Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
2.
J Virol ; 86(10): 5829-43, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438539

RESUMO

Pneumonia virus of mice (PVM), a relative of human respiratory syncytial virus (RSV), causes respiratory disease in mice. There is serologic evidence suggesting widespread exposure of humans to PVM. To investigate replication in primates, African green monkeys (AGM) and rhesus macaques (n = 4) were inoculated with PVM by the respiratory route. Virus was shed intermittently at low levels by a subset of animals, suggesting poor permissiveness. PVM efficiently replicated in cultured human cells and inhibited the type I interferon (IFN) response in these cells. This suggests that poor replication in nonhuman primates was not due to a general nonpermissiveness of primate cells or poor control of the IFN response. Seroprevalence in humans was examined by screening sera from 30 adults and 17 young children for PVM-neutralizing activity. Sera from a single child (6%) and 40% of adults had low neutralizing activity against PVM, which could be consistent with increasing incidence of exposure following early childhood. There was no cross-reaction of human or AGM sera between RSV and PVM and no cross-protection in the mouse model. In native Western blots, human sera reacted with RSV but not PVM proteins under conditions in which AGM immune sera reacted strongly. Serum reactivity was further evaluated by flow cytometry using unfixed Vero cells infected with PVM or RSV expressing green fluorescent protein (GFP) as a measure of viral gene expression. The reactivity of human sera against RSV-infected cells correlated with GFP expression, whereas reactivity against PVM-infected cells was low and uncorrelated with GFP expression. Thus, PVM specificity was not evident. Our results indicate that the PVM-neutralizing activity of human sera is not due to RSV- or PVM-specific antibodies but may be due to low-affinity, polyreactive natural antibodies of the IgG subclass. The absence of PVM-specific antibodies and restriction in nonhuman primates makes PVM unlikely to be a human pathogen.


Assuntos
Vírus da Pneumonia Murina/fisiologia , Infecções por Pneumovirus/virologia , Replicação Viral , Adulto , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Pré-Escolar , Chlorocebus aethiops , Proteção Cruzada , Feminino , Humanos , Lactente , Macaca mulatta , Masculino , Camundongos , Vírus da Pneumonia Murina/imunologia , Vírus da Pneumonia Murina/patogenicidade , Infecções por Pneumovirus/imunologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/fisiologia , Adulto Jovem
3.
J Virol ; 85(19): 10135-43, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21795345

RESUMO

Susceptibility to respiratory syncytial virus (RSV) infection in mice is genetically determined. While RSV causes little pathology in C57BL/6 mice, pulmonary inflammation and weight loss occur in BALB/c mice. Using major histocompatibility complex (MHC)-congenic mice, we observed that the H-2(d) allele can partially transfer disease susceptibility to C57BL/6 mice. This was not explained by altered viral elimination or differences in the magnitude of the overall virus-specific cytotoxic T lymphocyte (CTL) response. However, H-2(d) mice showed a more focused response, with 70% of virus-specific CTL representing Vß8.2(+) CTL directed against the immunodominant epitope M2-1 82, while in H-2(b) mice only 20% of antiviral CTL were Vß9(+) CTL specific for the immunodominant epitope M187. The immunodominant H-2(d)-restricted CTL lysed target cells less efficiently than the immunodominant H-2(b) CTL, probably contributing to prolonged CTL stimulation and cytokine-mediated immunopathology. Accordingly, reduction of dominance of the M2-1 82-specific CTL population by introduction of an M187 response in the F1 generation of a C57BL/6N × C57BL/6-H-2(d) mating (C57BL/6-H-2(dxb) mice) attenuated disease. Moreover, disease in H-2(d) mice was less pronounced after infection with an RSV mutant failing to activate M2-1 82-specific CTL or after depletion of Vß8.2(+) cells. These data illustrate how the MHC-determined diversity and functional avidity of CTL responses contribute to disease susceptibility after viral infection.


Assuntos
Suscetibilidade a Doenças , Antígenos H-2/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Antígenos H-2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sinciciais Respiratórios/patogenicidade , Doenças dos Roedores/imunologia , Doenças dos Roedores/patologia
4.
J Virol ; 85(9): 4071-84, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21307191

RESUMO

Infection of mice with pneumonia virus of mice (PVM) provides a convenient experimental pathogenesis model in a natural host for a human respiratory syncytial virus-related virus. Extending our previous work showing that the PVM nonstructural (NS) proteins were pathogenicity factors in mice, we identify both the NS1 and NS2 proteins as antagonists of alpha/beta interferon (IFN-α/ß) and IFN-λ by use of recombinant PVM (rPVM) with single and combined deletions of the NS proteins (ΔNS1, ΔNS2, and ΔNS1 ΔNS2). Wild-type and NS deletion PVMs were evaluated for growth and pathogenesis by infecting knockout mice that lack functional receptors to IFN-α/ß, IFN-λ, or both. The absence of the receptor to IFN-α/ß (IFNAR) or IFN-λ (interleukin-28 receptor α chain [IL-28Rα]) individually did not reverse the attenuated virulence of the NS deletion viruses although loss of IFNAR partially restored replication efficiency. When both receptors were deleted, replication and virulence were largely rescued for rPVM ΔNS1 and were significantly but not completely rescued for rPVM ΔNS2. As for rPVM ΔNS1 ΔNS2, the effect was mostly limited to partial enhancement of replication. This indicates that both IFN-α/ß and IFN-λ contributed to restricting the NS deletion viruses, with the former playing the greater role. Interestingly, the replication and virulence of wild-type PVM were completely unaffected by the presence or absence of functional receptors to IFN-α/ß and IFN-λ, indicating that both systems are strongly suppressed during infection. However, pretreatment of mice with IFN-α/ß was protective against lethal rPVM challenge, whereas pretreatment with IFN-λ delayed but did not prevent disease and, in some cases, reduced mortality. The fact that virulence of rPVM lacking NS2 was not recovered completely when both interferon receptors were deleted suggests that NS2 may have further functions outside the IFN system.


Assuntos
Citocinas/antagonistas & inibidores , Interferon Tipo I/antagonistas & inibidores , Vírus da Pneumonia Murina/imunologia , Infecções por Pneumovirus/veterinária , Doenças dos Roedores/virologia , Proteínas não Estruturais Virais/imunologia , Fatores de Virulência/imunologia , Animais , Deleção de Genes , Histocitoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia , Vírus da Pneumonia Murina/patogenicidade , Infecções por Pneumovirus/patologia , Infecções por Pneumovirus/virologia , Doenças dos Roedores/patologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Replicação Viral
5.
Nat Med ; 16(12): 1434-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21102458

RESUMO

Danger signals released upon cell damage can cause excessive immune-mediated tissue destruction such as that found in acute graft-versus-host disease (GVHD), allograft rejection and systemic inflammatory response syndrome. Given that ATP is found in small concentrations in the extracellular space under physiological conditions, and its receptor P2X(7)R is expressed on several immune cell types, ATP could function as a danger signal when released from dying cells. We observed increased ATP concentrations in the peritoneal fluid after total body irradiation, and during the development of GVHD in mice and in humans. Stimulation of antigen-presenting cells (APCs) with ATP led to increased expression of CD80 and CD86 in vitro and in vivo and actuated a cascade of proinflammatory events, including signal transducer and activator of transcription-1 (STAT1) phosphorylation, interferon-γ (IFN-γ) production and donor T cell expansion, whereas regulatory T cell numbers were reduced. P2X(7)R expression increased when GVHD evolved, rendering APCs more responsive to the detrimental effects of ATP, thereby providing positive feedback signals. ATP neutralization, early P2X(7)R blockade or genetic deficiency of P2X(7)R during GVHD development improved survival without immune paralysis. These data have major implications for transplantation medicine, as pharmacological interference with danger signals that act via P2X(7)R could lead to the development of tolerance without the need for intensive immunosuppression.


Assuntos
Trifosfato de Adenosina/metabolismo , Espaço Extracelular/metabolismo , Doença Enxerto-Hospedeiro/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animais , Células Apresentadoras de Antígenos/metabolismo , Ascite/metabolismo , Líquido Ascítico/metabolismo , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Transplante de Medula Óssea , Citocinas/imunologia , Citometria de Fluxo , Trato Gastrointestinal/metabolismo , Humanos , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Fosforilação , Receptores Purinérgicos P2X7/genética , Fator de Transcrição STAT1/metabolismo , Linfócitos T Reguladores/imunologia , Irradiação Corporal Total
6.
J Virol ; 83(4): 1969-80, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19052095

RESUMO

Pneumonia virus of mice (PVM) strain 15 causes fatal pneumonia in mice and provides a convenient model for human respiratory syncytial virus pathogenesis and immunobiology. We prepared PVM mutants lacking the genes for nonstructural proteins NS1 and/or NS2. In Vero cells, which lack type I interferon (IFN), deletion of these proteins had no effect on the efficiency of virus growth. In IFN-competent mouse embryo fibroblasts, wild-type (wt) PVM and the DeltaNS1 virus grew efficiently and strongly inhibited the IFN response, whereas virus lacking NS2 was highly attenuated and induced high levels of IFN and IFN-inducible genes. In BALB/c mice, intranasal infection with wt PVM caused overt disease that began on day 6 and was lethal by day 9 postinoculation. In comparison, DeltaNS1 induced transient, reduced disease, and DeltaNS2 and DeltaNS12 caused no disease. Thus, NS1 and NS2 are virulence factors, with NS2 being a major antagonist of the type I IFN system. The pulmonary titers of wt PVM and DeltaNS1 were high on day 3 and increased further by day 6; in addition, expression of IFN and representative proinflammatory cytokines/chemokines and T lymphocyte-related cytokines was undetectable on day 3 but increased dramatically by day 6 coincident with the onset of disease. The titers of DeltaNS2 and DeltaNS12 were somewhat lower on day 3 and decreased further by day 6; in addition, these viruses induced a more circumscribed set of cytokines/chemokines (IFN, interleukin-6 [IL-6], and CXCL10) that were detected on day 3 and had largely subsided by day 6. Lung immunohistology revealed abundant PVM-positive pneumocytes and bronchial and bronchiolar epithelial cells in wt PVM- and DeltaNS1-infected mice on day 6 compared to few PVM-positive foci with DeltaNS2 and DeltaNS12. These results indicate that severe PVM disease is associated with high, poorly controlled virus replication driving the expression of high levels of pulmonary IFN and a broad array of cytokines/chemokines. In contrast, in the absence of NS2, there was an early, transient innate response involving moderate levels of IFN, IL-6, and CXCL10 that restricted virus replication and prevented disease.


Assuntos
Citocinas/biossíntese , Pulmão/patologia , Vírus da Pneumonia Murina/fisiologia , Vírus da Pneumonia Murina/patogenicidade , Proteínas não Estruturais Virais/fisiologia , Fatores de Virulência/fisiologia , Replicação Viral , Animais , Peso Corporal , Linhagem Celular , Citocinas/imunologia , Deleção de Genes , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Pneumonia Murina/genética , Vírus da Pneumonia Murina/imunologia , Índice de Gravidade de Doença , Análise de Sobrevida , Proteínas não Estruturais Virais/genética , Virulência , Fatores de Virulência/genética
7.
J Virol ; 82(23): 11619-27, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18815308

RESUMO

Infection of mice with pneumonia virus of mice (PVM) is used as a natural host experimental model for studying the pathogenesis of infection with the closely related human respiratory syncytial virus. We analyzed the contribution of T cells to virus control and pathology after PVM infection. Control of a sublethal infection with PVM strain 15 in C57BL/6 mice was accompanied by a 100-fold increase in pulmonary cytotoxic T lymphocytes, 20% of which were specific for PVM. T-cell-deficient mice failed to eliminate PVM and became virus carriers in the absence of the clinical or histopathological signs of pneumonia that occurred after infection of control mice. Mice with limited T-cell numbers did not achieve virus control without weight loss, indicating that T-cell-mediated virus control was closely linked to immunopathology. Both CD4 and CD8 T cells independently contributed to virus elimination and disease. Virus control and disease were similar in the absence of perforin, gamma interferon, or tumor necrosis factor alpha. Interestingly, disease and mortality after lethal high-dose PVM infection were independent of T cells. These data illustrate a key role for T cells in control of PVM infection and demonstrate that both T-cell-dependent and -independent pathways contribute to disease in a viral dose-dependent fashion.


Assuntos
Vírus da Pneumonia Murina , Infecções por Pneumovirus/imunologia , Linfócitos T/fisiologia , Animais , Interferon gama/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Perforina/fisiologia , Infecções por Pneumovirus/patologia , Receptores de Antígenos de Linfócitos T/fisiologia , Linfócitos T Citotóxicos/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Replicação Viral , Redução de Peso
8.
J Immunol ; 179(12): 8264-73, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18056370

RESUMO

CTL are important for virus clearance but also contribute to immunopathology after the infection of BALB/c mice with respiratory syncytial virus (RSV). The pulmonary immune response to RSV is dominated by a CTL population directed against the CTL epitope M2-1 82-90. Infection with a virus carrying an M2-1 N89A mutation introduced by reverse genetics failed to activate this immunodominant CTL population, leading to a significant decrease in the overall antiviral CTL response. There was no compensatory increase in responses to the mutated epitope, to the subdominant epitope F 85-93, or to yet undefined minor epitopes in the N or the P protein. However, there was some increase in the response to the subdominant epitope M2-1 127-135, which is located in the same protein and presented by the same H-2Kd MHC molecule. Infection with the mutant virus reversed the oligoclonality of the T cell response elicited by the wild-type virus. These changes in the pattern and composition of the antiviral CTL response only slightly impaired virus clearance but significantly reduced RSV-induced weight loss. These data illustrate how T cell epitope mutations can influence the virus-host relationship and determine disease after an acute respiratory virus infection.


Assuntos
Epitopos de Linfócito T/imunologia , Epitopos Imunodominantes/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Linfócitos T Citotóxicos/imunologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Epitopos de Linfócito T/genética , Epitopos Imunodominantes/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação , Oligopeptídeos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Infecções por Vírus Respiratório Sincicial/genética
9.
J Virol ; 81(17): 9490-501, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17567693

RESUMO

Pneumonia virus of mice (PVM) is a murine relative of human respiratory syncytial virus (HRSV). Here we developed a reverse genetics system for PVM based on a consensus sequence for virulent strain 15. Recombinant PVM and a version engineered to express green fluorescent protein replicated as efficiently as the biological parent in vitro but were 4- and 12.5-fold attenuated in vivo, respectively. The G proteins of HRSV and PVM have been suggested to contribute to viral pathogenesis, but this had not been possible to study in a defined manner in a fully permissive host. As a first step, we evaluated recombinant mutants bearing a deletion of the entire G gene (Delta G) or expressing a G protein lacking its cytoplasmic tail (Gt). Both G mutants replicated as efficiently in vitro as their recombinant parent, but both were nonpathogenic in mice at doses that would otherwise be lethal. We could not detect replication of the Delta G mutant in mice, indicating that its attenuation is based on a severe reduction in the virus load. In contrast, the Gt mutant appeared to replicate as efficiently in mice as its recombinant parent. Thus, the reduction in virulence associated with the Gt mutant could not be accounted for by a reduction in viral replication. These results identified the cytoplasmic tail of G as a virulence factor whose effect is not mediated solely by the viral load. In addition to its intrinsic interest, a recombinant virus that replicates with wild-type-like efficiency but does not cause disease defines optimal properties for vaccine development.


Assuntos
Vírus da Pneumonia Murina/patogenicidade , Fatores de Virulência/genética , Animais , Linhagem Celular , Cricetinae , Deleção de Genes , Genes Reporter , Engenharia Genética , Glicoproteínas/genética , Glicoproteínas/fisiologia , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Pneumonia Murina/genética , Vírus da Pneumonia Murina/fisiologia , Recombinação Genética , Deleção de Sequência , Análise de Sobrevida , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Virulência/fisiologia , Replicação Viral
10.
Virus Genes ; 30(2): 237-49, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15744580

RESUMO

Pneumonia virus of mice (PVM) is an enveloped RNA-containing virus of Family Paramyxoviridae. Sequences had been determined previously for a number of PVM genes, although these represented cloned cDNAs rather than consensus sequences. Sequences were not available for the 3' -leader and 5' -trailer regions that constitute the genome termini or for the large polymerase L gene that accounts for 43% of the genome. Also, the available sequences were from an attenuated variant of strain 15, whereas the present study analyzed the version of strain 15 that is available from the American Type Culture Collection (ATCC) and is highly virulent in mice. Analysis of unclosed RT-PCR products yielded a complete consensus sequence of 14,886 nt (GenBank accession number AY729016). Of the regions for which sequences had been previously reported for the non-pathogenic strain, there were 13 nucleotide differences and 10 amino acid differences compared to the present consensus sequence for the virulent isolate. The various genes of PVM shared 29-62% nucleotide sequence identity and 10-60% amino acid sequence identity with human or bovine respiratory syncytial virus (HRSV and BRSV), its closest relatives.


Assuntos
Genoma Viral , Vírus da Pneumonia Murina/genética , RNA Viral/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Mapeamento Cromossômico , Sequência Consenso , DNA Viral/genética , Humanos , Camundongos , Dados de Sequência Molecular , Vírus da Pneumonia Murina/patogenicidade , Vírus Sincicial Respiratório Bovino/genética , Vírus Sincicial Respiratório Humano/genética , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Proteínas Virais/genética
11.
J Virol ; 78(23): 13362-5, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15542688

RESUMO

Prototypic strain 15 of pneumonia virus of mice (PVM) has been described as being nonpathogenic in mice, in contrast to the mouse-passaged, highly virulent strain J3666. Previous sequence analysis also indicated that strain 15 encodes an attachment G protein that is truncated at the amino terminus, which for the amino terminally anchored protein deletes the cytoplasmic tail. However, we found that PVM strain 15 obtained from the American Type Culture Collection was highly virulent in mice and was essentially indistinguishable on that basis from strain J3666. Sequence analysis showed that this preparation of virus encodes a G protein with an intact cytoplasmic tail: the truncated predicted protein in the previous sequence appeared to be due to a single nucleotide insertion that disrupted the upstream end of the open reading frame and shifted the translational start site to the next downstream AUG. Taken together, the two studies indicate that strain 15 is an inherently virulent strain but that a nonpathogenic variant that was generated during passage in vitro and encodes a truncated G protein exists. Interestingly, the majority sequence of strain J3666 was found to encode a G protein with an extended cytoplasmic tail, suggesting that there is the potential for considerable plasticity in the cytoplasmic tail of the G protein of PVM.


Assuntos
Vírus da Pneumonia Murina/patogenicidade , Animais , Glicoproteínas/genética , Glicoproteínas/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA