Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 21(12): 1464-72, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26523970

RESUMO

The TAM receptors Tyro3, Axl and Mertk are receptor tyrosine kinases that dampen host innate immune responses following engagement with their ligands Gas6 and Protein S, which recognize phosphatidylserine on apoptotic cells. In a form of apoptotic mimicry, many enveloped viruses display phosphatidylserine on the outer leaflet of their membranes, enabling TAM receptor activation and downregulation of antiviral responses. Accordingly, we hypothesized that a deficiency of TAM receptors would enhance antiviral responses and protect against viral infection. Unexpectedly, mice lacking Mertk and/or Axl, but not Tyro3, exhibited greater vulnerability to infection with neuroinvasive West Nile and La Crosse encephalitis viruses. This phenotype was associated with increased blood-brain barrier permeability, which enhanced virus entry into and infection of the brain. Activation of Mertk synergized with interferon-ß to tighten cell junctions and prevent virus transit across brain microvascular endothelial cells. Because TAM receptors restrict pathogenesis of neuroinvasive viruses, these findings have implications for TAM antagonists that are currently in clinical development.


Assuntos
Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/virologia , Encefalite da Califórnia/enzimologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Febre do Nilo Ocidental/enzimologia , Imunidade Adaptativa , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/patologia , Quimiocinas/sangue , Encefalite da Califórnia/patologia , Encefalite da Califórnia/virologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Interferon beta/metabolismo , Vírus La Crosse/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvasos/patologia , Permeabilidade , Substâncias Protetoras , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/deficiência , Tolerância a Radiação , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/deficiência , Transdução de Sinais , Análise de Sobrevida , Carga Viral , Febre do Nilo Ocidental/patologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/fisiologia , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
3.
J Virol ; 88(19): 11007-21, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25031348

RESUMO

UNLABELLED: Upon activation of Toll-like and RIG-I-like receptor signaling pathways, the transcription factor IRF5 translocates to the nucleus and induces antiviral immune programs. The recent discovery of a homozygous mutation in the immunoregulatory gene guanine exchange factor dedicator of cytokinesis 2 (Dock2mu/mu) in several Irf5-/- mouse colonies has complicated interpretation of immune functions previously ascribed to IRF5. To define the antiviral functions of IRF5 in vivo, we infected backcrossed Irf5-/-×Dock2wt/wt mice (here called Irf5-/- mice) and independently generated CMV-Cre Irf5fl/fl mice with West Nile virus (WNV), a pathogenic neurotropic flavivirus. Compared to congenic wild-type animals, Irf5-/- and CMV-Cre Irf5fl/fl mice were more vulnerable to WNV infection, and this phenotype was associated with increased infection in peripheral organs, which resulted in higher virus titers in the central nervous system. The loss of IRF5, however, was associated with only small differences in the type I interferon response systemically and in the draining lymph node during WNV infection. Instead, lower levels of several other proinflammatory cytokines and chemokines, as well as fewer and less activated immune cells, were detected in the draining lymph node 2 days after WNV infection. WNV-specific antibody responses in Irf5-/- mice also were blunted in the context of live or inactivated virus infection and this was associated with fewer antigen-specific memory B cells and long-lived plasma cells. Our results with Irf5-/- mice establish a key role for IRF5 in shaping the early innate immune response in the draining lymph node, which impacts the spread of virus infection, optimal B cell immunity, and disease pathogenesis. IMPORTANCE: Although the roles of IRF3 and IRF7 in orchestrating innate and adaptive immunity after viral infection are established, the function of the related transcription factor IRF5 remains less certain. Prior studies in Irf5-/- mice reported conflicting results as to the contribution of IRF5 in regulating type I interferon and adaptive immune responses. The lack of clarity may stem from a recently discovered homozygous loss-of-function mutation of the immunoregulatory gene Dock2 in several colonies of Irf5-/- mice. Here, using a mouse model with a deficiency in IRF5 and wild-type Dock2 alleles, we investigated how IRF5 modulates West Nile virus (WNV) pathogenesis and host immune responses. Our in vivo studies indicate that IRF5 has a key role in shaping the early proinflammatory cytokine response in the draining lymph node, which impacts immunity and control of WNV infection.


Assuntos
Anticorpos Antivirais/sangue , Imunidade Inata , Fatores Reguladores de Interferon/imunologia , Linfonodos/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Imunidade Adaptativa , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos B/virologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Cruzamentos Genéticos , Feminino , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/imunologia , Deleção de Genes , Expressão Gênica/imunologia , Fatores de Troca do Nucleotídeo Guanina , Fatores Reguladores de Interferon/deficiência , Fatores Reguladores de Interferon/genética , Linfonodos/patologia , Linfonodos/virologia , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Carga Viral , Febre do Nilo Ocidental/genética , Febre do Nilo Ocidental/virologia
4.
PLoS Pathog ; 10(4): e1004072, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24743696

RESUMO

We recently described our most potently neutralizing monoclonal antibody, E106, which protected against lethal Dengue virus type 1 (DENV-1) infection in mice. To further understand its functional properties, we determined the crystal structure of E106 Fab in complex with domain III (DIII) of DENV-1 envelope (E) protein to 2.45 Šresolution. Analysis of the complex revealed a small antibody-antigen interface with the epitope on DIII composed of nine residues along the lateral ridge and A-strand regions. Despite strong virus neutralizing activity of E106 IgG at picomolar concentrations, E106 Fab exhibited a ∼20,000-fold decrease in virus neutralization and bound isolated DIII, E, or viral particles with only a micromolar monovalent affinity. In comparison, E106 IgG bound DENV-1 virions with nanomolar avidity. The E106 epitope appears readily accessible on virions, as neutralization was largely temperature-independent. Collectively, our data suggest that E106 neutralizes DENV-1 infection through bivalent engagement of adjacent DIII subunits on a single virion. The isolation of anti-flavivirus antibodies that require bivalent binding to inhibit infection efficiently may be a rare event due to the unique icosahedral arrangement of envelope proteins on the virion surface.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vírus da Dengue , Dengue , Imunoglobulina G , Proteínas do Envelope Viral , Animais , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/química , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/farmacologia , Afinidade de Anticorpos , Dengue/tratamento farmacológico , Dengue/imunologia , Vírus da Dengue/química , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Imunoglobulina G/química , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Camundongos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vírion/química , Vírion/genética , Vírion/imunologia
5.
PLoS Pathog ; 10(4): e1004086, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24743949

RESUMO

The type I interferon (IFN) signaling response limits infection of many RNA and DNA viruses. To define key cell types that require type I IFN signaling to orchestrate immunity against West Nile virus (WNV), we infected mice with conditional deletions of the type I IFN receptor (IFNAR) gene. Deletion of the Ifnar gene in subsets of myeloid cells resulted in uncontrolled WNV replication, vasoactive cytokine production, sepsis, organ damage, and death that were remarkably similar to infection of Ifnar-/- mice completely lacking type I IFN signaling. In Mavs-/-×Ifnar-/- myeloid cells and mice lacking both Ifnar and the RIG-I-like receptor adaptor gene Mavs, cytokine production was muted despite high levels of WNV infection. Thus, in myeloid cells, viral infection triggers signaling through MAVS to induce proinflammatory cytokines that can result in sepsis and organ damage. Viral pathogenesis was caused in part by massive complement activation, as liver damage was minimized in animals lacking complement components C3 or factor B or treated with neutralizing anti-C5 antibodies. Disease in Ifnar-/- and CD11c Cre+Ifnarf/f mice also was facilitated by the proinflammatory cytokine TNF-α, as blocking antibodies diminished complement activation and prolonged survival without altering viral burden. Collectively, our findings establish the dominant role of type I IFN signaling in myeloid cells in restricting virus infection and controlling pathological inflammation and tissue injury.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Sepse/metabolismo , Transdução de Sinais , Febre do Nilo Ocidental/metabolismo , Vírus do Nilo Ocidental/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Complemento C3/genética , Complemento C3/metabolismo , Fator B do Complemento/genética , Fator B do Complemento/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Sepse/genética , Sepse/patologia , Sepse/virologia , Febre do Nilo Ocidental/genética , Febre do Nilo Ocidental/patologia
6.
Nature ; 505(7485): 691-5, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24284630

RESUMO

The type I interferon (IFN) response protects cells from viral infection by inducing hundreds of interferon-stimulated genes (ISGs), some of which encode direct antiviral effectors. Recent screening studies have begun to catalogue ISGs with antiviral activity against several RNA and DNA viruses. However, antiviral ISG specificity across multiple distinct classes of viruses remains largely unexplored. Here we used an ectopic expression assay to screen a library of more than 350 human ISGs for effects on 14 viruses representing 7 families and 11 genera. We show that 47 genes inhibit one or more viruses, and 25 genes enhance virus infectivity. Comparative analysis reveals that the screened ISGs target positive-sense single-stranded RNA viruses more effectively than negative-sense single-stranded RNA viruses. Gene clustering highlights the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS, also known as MB21D1) as a gene whose expression also broadly inhibits several RNA viruses. In vitro, lentiviral delivery of enzymatically active cGAS triggers a STING-dependent, IRF3-mediated antiviral program that functions independently of canonical IFN/STAT1 signalling. In vivo, genetic ablation of murine cGAS reveals its requirement in the antiviral response to two DNA viruses, and an unappreciated contribution to the innate control of an RNA virus. These studies uncover new paradigms for the preferential specificity of IFN-mediated antiviral pathways spanning several virus families.


Assuntos
Imunidade Inata/genética , Imunidade Inata/imunologia , Interferons/imunologia , Nucleotidiltransferases/imunologia , Nucleotidiltransferases/metabolismo , Vírus/imunologia , Animais , Análise por Conglomerados , Vírus de DNA/imunologia , Vírus de DNA/patogenicidade , Citometria de Fluxo , Biblioteca Gênica , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferons/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Nucleotidiltransferases/deficiência , Nucleotidiltransferases/genética , Vírus de RNA/imunologia , Vírus de RNA/patogenicidade , Fator de Transcrição STAT1/metabolismo , Especificidade por Substrato , Vírus/classificação , Vírus/patogenicidade
7.
J Virol ; 87(21): 11401-15, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23966390

RESUMO

Many viruses induce type I interferon responses by activating cytoplasmic RNA sensors, including the RIG-I-like receptors (RLRs). Although two members of the RLR family, RIG-I and MDA5, have been implicated in host control of virus infection, the relative role of each RLR in restricting pathogenesis in vivo remains unclear. Recent studies have demonstrated that MAVS, the adaptor central to RLR signaling, is required to trigger innate immune defenses and program adaptive immune responses, which together restrict West Nile virus (WNV) infection in vivo. In this study, we examined the specific contribution of MDA5 in controlling WNV in animals. MDA5(-/-) mice exhibited enhanced susceptibility, as characterized by reduced survival and elevated viral burden in the central nervous system (CNS) at late times after infection, even though small effects on systemic type I interferon response or viral replication were observed in peripheral tissues. Intracranial inoculation studies and infection experiments with primary neurons ex vivo revealed that an absence of MDA5 did not impact viral infection in neurons directly. Rather, subtle defects were observed in CNS-specific CD8(+) T cells in MDA5(-/-) mice. Adoptive transfer into recipient MDA5(+/+) mice established that a non-cell-autonomous deficiency of MDA5 was associated with functional defects in CD8(+) T cells, which resulted in a failure to clear WNV efficiently from CNS tissues. Our studies suggest that MDA5 in the immune priming environment shapes optimal CD8(+) T cell activation and subsequent clearance of WNV from the CNS.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Sistema Nervoso Central/imunologia , RNA Helicases DEAD-box/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Linfócitos T CD8-Positivos/virologia , Sistema Nervoso Central/virologia , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/metabolismo , Modelos Animais de Doenças , Helicase IFIH1 Induzida por Interferon , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Reconhecimento de Padrão/metabolismo , Análise de Sobrevida , Carga Viral , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/isolamento & purificação
8.
Cell Host Microbe ; 14(2): 136-47, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23954153

RESUMO

Upon activation by the ligands Gas6 and Protein S, Tyro3/Axl/Mer (TAM) receptor tyrosine kinases promote phagocytic clearance of apoptotic cells and downregulate immune responses initiated by Toll-like receptors and type I interferons (IFNs). Many enveloped viruses display the phospholipid phosphatidylserine on their membranes, through which they bind Gas6 and Protein S and engage TAM receptors. We find that ligand-coated viruses activate TAM receptors on dendritic cells (DCs), dampen type I IFN signaling, and thereby evade host immunity and promote infection. Upon virus challenge, TAM-deficient DCs display type I IFN responses that are elevated in comparison to wild-type cells. As a consequence, TAM-deficient DCs are relatively resistant to infection by flaviviruses and pseudotyped retroviruses, but infection can be restored with neutralizing type I IFN antibodies. Correspondingly, a TAM kinase inhibitor antagonizes the infection of wild-type DCs. Thus, TAM receptors are engaged by viruses in order to attenuate type I IFN signaling and represent potential therapeutic targets.


Assuntos
Células Dendríticas/imunologia , Flavivirus/imunologia , Tolerância Imunológica , Imunidade Inata , Retroviridae/imunologia , Animais , Linhagem Celular , Humanos , Interferon Tipo I/biossíntese , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
9.
J Virol ; 87(15): 8363-71, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23740986

RESUMO

Previous studies have demonstrated that type I interferon (IFN-I) restricts West Nile virus (WNV) replication and pathogenesis in peripheral and central nervous system (CNS) tissues. However, the in vivo role of specific antiviral genes that are induced by IFN-I against WNV infection remains less well characterized. Here, using Ifit2(-/-) mice, we defined the antiviral function of the interferon-stimulated gene (ISG) Ifit2 in limiting infection and disease in vivo by a virulent North American strain of WNV. Compared to congenic wild-type controls, Ifit2(-/-) mice showed enhanced WNV infection in a tissue-restricted manner, with preferential replication in the CNS of animals lacking Ifit2. Virological analysis of cultured macrophages, dendritic cells, fibroblasts, cerebellar granule cell neurons, and cortical neurons revealed cell type-specific antiviral functions of Ifit2 against WNV. In comparison, small effects of Ifit2 were observed on the induction or magnitude of innate or adaptive immune responses. Our results suggest that Ifit2 restricts WNV infection and pathogenesis in different tissues in a cell type-specific manner.


Assuntos
Encéfalo/imunologia , Encéfalo/virologia , Proteínas/metabolismo , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Proteínas Reguladoras de Apoptose , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas/genética , Proteínas de Ligação a RNA , Febre do Nilo Ocidental/patologia , Febre do Nilo Ocidental/virologia
10.
J Virol ; 87(16): 8826-42, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23785205

RESUMO

Although prior studies have characterized the neutralizing activities of monoclonal antibodies (MAbs) against dengue virus (DENV) serotypes 1, 2, and 3 (DENV-1, DENV-2, and DENV-3), few reports have assessed the activity of MAbs against DENV-4. Here, we evaluated the inhibitory activity of 81 new mouse anti-DENV-4 MAbs. We observed strain- and genotype-dependent differences in neutralization of DENV-4 by MAbs mapping to epitopes on domain II (DII) and DIII of the envelope (E) protein. Several anti-DENV-4 MAbs inefficiently inhibited at least one strain and/or genotype, suggesting that the exposure or sequence of neutralizing epitopes varies within isolates of this serotype. Remarkably, flavivirus cross-reactive MAbs, which bound to the highly conserved fusion loop in DII and inhibited infection of DENV-1, DENV-2, and DENV-3, more weakly neutralized five different DENV-4 strains encompassing the genetic diversity of the serotype after preincubation at 37°C. However, increasing the time of preincubation at 37°C or raising the temperature to 40°C enhanced the potency of DII fusion loop-specific MAbs and some DIII-specific MAbs against DENV-4 strains. Prophylaxis studies in two new DENV-4 mouse models showed that neutralization titers of MAbs after preincubation at 37°C correlated with activity in vivo. Our studies establish the complexity of MAb recognition against DENV-4 and suggest that differences in epitope exposure relative to other DENV serotypes affect antibody neutralization and protective activity.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus da Dengue/imunologia , Epitopos/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Reações Cruzadas , Dengue/prevenção & controle , Modelos Animais de Doenças , Mapeamento de Epitopos , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Temperatura , Fatores de Tempo
11.
PLoS Pathog ; 8(10): e1002930, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23055922

RESUMO

We previously developed a panel of neutralizing monoclonal antibodies against Dengue virus (DENV)-1, of which few exhibited inhibitory activity against all DENV-1 genotypes. This finding is consistent with reports observing variable neutralization of different DENV strains and genotypes using serum from individuals that experienced natural infection or immunization. Herein, we describe the crystal structures of DENV1-E111 bound to a novel CC' loop epitope on domain III (DIII) of the E protein from two different DENV-1 genotypes. Docking of our structure onto the available cryo-electron microscopy models of DENV virions revealed that the DENV1-E111 epitope was inaccessible, suggesting that this antibody recognizes an uncharacterized virus conformation. While the affinity of binding between DENV1-E111 and DIII varied by genotype, we observed limited correlation with inhibitory activity. Instead, our results support the conclusion that potent neutralization depends on genotype-dependent exposure of the CC' loop epitope. These findings establish new structural complexity of the DENV virion, which may be relevant for the choice of DENV strain for induction or analysis of neutralizing antibodies in the context of vaccine development.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Vírus da Dengue/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Anticorpos Monoclonais/imunologia , Sítios de Ligação de Anticorpos , Dengue/imunologia , Dengue/prevenção & controle , Vírus da Dengue/genética , Mapeamento de Epitopos , Epitopos/genética , Epitopos/imunologia , Humanos , Estrutura Terciária de Proteína
12.
J Virol ; 86(17): 8937-48, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740407

RESUMO

Previous studies of mice have demonstrated that an orchestrated sequence of innate and adaptive immune responses is required to control West Nile virus (WNV) infection in peripheral and central nervous system (CNS) tissues. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL; also known as CD253) has been reported to inhibit infection with dengue virus, a closely related flavivirus, in cell culture. To determine the physiological function of TRAIL in the context of flavivirus infection, we compared the pathogenesis of WNV in wild-type and TRAIL(-/-) mice. Mice lacking TRAIL showed increased vulnerability and death after subcutaneous WNV infection. Although no difference in viral burden was detected in peripheral tissues, greater viral infection was detected in the brain and spinal cord at late times after infection, and this was associated with delayed viral clearance in the few surviving TRAIL(-/-) mice. While priming of adaptive B and T cell responses and trafficking of immune and antigen-specific cells to the brain were undistinguishable from those in normal mice, in TRAIL(-/-) mice, CD8(+) T cells showed qualitative defects in the ability to clear WNV infection. Adoptive transfer of WNV-primed wild-type but not TRAIL(-/-) CD8(+) T cells to recipient CD8(-/-) mice efficiently limited infection in the brain and spinal cord, and analogous results were obtained when wild-type or TRAIL(-/-) CD8(+) T cells were added to WNV-infected primary cortical neuron cultures ex vivo. Collectively, our results suggest that TRAIL produced by CD8(+) T cells contributes to disease resolution by helping to clear WNV infection from neurons in the central nervous system.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neurônios/virologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Transferência Adotiva , Animais , Anticorpos Antivirais/imunologia , Encéfalo/imunologia , Encéfalo/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/imunologia , Medula Espinal/imunologia , Medula Espinal/virologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Carga Viral , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia
13.
Cell Host Microbe ; 11(4): 397-409, 2012 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-22520467

RESUMO

Host resistance to viral infection requires type I (α/ß) and II (γ) interferon (IFN) production. Another important defense mechanism is the degradative activity of macroautophagy (herein autophagy), mediated by the coordinated action of evolutionarily conserved autophagy proteins (Atg). We show that the Atg5-Atg12/Atg16L1 protein complex, whose prior known function is in autophagosome formation, is required for IFNγ-mediated host defense against murine norovirus (MNV) infection. Importantly, the direct antiviral activity of IFNγ against MNV in macrophages required Atg5-Atg12, Atg7, and Atg16L1, but not induction of autophagy, the degradative activity of lysosomal proteases, fusion of autophagosomes and lysosomes, or the Atg8-processing protein Atg4B. IFNγ, via Atg5-Atg12/Atg16L1, inhibited formation of the membranous cytoplasmic MNV replication complex, where Atg16L1 localized. Thus, the Atg5-Atg12/Atg16L1 complex performs a pivotal, nondegradative role in IFNγ-mediated antiviral defense, establishing that multicellular organisms have evolved to use portions of the autophagy pathway machinery in a cassette-like fashion for host defense.


Assuntos
Infecções por Caliciviridae/metabolismo , Proteínas de Transporte/metabolismo , Interferon gama/imunologia , Proteínas Associadas aos Microtúbulos/metabolismo , Norovirus/fisiologia , Proteínas/metabolismo , Animais , Autofagia , Proteína 12 Relacionada à Autofagia , Proteína 5 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Infecções por Caliciviridae/genética , Infecções por Caliciviridae/imunologia , Proteínas de Transporte/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Ligação Proteica , Proteínas/genética
14.
Virology ; 427(2): 127-34, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22406169

RESUMO

DENV1-E106 is a monoclonal antibody (MAb) with strong neutralizing activity against all five DENV-1 genotypes and therapeutic activity in mice. Here, we evaluated the potential for DENV-1 to escape neutralization by DENV1-E106. A single mutation in domain III of the envelope protein (T329A) emerged, which conferred resistance to DENV1-E106. However, the T329A variant virus had differing phenotypes in vitro and in vivo with attenuation in cell culture yet increased infectivity in Aedes aegypti mosquitoes. Mice infected with this T329A variant still were protected against lethal infection by DENV1-E106 even though much of the neutralizing activity was lost. This study reveals the complex dynamics of neutralization escape of an inhibitory MAb against DENV, and suggests that evaluation of therapeutic MAbs requires detailed investigation in relevant hosts.


Assuntos
Aedes/virologia , Vírus da Dengue/imunologia , Dengue/virologia , Animais , Anticorpos Monoclonais , Linhagem Celular , Cricetinae , Dengue/imunologia , Vírus da Dengue/classificação , Vírus da Dengue/genética , Vírus da Dengue/patogenicidade , Insetos Vetores , Camundongos , Camundongos Knockout , Mutação , Fenótipo , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Virulência
15.
PLoS Pathog ; 6(4): e1000823, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20369024

RESUMO

Antibody protection against flaviviruses is associated with the development of neutralizing antibodies against the viral envelope (E) protein. Prior studies with West Nile virus (WNV) identified therapeutic mouse and human monoclonal antibodies (MAbs) that recognized epitopes on domain III (DIII) of the E protein. To identify an analogous panel of neutralizing antibodies against DENV type-1 (DENV-1), we immunized mice with a genotype 2 strain of DENV-1 virus and generated 79 new MAbs, 16 of which strongly inhibited infection by the homologous virus and localized to DIII. Surprisingly, only two MAbs, DENV1-E105 and DENV1-E106, retained strong binding and neutralizing activity against all five DENV-1 genotypes. In an immunocompromised mouse model of infection, DENV1-E105 and DENV1-E106 exhibited therapeutic activity even when administered as a single dose four days after inoculation with a heterologous genotype 4 strain of DENV-1. Using epitope mapping and X-ray crystallographic analyses, we localized the neutralizing determinants for the strongly inhibitory MAbs to distinct regions on DIII. Interestingly, sequence variation in DIII alone failed to explain disparities in neutralizing potential of MAbs among different genotypes. Overall, our experiments define a complex structural epitope on DIII of DENV-1 that can be recognized by protective antibodies with therapeutic potential.


Assuntos
Anticorpos Neutralizantes/imunologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
16.
J Exp Med ; 205(10): 2319-37, 2008 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-18779347

RESUMO

In a lethal West Nile virus (WNV) model, central nervous system infection triggered a threefold increase in CD45(int)/CD11b(+)/CD11c(-) microglia at days 6-7 postinfection (p.i.). Few microglia were proliferating, suggesting that the increased numbers were derived from a migratory precursor cell. Depletion of "circulating" (Gr1(-)(Ly6C(lo))CX3CR1(+)) and "inflammatory" (Gr1(hi)/Ly6C(hi)/CCR2(+)) classical monocytes during infection abrogated the increase in microglia. C57BL/6 chimeras reconstituted with cFMS-enhanced green fluorescent protein (EGFP) bone marrow (BM) showed large numbers of peripherally derived (GFP(+)) microglia expressing GR1(+)(Ly6C(+)) at day 7 p.i., suggesting that the inflammatory monocyte is a microglial precursor. This was confirmed by adoptive transfer of labeled BM (Ly6C(hi)/CD115(+)) or circulating inflammatory monocytes that trafficked to the WNV-infected brain and expressed a microglial phenotype. CCL2 is a chemokine that is highly expressed during WNV infection and important in inflammatory monocyte trafficking. Neutralization of CCL2 not only reduced the number of GFP(+) microglia in the brain during WNV infection but prolonged the life of infected animals. Therefore, CCL2-dependent inflammatory monocyte migration is critical for increases in microglia during WNV infection and may also play a pathogenic role during WNV encephalitis.


Assuntos
Antígenos Ly/imunologia , Encéfalo/imunologia , Encéfalo/virologia , Microglia/imunologia , Monócitos/imunologia , Células-Tronco/imunologia , Febre do Nilo Ocidental/imunologia , Transferência Adotiva , Animais , Barreira Hematoencefálica/fisiologia , Encéfalo/citologia , Encéfalo/patologia , Quimiocina CCL2/imunologia , Ácido Clodrônico/metabolismo , Feminino , Antígenos Comuns de Leucócito/imunologia , Lipossomos/química , Lipossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/fisiologia , Monócitos/citologia , Células-Tronco/citologia , Taxa de Sobrevida , Febre do Nilo Ocidental/patologia , Vírus do Nilo Ocidental
17.
J Virol ; 82(18): 8956-64, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18632856

RESUMO

West Nile virus (WNV) is a neurotropic flavivirus that has emerged globally as a significant cause of viral encephalitis in humans, especially in immunocompromised individuals. Previous studies have shown essential protective roles for antiviral cytokines (e.g., alpha interferon [IFN-alpha] and IFN-gamma) against WNV in mice. However, studies using cell culture offer conflicting answers regarding whether tumor necrosis factor alpha (TNF-alpha) has an anti-WNV function. To test the biological significance of TNF-alpha against WNV in vivo, experiments were performed with TNF receptor-1 (TNF-R1)-deficient and TNF-alpha-depleted C57BL/6 mice. TNF-R1(-/-) mice had enhanced mortality and decreased survival time after WNV infection compared to congenic wild-type mice. Consistent with this, administration of a neutralizing anti-TNF-alpha monoclonal antibody also decreased survival after WNV infection. Relatively small differences in viral burdens in peripheral tissues of TNF-R1(-/-) mice were observed, and this occurrence correlated with a modest antiviral effect of TNF-alpha on primary macrophages but not dendritic cells. In contrast, the viral titers detected in the central nervous systems of TNF-R1(-/-) mice were significantly increased compared to those of wild-type mice, although TNF-alpha did not have a direct antiviral effect in primary neuron cultures. Whereas no defect in priming of adaptive B- and T-cell responses in TNF-R1(-/-) mice was observed, there were significant reductions in accumulations of CD8+ T cells and macrophages in the brain. Our data are most consistent with a model in which interaction of TNF-alpha with TNF-R1 protects against WNV infection by regulating migration of protective inflammatory cells into the brain during acute infection.


Assuntos
Movimento Celular/imunologia , Sistema Nervoso Central/imunologia , Leucócitos Mononucleares/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Animais , Anticorpos Antivirais/sangue , Encéfalo/imunologia , Encéfalo/virologia , Linfócitos T CD8-Positivos/imunologia , Sistema Nervoso Central/virologia , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/genética , Febre do Nilo Ocidental/mortalidade , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/imunologia
18.
Vaccine ; 26(16): 2020-33, 2008 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-18339459

RESUMO

West Nile virus (WNV) is a mosquito borne, neurotropic flavivirus that causes a severe central nervous system (CNS) infection in humans and animals. Although commercial vaccines are available for horses, none is currently approved for human use. In this study, we evaluated the efficacy and mechanism of immune protection of two candidate WNV vaccines in mice. A formalin-inactivated WNV vaccine induced higher levels of specific and neutralizing antibodies compared to a DNA plasmid vaccine that produces virus-like particles. Accordingly, partial and almost complete protection against a highly stringent lethal intracranial WNV challenge were observed in mice 60 days after single dose immunization with the DNA plasmid and inactivated virus vaccines, respectively. In mice immunized with a single dose of DNA plasmid or inactivated vaccine, antigen-specific CD8(+) T cells were induced and contributed to protective immunity as acquired or genetic deficiencies of CD8(+) T cells lowered the survival rates. In contrast, in boosted animals, WNV-specific antibody titers were higher, survival rates after challenge were greater, and an absence of CD8(+) T cells did not appreciably affect mortality. Overall, our experiments suggest that in mice, both inactivated WNV and DNA plasmid vaccines are protective after two doses, and the specific contribution of antibody and CD8(+) T cells to vaccine immunity against WNV is modulated by the prime-boost strategy.


Assuntos
Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Animais , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Antígenos CD8/genética , Imunização Secundária , Injeções Intramusculares , Injeções Intraperitoneais , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Plasmídeos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Febre do Nilo Ocidental/sangue
19.
J Neurochem ; 103(3): 1019-30, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17854352

RESUMO

Seizures are a major complication of viral encephalitis. However, the mechanisms of seizure-associated neuronal dysfunction remain poorly understood. We report that intranasal inoculation with West Nile virus (WNV) (Sarafend) causes limbic seizures in C57BL/6 mice, but not in interferon (IFN)-gamma-deficient (IFN-gamma-/-) mice. Both strains showed similar levels of virus in the brain, as well as similar concentrations of the cytokines, tumor necrosis factor and interleukin-6, both of which can alter neuronal excitability. Experiments in chimeric IFN-gamma-/- mice reconstituted with IFN-gamma-producing leukocytes showed that IFN-gamma is not required during central nervous system infection for limbic seizure development, suggesting a role for IFN-gamma in the developing brain. This was supported responses to pentylenetetrazole, kainic acid (KA), and N-methyl-d-aspartate (NMDA). Both strains of mice exhibited similar behavior after pentylenetetrazole challenge. However, while NMDA and KA treatment resulted in characteristic seizures in C57BL/6 mice, these responses were diminished (NMDA treatment) or absent (KA treatment) in IFN-gamma-/- mice. Furthermore, NMDA-receptor blockade with MK-801 in WNV-infected C57BL/6 mice abrogated seizures and prolonged survival. Our data show that IFN-gamma plays an important role in the development of the excitatory seizure pathways in the brain and that these cascades become pathogenic in encephalitic WNV infection.


Assuntos
Encéfalo/imunologia , Encéfalo/virologia , Interferon gama/imunologia , Convulsões/imunologia , Convulsões/virologia , Febre do Nilo Ocidental/imunologia , Animais , Encéfalo/fisiopatologia , Chlorocebus aethiops , Convulsivantes/farmacologia , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Predisposição Genética para Doença/genética , Ácido Glutâmico/metabolismo , Interferon gama/genética , Interleucina-6/imunologia , Interleucina-6/metabolismo , Encefalite Límbica/genética , Encefalite Límbica/imunologia , Encefalite Límbica/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/imunologia , Vias Neurais/fisiopatologia , Vias Neurais/virologia , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/genética , Transmissão Sináptica/genética , Transmissão Sináptica/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Células Vero , Febre do Nilo Ocidental/complicações , Febre do Nilo Ocidental/fisiopatologia
20.
J Virol ; 81(21): 11749-57, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17804505

RESUMO

West Nile virus (WNV) is a neurotropic flavivirus that causes encephalitis, most frequently in elderly and immunocompromised humans. Previous studies demonstrated that CD8+ T cells utilize perforin-dependent cytolytic mechanisms to limit WNV infection. Nonetheless, the phenotype of perforin-deficient CD8+ T cells was not as severe as that of an absence of CD8+ T cells, suggesting additional effector control mechanisms. In this study, we evaluated the contribution of Fas-Fas ligand (FasL) interactions to CD8+ T-cell-mediated control of WNV infection. Notably, the cell death receptor Fas was strongly upregulated on neurons in culture and in vivo after WNV infection. gld mice that were functionally deficient in FasL expression showed increased susceptibility to lethal WNV infection. Although antigen-specific priming of CD8+ T cells in peripheral lymphoid tissues was normal in gld mice, increased central nervous system (CNS) viral burdens and delayed clearance were observed. Moreover, the adoptive transfer of WNV-primed wild-type but not gld CD8+ T cells to recipient CD8(-/-) or gld mice efficiently limited infection in the CNS and enhanced survival rates. Overall, our data suggest that CD8+ T cells also utilize FasL effector mechanisms to contain WNV infection in Fas-expressing neurons in the CNS.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Infecções do Sistema Nervoso Central/virologia , Proteína Ligante Fas/fisiologia , Regulação Viral da Expressão Gênica , Febre do Nilo Ocidental/virologia , Animais , Linfócitos T CD8-Positivos/virologia , Cricetinae , Proteína Ligante Fas/metabolismo , Humanos , Interferon gama/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neurônios/metabolismo , Vírus do Nilo Ocidental/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...