Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 2(13)2017 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-28679950

RESUMO

Dengue virus (DENV) is the most prevalent mosquito-borne virus causing human disease. Of the 4 DENV serotypes, epidemiological data suggest that DENV-2 secondary infections are associated with more severe disease than DENV-4 infections. Mass cytometry by time-of-flight (CyTOF) was used to dissect immune changes induced by DENV-2 and DENV-4 in human DCs, the initial targets of primary infections that likely affect infection outcomes. Strikingly, DENV-4 replication peaked earlier and promoted stronger innate immune responses, with increased expression of DC activation and migration markers and increased cytokine production, compared with DENV-2. In addition, infected DCs produced higher levels of inflammatory cytokines compared with bystander DCs, which mainly produced IFN-induced cytokines. These high-dimensional analyses during DENV-2 and DENV-4 infections revealed distinct viral signatures marked by different replication strategies and antiviral innate immune induction in DCs, which may result in different viral fitness, transmission, and pathogenesis.

2.
Biochem Biophys Res Commun ; 492(4): 587-596, 2017 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-28576494

RESUMO

The prompt and tightly controlled induction of type I interferon is a central event of the immune defense against viral infection. Flaviviruses comprise a large family of arthropod-borne positive-stranded RNA viruses, many of which represent a serious threat to global human health due to their high rates of morbidity and mortality. All flaviviruses studied so far have been shown to counteract the host's immune response to establish a productive infection and facilitate viral spread. Here, we review the current knowledge on the main strategies that human pathogenic flaviviruses utilize to escape both type I IFN induction and effector pathways. A better understanding of the specific mechanisms by which flaviviruses activate and evade innate immune responses is critical for the development of better therapeutics and vaccines.


Assuntos
Infecções por Flavivirus/imunologia , Infecções por Flavivirus/virologia , Flavivirus/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Animais , Humanos , Modelos Imunológicos , Replicação Viral/imunologia
3.
PLoS Pathog ; 13(3): e1006258, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28278235

RESUMO

Zika virus (ZIKV) is a mosquito borne flavivirus, which was a neglected tropical pathogen until it emerged and spread across the Pacific Area and the Americas, causing large human outbreaks associated with fetal abnormalities and neurological disease in adults. The factors that contributed to the emergence, spread and change in pathogenesis of ZIKV are not understood. We previously reported that ZIKV evades cellular antiviral responses by targeting STAT2 for degradation in human cells. In this study, we demonstrate that Stat2-/- mice are highly susceptible to ZIKV infection, recapitulate virus spread to the central nervous system (CNS), gonads and other visceral organs, and display neurological symptoms. Further, we exploit this model to compare ZIKV pathogenesis caused by a panel of ZIKV strains of a range of spatiotemporal history of isolation and representing African and Asian lineages. We observed that African ZIKV strains induce short episodes of severe neurological symptoms followed by lethality. In comparison, Asian strains manifest prolonged signs of neuronal malfunctions, occasionally causing death of the Stat2-/- mice. African ZIKV strains induced higher levels of inflammatory cytokines and markers associated with cellular infiltration in the infected brain in mice, which may explain exacerbated pathogenesis in comparison to those of the Asian lineage. Interestingly, viral RNA levels in different organs did not correlate with the pathogenicity of the different strains. Taken together, we have established a new murine model that supports ZIKV infection and demonstrate its utility in highlighting intrinsic differences in the inflammatory response induced by different ZIKV strains leading to severity of disease. This study paves the way for the future interrogation of strain-specific changes in the ZIKV genome and their contribution to viral pathogenesis.


Assuntos
Modelos Animais de Doenças , Infecção por Zika virus/imunologia , Zika virus/imunologia , Zika virus/patogenicidade , Animais , Inflamação/imunologia , Inflamação/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Filogenia , Reação em Cadeia da Polimerase em Tempo Real , Zika virus/genética
4.
Nat Microbiol ; 2: 17037, 2017 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-28346446

RESUMO

During the last few decades, the global incidence of dengue virus (DENV) has increased dramatically, and it is now endemic in more than 100 countries. To establish a productive infection in humans, DENV uses different strategies to inhibit or avoid the host innate immune system. Several DENV proteins have been shown to strategically target crucial components of the type I interferon system. Here, we report that the DENV NS2B protease cofactor targets the DNA sensor cyclic GMP-AMP synthase (cGAS) for lysosomal degradation to avoid the detection of mitochondrial DNA during infection. Such degradation subsequently results in the inhibition of type I interferon production in the infected cell. Our data demonstrate a mechanism by which cGAS senses cellular damage upon DENV infection.


Assuntos
DNA Mitocondrial/fisiologia , Vírus da Dengue/genética , Interações Hospedeiro-Patógeno , Nucleotidiltransferases/metabolismo , Proteínas não Estruturais Virais/metabolismo , DNA Mitocondrial/genética , Células Dendríticas/virologia , Dengue/imunologia , Dengue/virologia , Vírus da Dengue/química , Vírus da Dengue/enzimologia , Vírus da Dengue/imunologia , Células HEK293 , Humanos , Imunidade Inata , Interferon Tipo I/imunologia , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/deficiência , Nucleotidiltransferases/genética , Nucleotidiltransferases/imunologia , Transdução de Sinais , Proteínas não Estruturais Virais/genética
5.
PLoS Pathog ; 13(3): e1006265, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28264033

RESUMO

The Flavivirus genus includes a large number of medically relevant pathogens that cycle between humans and arthropods. This host alternation imposes a selective pressure on the viral population. Here, we found that dengue virus, the most important viral human pathogen transmitted by insects, evolved a mechanism to differentially regulate the production of viral non-coding RNAs in mosquitos and humans, with a significant impact on viral fitness in each host. Flavivirus infections accumulate non-coding RNAs derived from the viral 3'UTRs (known as sfRNAs), relevant in viral pathogenesis and immune evasion. We found that dengue virus host adaptation leads to the accumulation of different species of sfRNAs in vertebrate and invertebrate cells. This process does not depend on differences in the host machinery; but it was found to be dependent on the selection of specific mutations in the viral 3'UTR. Dissecting the viral population and studying phenotypes of cloned variants, the molecular determinants for the switch in the sfRNA pattern during host change were mapped to a single RNA structure. Point mutations selected in mosquito cells were sufficient to change the pattern of sfRNAs, induce higher type I interferon responses and reduce viral fitness in human cells, explaining the rapid clearance of certain viral variants after host change. In addition, using epidemic and pre-epidemic Zika viruses, similar patterns of sfRNAs were observed in mosquito and human infected cells, but they were different from those observed during dengue virus infections, indicating that distinct selective pressures act on the 3'UTR of these closely related viruses. In summary, we present a novel mechanism by which dengue virus evolved an RNA structure that is under strong selective pressure in the two hosts, as regulator of non-coding RNA accumulation and viral fitness. This work provides new ideas about the impact of host adaptation on the variability and evolution of flavivirus 3'UTRs with possible implications in virulence and viral transmission.


Assuntos
Adaptação Biológica/genética , Culicidae/virologia , Vírus da Dengue/genética , Aptidão Genética/genética , RNA Viral/genética , Regiões 3' não Traduzidas/genética , Animais , Northern Blotting , Dengue/genética , Variação Genética , Genoma Viral , Interações Hospedeiro-Patógeno/genética , Humanos , Insetos Vetores/virologia , Filogenia , Reação em Cadeia da Polimerase , Transfecção
6.
JCI Insight ; 2(4): e88226, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28239647

RESUMO

HIV-1 viremic controllers (VC) spontaneously control infection without antiretroviral treatment. Several studies indicate that IgG Abs from VCs induce enhanced responses from immune effector cells. Since signaling through Fc-γ receptors (FCGRs) modulate these Ab-driven responses, here we examine if enhanced FCGR activation is a common feature of IgG from VCs. Using an infected cell-based system, we observed that VC IgG stimulated greater FCGR2A and FCGR3A activation as compared with noncontrollers, independent of the magnitude of HIV-specific Ab binding or virus neutralization activities. Multivariate regression analysis showed that enhanced FCGR signaling was a significant predictor of VC status as compared with chronically infected patients (CIP) on highly active antiretroviral therapy (HAART). Unsupervised hierarchical clustering of patient IgG functions primarily grouped VC IgG profiles by enhanced FCGR2A, FCGR3A, or dual signaling activity. Our findings demonstrate that enhanced FCGR signaling is a common and significant predictive feature of VC IgG, with VCs displaying a distinct spectrum of FCGR activation profiles. Thus, profiling FCGR activation may provide a useful method for screening and distinguishing protective anti-HIV IgG responses in HIV-infected patients and in monitoring HIV vaccination regimens.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , Imunoglobulina G/imunologia , Receptores de IgG/imunologia , Viremia/imunologia , Terapia Antirretroviral de Alta Atividade , Resistência à Doença , Feminino , Infecções por HIV/tratamento farmacológico , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Transdução de Sinais
10.
Cell Host Microbe ; 18(3): 345-53, 2015 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-26355217

RESUMO

Combination antiretroviral therapy (ART) is able to suppress HIV-1 replication to undetectable levels. However, the persistence of latent viral reservoirs allows for a rebound of viral load upon cessation of therapy. Thus, therapeutic strategies to eradicate the viral latent reservoir are critically needed. Employing a targeted RNAi screen, we identified the ubiquitin ligase BIRC2 (cIAP1), a repressor of the noncanonical NF-κB pathway, as a potent negative regulator of LTR-dependent HIV-1 transcription. Depletion of BIRC2 through treatment with small molecule antagonists known as Smac mimetics enhanced HIV-1 transcription, leading to a reversal of latency in a JLat latency model system. Critically, treatment of resting CD4+ T cells isolated from ART-suppressed patients with the histone deacetylase inhibitor (HDACi) panobinostat together with Smac mimetics resulted in synergistic activation of the latent reservoir. These data implicate Smac mimetics as useful agents for shock-and-kill strategies to eliminate the latent HIV reservoir.


Assuntos
Regulação Viral da Expressão Gênica , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Inibidoras de Apoptose/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Humanos , Ácidos Hidroxâmicos/metabolismo , Indóis/metabolismo , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Oligopeptídeos/metabolismo , Panobinostat , Ubiquitina-Proteína Ligases/antagonistas & inibidores
11.
J Virol ; 89(19): 9781-90, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26178989

RESUMO

UNLABELLED: The accessory HIV protein Vpu inhibits a number of cellular pathways that trigger host innate restriction mechanisms. HIV Vpu-mediated degradation of tetherin allows efficient particle release and hampers the activation of the NF-κB pathway thereby limiting the expression of proinflammatory genes. In addition, Vpu reduces cell surface expression of several cellular molecules such as newly synthesized CD4. However, the role of HIV Vpu in regulating the type 1 interferon response to viral infection by degradation of the interferon regulatory factor 3 (IRF3) has been subject of conflicting reports. We therefore systematically investigated the expression of IRF3 in primary CD4(+) T cells and macrophages infected with HIV at different time points. In addition, we also tested the ability of Vpu to interfere with innate immune signaling pathways such as the NF-κB and the IRF3 pathways. We report here that HIV Vpu failed to degrade IRF3 in infected primary cells. Moreover, we observed that HIV NL4.3 Vpu had no effect on IRF3-dependent gene expression in reporter assays. On the other hand, HIV NL4.3 Vpu downmodulated NF-κB-dependent transcription. Mutation of two serines (positions 52 and 56) involved in the binding of NL4.3 Vpu to the ßTrCP ubiquitin ligase abolishes its ability to inhibit NF-κB activity. Taken together, these results suggest that HIV Vpu regulates antiviral innate response in primary human cells by acting specifically on the NF-κB pathway. IMPORTANCE: HIV Vpu plays a pivotal role in enhancing HIV infection by counteraction of Tetherin. However, Vpu also regulates host response to HIV infection by hampering the type 1 interferon response. The molecular mechanism by which Vpu inhibits the interferon response is still controversial. Here we report that Vpu affects interferon expression by inhibiting NF-κB activity without affecting IRF3 levels or activity. These data suggest that Vpu facilitates HIV infection by regulating NF-κB transcription to levels sufficient for viral transcription while limiting cellular responses to infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Regulação da Expressão Gênica/imunologia , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Linfócitos T CD4-Positivos/virologia , Primers do DNA/genética , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , HIV-1/metabolismo , Humanos , Immunoblotting , Fator Regulador 3 de Interferon/metabolismo , Luciferases , Plasmídeos/genética
12.
Nat Immunol ; 16(5): 485-94, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25822250

RESUMO

The human helicase senataxin (SETX) has been linked to the neurodegenerative diseases amyotrophic lateral sclerosis (ALS4) and ataxia with oculomotor apraxia (AOA2). Here we identified a role for SETX in controlling the antiviral response. Cells that had undergone depletion of SETX and SETX-deficient cells derived from patients with AOA2 had higher expression of antiviral mediators in response to infection than did wild-type cells. Mechanistically, we propose a model whereby SETX attenuates the activity of RNA polymerase II (RNAPII) at genes stimulated after a virus is sensed and thus controls the magnitude of the host response to pathogens and the biogenesis of various RNA viruses (e.g., influenza A virus and West Nile virus). Our data indicate a potentially causal link among inborn errors in SETX, susceptibility to infection and the development of neurologic disorders.


Assuntos
Esclerose Lateral Amiotrófica/genética , Influenza Humana/imunologia , Orthomyxoviridae/fisiologia , RNA Helicases/metabolismo , RNA Polimerase II/metabolismo , Degenerações Espinocerebelares/genética , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Citocinas/metabolismo , DNA Helicases , Cães , Regulação para Baixo , Humanos , Imunidade Inata/genética , Fator Regulador 3 de Interferon/metabolismo , Células Madin Darby de Rim Canino , Camundongos , Camundongos Knockout , Análise em Microsséries , Enzimas Multifuncionais , RNA Helicases/genética , RNA Polimerase II/genética , RNA Interferente Pequeno/genética , Ataxias Espinocerebelares/congênito , Células Vero , Replicação Viral/genética
13.
Immunity ; 40(6): 880-95, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24882218

RESUMO

Type I interferons (IFN-I) are essential antiviral cytokines produced upon microbial infection. IFN-I elicits this activity through the upregulation of hundreds of IFN-I-stimulated genes (ISGs). The full breadth of ISG induction demands activation of a number of cellular factors including the IκB kinase epsilon (IKKε). However, the mechanism of IKKε activation upon IFN receptor signaling has remained elusive. Here we show that TRIM6, a member of the E3-ubiquitin ligase tripartite motif (TRIM) family of proteins, interacted with IKKε and promoted induction of IKKε-dependent ISGs. TRIM6 and the E2-ubiquitin conjugase UbE2K cooperated in the synthesis of unanchored K48-linked polyubiquitin chains, which activated IKKε for subsequent STAT1 phosphorylation. Our work attributes a previously unrecognized activating role of K48-linked unanchored polyubiquitin chains in kinase activation and identifies the UbE2K-TRIM6-ubiquitin axis as critical for IFN signaling and antiviral response.


Assuntos
Quinase I-kappa B/imunologia , Interferon Tipo I/imunologia , Poliubiquitina/biossíntese , Ubiquitina-Proteína Ligases/imunologia , Animais , Antivirais , Células Cultivadas , Ativação Enzimática/imunologia , Humanos , Janus Quinase 1 , Camundongos , Fosforilação/imunologia , Interferência de RNA , RNA Interferente Pequeno , Fator de Transcrição STAT1/imunologia , Transdução de Sinais/imunologia , Proteínas com Motivo Tripartido , Enzimas de Conjugação de Ubiquitina/imunologia , Ubiquitina-Proteína Ligases/genética
14.
PLoS One ; 9(2): e89284, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586659

RESUMO

Innate immune sensors such as Toll-like receptors (TLRs) differentially utilize adaptor proteins and additional molecular mediators to ensure robust and precise immune responses to pathogen challenge. Through a gain-of-function genetic screen, we identified the gamma catalytic subunit of protein phosphatase 1 (PP1-γ) as a positive regulator of MyD88-dependent proinflammatory innate immune activation. PP1-γ physically interacts with the E3 ubiquitin ligase TRAF6, and enhances the activity of TRAF6 towards itself and substrates such as IKKγ, whereas enzymatically inactive PP1-γ represses these events. Importantly, these activities were found to be critical for cellular innate responses to pathogen challenge and microbial clearance in both mouse macrophages and human monocyte lines. These data indicate that PP1-γ phosphatase activity regulates overall TRAF6 E3 ubiquitin ligase function and promotes NF-κB-mediated innate signaling responses.


Assuntos
Células Dendríticas/imunologia , Regulação da Expressão Gênica , Imunidade Inata , Macrófagos/imunologia , Proteína Fosfatase 1/fisiologia , Infecções Estreptocócicas/imunologia , Fator 6 Associado a Receptor de TNF/metabolismo , Animais , Western Blotting , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Ensaio de Imunoadsorção Enzimática , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunoprecipitação , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Mutação/genética , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus/patogenicidade , Fator 6 Associado a Receptor de TNF/antagonistas & inibidores , Fator 6 Associado a Receptor de TNF/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
15.
PLoS Pathog ; 9(3): e1003265, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23555265

RESUMO

An estimated 50 million dengue virus (DENV) infections occur annually and more than forty percent of the human population is currently at risk of developing dengue fever (DF) or dengue hemorrhagic fever (DHF). Despite the prevalence and potential severity of DF and DHF, there are no approved vaccines or antiviral therapeutics available. An improved understanding of DENV immune evasion is pivotal for the rational development of anti-DENV therapeutics. Antagonism of type I interferon (IFN-I) signaling is a crucial mechanism of DENV immune evasion. DENV NS5 protein inhibits IFN-I signaling by mediating proteasome-dependent STAT2 degradation. Only proteolytically-processed NS5 can efficiently mediate STAT2 degradation, though both unprocessed and processed NS5 bind STAT2. Here we identify UBR4, a 600-kDa member of the N-recognin family, as an interacting partner of DENV NS5 that preferentially binds to processed NS5. Our results also demonstrate that DENV NS5 bridges STAT2 and UBR4. Furthermore, we show that UBR4 promotes DENV-mediated STAT2 degradation, and most importantly, that UBR4 is necessary for efficient viral replication in IFN-I competent cells. Our data underscore the importance of NS5-mediated STAT2 degradation in DENV replication and identify UBR4 as a host protein that is specifically exploited by DENV to inhibit IFN-I signaling via STAT2 degradation.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Vírus da Dengue/fisiologia , Interferon Tipo I/metabolismo , Fator de Transcrição STAT2/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Vírus da Dengue/patogenicidade , Humanos , Evasão da Resposta Imune , Ligação Proteica , Transdução de Sinais , Ubiquitina-Proteína Ligases , Células Vero , Proteínas não Estruturais Virais/metabolismo
16.
Immunity ; 38(2): 384-98, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23438823

RESUMO

Innate immunity conferred by the type I interferon is critical for antiviral defense. To date only a limited number of tripartite motif (TRIM) proteins have been implicated in modulation of innate immunity and anti-microbial activity. Here we report the complementary DNA cloning and systematic analysis of all known 75 human TRIMs. We demonstrate that roughly half of the 75 TRIM-family members enhanced the innate immune response and that they do this at multiple levels in signaling pathways. Moreover, messenger RNA levels and localization of most of these TRIMs were found to be altered during viral infection, suggesting that their regulatory activities are highly controlled at both pre- and posttranscriptional levels. Taken together, our data demonstrate a very considerable dedication of this large protein family to the positive regulation of the antiviral response, which supports the notion that this family of proteins evolved as a component of innate immunity.


Assuntos
Proteínas de Transporte/genética , Imunidade Inata , Leucócitos Mononucleares/metabolismo , RNA Mensageiro/genética , Receptores de Reconhecimento de Padrão/genética , Infecções por Rhabdoviridae/metabolismo , Dedos de Zinco/genética , Processamento Alternativo , Fatores de Restrição Antivirais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/imunologia , Linhagem Celular , Clonagem Molecular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , RNA Mensageiro/imunologia , RNA Interferente Pequeno/genética , Receptores de Reconhecimento de Padrão/imunologia , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/virologia , Transdução de Sinais , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Vesiculovirus/imunologia , Dedos de Zinco/imunologia
17.
PLoS Pathog ; 8(10): e1002934, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23055924

RESUMO

Dengue virus (DENV) is a pathogen with a high impact on human health. It replicates in a wide range of cells involved in the immune response. To efficiently infect humans, DENV must evade or inhibit fundamental elements of the innate immune system, namely the type I interferon response. DENV circumvents the host immune response by expressing proteins that antagonize the cellular innate immunity. We have recently documented the inhibition of type I IFN production by the proteolytic activity of DENV NS2B3 protease complex in human monocyte derived dendritic cells (MDDCs). In the present report we identify the human adaptor molecule STING as a target of the NS2B3 protease complex. We characterize the mechanism of inhibition of type I IFN production in primary human MDDCs by this viral factor. Using different human and mouse primary cells lacking STING, we show enhanced DENV replication. Conversely, mutated versions of STING that cannot be cleaved by the DENV NS2B3 protease induced higher levels of type I IFN after infection with DENV. Additionally, we show that DENV NS2B3 is not able to degrade the mouse version of STING, a phenomenon that severely restricts the replication of DENV in mouse cells, suggesting that STING plays a key role in the inhibition of DENV infection and spread in mice.


Assuntos
Células Dendríticas/metabolismo , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Interferon Tipo I/biossíntese , Proteínas de Membrana/metabolismo , Proteínas não Estruturais Virais/metabolismo , Aedes , Animais , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Células Dendríticas/virologia , Vírus da Dengue/metabolismo , Células HEK293 , Humanos , Evasão da Resposta Imune , Macrófagos/metabolismo , Macrófagos/virologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Células Vero , Replicação Viral
18.
PLoS One ; 7(10): e46768, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056444

RESUMO

Infectious bursal disease virus (IBDV) is an avian pathogen responsible for an acute immunosuppressive disease that causes major losses to the poultry industry. Despite having a bipartite dsRNA genome, IBDV, as well as other members of the Birnaviridae family, possesses a single capsid layer formed by trimers of the VP2 capsid protein. The capsid encloses a ribonucleoprotein complex formed by the genome associated to the RNA-dependent RNA polymerase and the RNA-binding polypeptide VP3. A previous report evidenced that expression of the mature VP2 IBDV capsid polypeptide triggers a swift programmed cell death response in a wide variety of cell lines. The mechanism(s) underlying this effect remained unknown. Here, we show that VP2 expression in HeLa cells activates the double-stranded RNA (dsRNA)-dependent protein kinase (PKR), which in turn triggers the phosphorylation of the eukaryotic initiation factor 2α (eIF2α). This results in a strong blockade of protein synthesis and the activation of an apoptotic response which is efficiently blocked by coexpression of a dominant negative PKR polypeptide. Our results demonstrate that coexpression of the VP3 polypeptide precludes phosphorylation of both PKR and eIF2α and the onset of programmed cell death induced by VP2 expression. A mutation blocking the capacity of VP3 to bind dsRNA also abolishes its capacity to prevent PKR activation and apoptosis. Further experiments showed that VP3 functionally replaces the host-range vaccinia virus (VACV) E3 protein, thus allowing the E3 deficient VACV deletion mutant WRΔE3L to grow in non-permissive cell lines. According to results presented here, VP3 can be categorized along with other well characterized proteins such us VACV E3, avian reovirus sigmaA, and influenza virus NS1 as a virus-encoded dsRNA-binding polypeptide with antiapoptotic properties. Our results suggest that VP3 plays a central role in ensuring the viability of the IBDV replication cycle by preventing programmed cell death.


Assuntos
Apoptose/fisiologia , Proteínas do Capsídeo/metabolismo , Vírus da Doença Infecciosa da Bursa/metabolismo , Vírus da Doença Infecciosa da Bursa/patogenicidade , Apoptose/genética , Western Blotting , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Células HeLa , Humanos , Vírus da Doença Infecciosa da Bursa/genética , Fosforilação/genética , Fosforilação/fisiologia , RNA de Cadeia Dupla/genética , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , eIF-2 Quinase
19.
Cell Host Microbe ; 11(3): 306-18, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22423970

RESUMO

Pathogens commonly utilize endocytic pathways to gain cellular access. The endosomal pattern recognition receptors TLR7 and TLR9 detect pathogen-encoded nucleic acids to initiate MyD88-dependent proinflammatory responses to microbial infection. Using genome-wide RNAi screening and integrative systems-based analysis, we identify 190 cofactors required for TLR7- and TLR9-directed signaling responses. A set of cofactors were crossprofiled for their activities downstream of several immunoreceptors and then functionally mapped based on the known architecture of NF-κB signaling pathways. Protein complexes and pathways involved in ubiquitin-protein ligase activities, sphingolipid metabolism, chromatin modifications, and ancient stress responses were found to modulate innate recognition of endosomal nucleic acids. Additionally, hepatocyte growth factor-regulated tyrosine kinase substrate (HRS) was characterized as necessary for ubiquitin-dependent TLR9 targeting to the endolysosome. Proteins and pathways identified here should prove useful in delineating strategies to manipulate innate responses for treatment of autoimmune disorders and microbial infection.


Assuntos
Imunidade Inata/genética , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Animais , Embrião de Galinha , Simulação por Computador , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Endossomos/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Biológicos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Transporte Proteico , Interferência de RNA , Transdução de Sinais , Máquina de Vetores de Suporte
20.
PLoS One ; 6(6): e20972, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21698249

RESUMO

Toroviruses are gastroenteritis causing agents that infect different animal species and humans. To date, very little is known about how toroviruses cause disease. Here, we describe for the first time that the prototype member of this genus, the equine torovirus Berne virus (BEV), induces apoptosis in infected cells at late times postinfection. Observation of BEV infected cells by electron microscopy revealed that by 24 hours postinfection some cells exhibited morphological characteristics of apoptotic cells. Based on this finding, we analyzed several apoptotic markers, and observed protein synthesis inhibition, rRNA and DNA degradation, nuclear fragmentation, caspase-mediated cleavage of PARP and eIF4GI, and PKR and eIF2α phosphorylation, all these processes taking place after peak virus production. We also determined that both cell death receptor and mitochondrial pathways are involved in the apoptosis process induced by BEV. BEV-induced apoptosis at late times postinfection, once viral progeny are produced, could facilitate viral dissemination in vivo and contribute to viral pathogenesis.


Assuntos
Apoptose/fisiologia , Caspases/fisiologia , Torovirus/fisiologia , Animais , Western Blotting , Linhagem Celular , Citometria de Fluxo , Microscopia Eletrônica , Fosforilação , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...