. While all three viruses replicated efficiently in the brain, only RA59 and SJHM/RA59 replicated to appreciable levels in the cervical lymph nodes (CLN), the site of T-cell priming during acute CNS infection. RJHM was unable to suppress the CD8(+) T-cell response elicited by RA59 in mice simultaneously infected with both strains, suggesting that RJHM does not cause generalized immunosuppression. RJHM was also unable to elicit a secondary CD8(+) T-cell response in the brain following peripheral immunization against a viral epitope. Notably, the weak CD8(+) T-cell response elicited by RJHM was unique to CNS infection, since peripheral inoculation induced a robust CD8(+) T-cell response in the spleen. These findings suggest that the failure of RJHM to prime a robust CD8(+) T-cell response during CNS infection is likely due to its failure to replicate in the CLN.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Sistema Nervoso Central/imunologia , Infecções por Coronavirus/imunologia , Vírus da Hepatite Murina/imunologia , Transferência Adotiva , Animais , Sistema Nervoso Central/virologia , Citometria de Fluxo , Linfonodos/imunologia , Camundongos , Especificidade da Espécie
5.
J Virol ; 81(2): 1022-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17079303

RESUMO

The important roles of the spike protein and other structural proteins in murine coronavirus (MHV) pathogenesis have been demonstrated; however, the role of the replicase gene remains unexplored. We assessed the influence of the replicase genes of the highly neurovirulent MHV-JHM strain and the hepatotropic and mildly neurovirulent A59 strain in acute infection of the mouse. Analysis of chimeric A59/JHM recombinant viruses indicates that the replicase genes are interchangeable and that it is the 3' end of the genome, encoding the structural proteins, rather than the replicase gene, that determines the pathogenic properties of these chimeras.


Assuntos
Regiões 3' não Traduzidas/genética , Genoma Viral , Vírus da Hepatite Murina/patogenicidade , RNA Polimerase Dependente de RNA/genética , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Encefalite Viral/patologia , Encefalite Viral/virologia , Hepatite Viral Animal/patologia , Hepatite Viral Animal/virologia , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Recombinação Genética
6.
J Virol ; 79(14): 9108-18, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15994805

RESUMO

The immunodominant CD8+ T-cell epitope of a highly neurovirulent strain of mouse hepatitis virus (MHV), JHM, is thought to be essential for protection against virus persistence within the central nervous system. To test whether abrogation of this H-2Db-restricted epitope, located within the spike glycoprotein at residues S510 to 518 (S510), resulted in delayed virus clearance and/or virus persistence we selected isogenic recombinants which express either the wild-type JHM spike protein (RJHM) or spike containing the N514S mutation (RJHM(N514S)), which abrogates the response to S510. In contrast to observations in suckling mice in which viruses encoding inactivating mutations within the S510 epitope (epitope escape mutants) were associated with persistent virus and increased neurovirulence (Pewe et al., J Virol. 72:5912-5918, 1998), RJHM(N514S) was not more virulent than the parental, RJHM, in 4-week-old C57BL/6 (H-2b) mice after intracranial injection. Recombinant viruses expressing the JHM spike, wild type or encoding the N514S substitution, were also selected in which background genes were derived from the neuroattenuated A59 strain of MHV. Whereas recombinants expressing the wild-type JHM spike (SJHM/RA59) were highly neurovirulent, A59 recombinants containing the N514S mutation (SJHM(N514S)/RA59) were attenuated, replicated less efficiently, and exhibited reduced virus spread in the brain at 5 days postinfection (peak of infectious virus titers in the central nervous system) compared to parental virus encoding wild-type spike. Virulence assays in BALB/c mice (H-2d), which do not recognize the S510 epitope, revealed that attenuation of the epitope escape mutants was not due to the loss of a pathogenic immune response directed against the S510 epitope. Thus, an intact immunodominant S510 epitope is not essential for virus clearance from the CNS, the S510 inactivating mutation results in decreased virulence in weanling mice but not in suckling mice, suggesting that specific host conditions are required for epitope escape mutants to display increased virulence, and the N514S mutation causes increased attenuation in the context of A59 background genes, demonstrating that genes other than that for the spike are also important in determining neurovirulence.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T , Epitopos Imunodominantes , Vírus da Hepatite Murina/patogenicidade , Substituição de Aminoácidos , Animais , Antígenos Virais/análise , Encéfalo/virologia , Doenças Desmielinizantes/etiologia , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/imunologia , Virulência , Replicação Viral
7.
J Virol ; 79(6): 3370-81, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15731231

RESUMO

CD8+ T cells are important for clearance of neurotropic mouse hepatitis virus (MHV) strain A59, although their possible role in A59-induced demyelination is not well understood. We developed an adoptive-transfer model to more clearly elucidate the role of virus-specific CD8+ T cells during the acute and chronic phases of infection with A59 that is described as follows. C57BL/6 mice were infected with a recombinant A59 virus expressing the gp33 epitope, an H-2Db-restricted CD8+ T-cell epitope encoded in the glycoprotein of lymphocytic choriomeningitis virus, as a fusion with the enhanced green fluorescent protein (RA59-gfp/gp33). P14 splenocytes (transgenic for a T-cell receptor specific for the gp33 epitope) were transferred at different times pre- and postinfection (p.i.). Adoptive transfer of P14 splenocytes 1 day prior to infection with RA59-gfp/gp33, but not control virus lacking the gp33 epitope, RA59-gfp, reduced weight loss and viral replication and spread in the brain and to the spinal cord. Furthermore, demyelination was significantly reduced compared to that in nonrecipients. However, when P14 cells were transferred on day 3 or 5 p.i., no difference in acute or chronic disease was observed compared to that in nonrecipients. Protection in mice receiving P14 splenocytes prior to infection correlated with a robust gp33-specific immune response that was not observed in mice receiving the later transfers. Thus, an early robust CD8+ T-cell response was necessary to reduce virus replication and spread, specifically to the spinal cord, which protected against demyelination in the chronic phase of the disease.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Coronavirus/imunologia , Coronavirus/imunologia , Doenças Desmielinizantes/prevenção & controle , Epitopos de Linfócito T/imunologia , Medula Espinal/virologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Coronavirus/crescimento & desenvolvimento , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Medula Espinal/patologia , Ensaio de Placa Viral
8.
J Virol ; 78(3): 1150-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14722270

RESUMO

Both CD4(+) and CD8(+) T cells are required for clearance of the murine coronavirus mouse hepatitis virus (MHV) during acute infection. We investigated the effects of an epitope-specific CD8(+) T-cell response on acute infection of MHV, strain A59, in the murine CNS. Mice with CD8(+) T cells specific for gp33-41 (an H-2D(b)-restricted CD8(+) T-cell epitope derived from lymphocytic choriomeningitis glycoprotein) were infected with a recombinant MHV-A59, also expressing gp33-41, as a fusion protein with enhanced green fluorescent protein (EGFP). By 5 days postinfection, these mice showed significantly (approximately 20-fold) lower titers of infectious virus in the brain compared to control mice. Furthermore mice with gp33-41-specific CD8(+) cells exhibited much reduced levels of viral antigen in the brain as measured by immunohistochemistry using an antibody directed against viral nucleocapsid. More than 90% of the viruses recovered from brain lysates of such protected mice, at 5 days postinfection, had lost the ability to express EGFP and had deletions in their genomes encompassing EGFP and gp33-41. In addition, genomes of viruses from about half the plaques that retained the EGFP gene had mutations within the gp33-41 epitope. On the other hand, gp33-41-specific cells failed to protect perforin-deficient mice from infection by the recombinant MHV expressing gp33, indicating that perforin-mediated mechanisms were needed. Virus recovered from perforin-deficient mice did not exhibit loss of EGFP expression and the gp33-41 epitope. These observations suggest that the cytotoxic T-cell response to gp33-41 exerts a strong immune pressure that quickly selects epitope escape mutants to gp33-41.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Viroses do Sistema Nervoso Central/imunologia , Epitopos/imunologia , Glicoproteínas/imunologia , Vírus da Hepatite Murina/imunologia , Fragmentos de Peptídeos/imunologia , Proteínas Virais/imunologia , Animais , Antígenos Virais/metabolismo , Viroses do Sistema Nervoso Central/prevenção & controle , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Epitopos/genética , Proteínas de Fluorescência Verde , Humanos , Imunização , Proteínas Luminescentes/genética , Proteínas Luminescentes/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/patogenicidade , Mutação , Perforina , Proteínas Citotóxicas Formadoras de Poros , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Replicação Viral
9.
Virology ; 301(1): 109-20, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12359451

RESUMO

The mouse hepatitis virus (MHV) spike glycoprotein is a major determinant of neurovirulence. We investigated how alterations in spike affect neurovirulence using two isogenic recombinant viruses differing exclusively in spike. S(4)R, containing the MHV-4 spike gene, is dramatically more neurovirulent than S(A59)R, containing the MHV-A59 spike gene (J. J. Phillips, M. M. Chua, E. Lavi, and S. R. Weiss, 1999, J. Virol. 73, 7752-7760). We examined the contribution of differences in cellular tropism, viral spread, and the immune response to infection to the differential neurovirulence of S(4)R and S(A59)R. MHV-4 spike-mediated neurovirulence was associated with extensive viral spread in the brain in both neurons and astrocytes. Infection of primary hippocampal neuron cultures demonstrated that S(4)R spread more rapidly than S(A59)R and suggested that spread may occur between cells in close physical contact. In addition, S(4)R infection induced a massive influx of lymphocytes into the brain, a higher percentage of CD8(+) T cells, and a higher frequency of MHV-specific CD8(+) T cells relative S(A59)R infection. Despite this robust and viral-specific immune response to S(4)R infection, infection of RAG1-/- mice suggested that immune-mediated pathology also contributes to the high neurovirulence of S(4)R.


Assuntos
Viroses do Sistema Nervoso Central/etiologia , Inflamação/etiologia , Glicoproteínas de Membrana/fisiologia , Vírus da Hepatite Murina/patogenicidade , Proteínas do Envelope Viral/fisiologia , Animais , Células Cultivadas , Viroses do Sistema Nervoso Central/imunologia , Citometria de Fluxo , Interferon gama/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/virologia , Glicoproteína da Espícula de Coronavírus , Linfócitos T/imunologia , Virulência
10.
J Immunol ; 169(9): 5202-8, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391238

RESUMO

The immune system has evolved various effector cells and functions to combat diverse infectious agents equipped with different virulence strategies. CD8 T cells play a critical role in protective immunity to Listeria monocytogenes (Lm), a bacterium that grows within the host cell cytosol and spreads directly into neighboring cells. The importance of CD8 T cells during Lm infection is currently attributed to the cytosolic niche of this organism, which allows it to evade many aspects of immune surveillance. CTL lysis of infected cells is believed to be an essential protective mechanism, presumably functioning to release intracellular bacteria, although its precise role remains to be fully defined. In this study, we examined the contribution of perforin-mediated CTL cytolysis to protective immunity against recombinant Lm capable of or defective in cell-cell spread. We found that CTL cytolysis is critical for protective immunity to Lm capable of cell-cell spread while protective immunity against spread-defective Lm is largely independent of CTL cytolysis. These results demonstrate that an important function of CTL cytolysis is to counter the microbial virulence strategy of direct cell-cell spread. We propose a model that advances the current view of the role of CTL cytolysis in immunity to intracellular pathogens.


Assuntos
Citotoxicidade Imunológica , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Glicoproteínas de Membrana/fisiologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/microbiologia , Animais , Antígenos Virais/imunologia , Citotoxicidade Imunológica/genética , Epitopos de Linfócito T/imunologia , Glicoproteínas/imunologia , Imunidade Celular/genética , Memória Imunológica/genética , Listeria monocytogenes/genética , Listeriose/genética , Listeriose/imunologia , Listeriose/microbiologia , Vírus da Coriomeningite Linfocítica/imunologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/imunologia , Perforina , Proteínas Citotóxicas Formadoras de Poros , Recombinação Genética , Linfócitos T Citotóxicos/virologia , Proteínas Virais/imunologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA