Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Virol ; 91(17)2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28637751

RESUMO

Understanding the interactions between rabies virus (RABV) and individual host cell proteins is critical for the development of targeted therapies. Here we report that interferon-induced protein with tetratricopeptide repeats 2 (Ifit2), an interferon-stimulated gene (ISG) with possible RNA-binding capacity, is an important restriction factor for rabies virus. When Ifit2 was depleted, RABV grew more quickly in mouse neuroblastoma cells in vitro This effect was replicated in vivo, where Ifit2 knockout mice displayed a dramatically more severe disease phenotype than wild-type mice after intranasal inoculation of RABV. This increase in pathogenicity correlated to an increase in RABV mRNA and live viral load in the brain, as well as to an accelerated spread to brain regions normally affected by this RABV model. These results suggest that Ifit2 exerts its antiviral effect mainly at the level of viral replication, as opposed to functioning as a mechanism that restricts viral entry/egress or transports RABV particles through axons.IMPORTANCE Rabies is a fatal zoonotic disease with a nearly 100% case fatality rate. Although there are effective vaccines for rabies, this disease still takes the lives of about 50,000 people each year. Victims tend to be children living in regions without comprehensive medical infrastructure who present to health care workers too late for postexposure prophylaxis. The protein discussed in our report, Ifit2, is found to be an important restriction factor for rabies virus, acting directly or indirectly against viral replication. A more nuanced understanding of this interaction may reveal a step of a pathway or site at which the system could be exploited for the development of a targeted therapy.


Assuntos
Encéfalo/virologia , Proteínas/metabolismo , Vírus da Raiva/patogenicidade , Raiva/patologia , Animais , Proteínas Reguladoras de Apoptose , Encéfalo/patologia , Linhagem Celular Tumoral , Feminino , Interferons/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroblastoma/virologia , Proteínas/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA , Raiva/virologia , Vírus da Raiva/fisiologia , Virulência , Replicação Viral
2.
EMBO Rep ; 16(11): 1535-47, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26341626

RESUMO

Mammalian Toll-like receptors (TLR) recognize microbial products and elicit transient immune responses that protect the infected host from disease. TLR4--which signals from both plasma and endosomal membranes--is activated by bacterial lipopolysaccharides (LPS) and induces many cytokine genes, the prolonged expression of which causes septic shock in mice. We report here that the expression of some TLR4-induced genes in myeloid cells requires the protein kinase activity of the epidermal growth factor receptor (EGFR). EGFR inhibition affects TLR4-induced responses differently depending on the target gene. The induction of interferon-ß (IFN-ß) and IFN-inducible genes is strongly inhibited, whereas TNF-α induction is enhanced. Inhibition is specific to the IFN-regulatory factor (IRF)-driven genes because EGFR is required for IRF activation downstream of TLR--as is IRF co-activator ß-catenin--through the PI3 kinase/AKT pathway. Administration of an EGFR inhibitor to mice protects them from LPS-induced septic shock and death by selectively blocking the IFN branch of TLR4 signaling. These results demonstrate a selective regulation of TLR4 signaling by EGFR and highlight the potential use of EGFR inhibitors to treat septic shock syndrome.


Assuntos
Receptores ErbB/metabolismo , Choque Séptico/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Animais , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Gefitinibe , Fatores Reguladores de Interferon/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Lipopolissacarídeos/administração & dosagem , Camundongos , Análise em Microsséries , Células Mieloides/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Células RAW 264.7 , Choque Séptico/prevenção & controle , Choque Séptico/terapia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
3.
Annu Rev Virol ; 2(1): 549-72, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26958928

RESUMO

The interferon system protects mammals against virus infections. There are several types of interferons, which are characterized by their ability to inhibit virus replication and resultant pathogenesis by triggering both innate and cell-mediated immune responses. Virus infection is sensed by a variety of cellular pattern-recognition receptors and triggers the synthesis of interferons, which are secreted by the infected cells. In uninfected cells, cell surface receptors recognize the secreted interferons and activate intracellular signaling pathways that induce the expression of interferon-stimulated genes; the proteins encoded by these genes inhibit different stages of virus replication. To avoid extinction, almost all viruses have evolved mechanisms to defend themselves against the interferon system. Consequently, a dynamic equilibrium of survival is established between the virus and its host, an equilibrium that can be shifted to the host's favor by the use of exogenous interferon as a therapeutic antiviral agent.


Assuntos
Interferons/imunologia , Viroses/imunologia , Vírus/imunologia , Animais , Humanos , Imunidade Inata , Interferons/genética , Viroses/genética , Viroses/virologia , Vírus/genética
4.
J Virol ; 89(5): 2462-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25428874

RESUMO

A major component of the protective antiviral host defense is contributed by the intracellular actions of the proteins encoded by interferon-stimulated genes (ISGs); among these are the interferon-induced proteins with tetratricopeptide repeats (IFITs), consisting of four members in human and three in mouse. IFIT proteins do not have any known enzyme activity. Instead, they inhibit virus replication by binding and regulating the functions of cellular and viral proteins and RNAs. Although all IFITs are comprised of multiple copies of the degenerate tetratricopeptide repeats, their distinct tertiary structures enable them to bind different partners and affect host-virus interactions differently. The recent use of Ifit knockout mouse models has revealed novel antiviral functions of these proteins and new insights into the specificities of ISG actions. This article focuses on human and murine IFIT1 and IFIT2 by reviewing their mechanisms of action, their critical roles in protecting mice from viral pathogenesis, and viral strategies to evade IFIT action.


Assuntos
Antivirais/metabolismo , Regulação da Expressão Gênica , Interferons/imunologia , Biossíntese de Proteínas , Proteínas/metabolismo , Vírus/imunologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Interferons/metabolismo , Camundongos
5.
J Virol ; 88(23): 13593-601, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25231314

RESUMO

UNLABELLED: The type I/III interferon (IFN) system has major roles in regulating viral pathogenesis, usually ameliorating pathogenesis by impairing virus replication through the antiviral actions of one or more IFN-induced proteins. Ifit2 is one such protein which can be induced by IFN or virus infection, and it is responsible for protecting mice from neuropathogenesis caused by vesicular stomatitis virus. Here, we show that Ifit2 also protects mice from pathogenesis caused by the respirovirus Sendai virus (SeV). Mice lacking Ifit2 (Ifit2(-/-)) suffered severe weight loss and succumbed to intranasal infection with SeV strain 52 at a dose that killed only a few wild-type mice. Viral RNA was detectable only in lungs, and SeV titers were higher in Ifit2(-/-) mice than in wild-type mice. Similar infiltration of immune cells was found in the lungs of both mouse lines, corresponding to similar levels of many induced cytokines and chemokines. In contrast, IFN-ß and IFN-λ3 expression were considerably higher in the lungs of Ifit2(-/-) mice. Surprisingly, type I IFN receptor knockout (IFNAR(-/-)) mice were less susceptible to SeV than Ifit2(-/-) mice, although their pulmonary virus titers were similarly high. To test the intriguing possibility that type I IFN action enhances pathogenesis in the context of elevated SeV replication in lungs, we generated Ifit2/IFNAR(-/-) double knockout mice. These mice were less susceptible to SeV than Ifit2(-/-) mice, although viral titers in their lungs were even higher. Our results indicate that high SeV replication in the lungs of infected Ifit2(-/-) mice cooperates with elevated IFN-ß induction to cause disease. IMPORTANCE: The IFN system is an innate defense against virus infections. It is triggered quickly in infected cells, which then secrete IFN. Via their cell surface receptors on surrounding cells, they induce transcription of numerous IFN-stimulated genes (ISG), which in turn protect these cells by inhibiting virus life cycles. Hence, IFNs are commonly considered beneficial during virus infections. Here, we report two key findings. First, lack of a single ISG in mice, Ifit2, resulted in high mortality after SeV infection of the respiratory tract, following higher virus loads and higher IFN production in Ifit2(-/-) lungs. Second, mortality of Ifit2(-/-) mice was reduced when mice also lacked the type I IFN receptor, while SeV loads in lungs still were high. This indicates that type I IFN exacerbates pathogenesis in the SeV model, and that limitation of both viral replication and IFN production is needed for effective prevention of disease.


Assuntos
Interações Hospedeiro-Patógeno , Interferons/metabolismo , Proteínas/metabolismo , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/patologia , Vírus Sendai/imunologia , Animais , Proteínas Reguladoras de Apoptose , Feminino , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas/genética , Proteínas de Ligação a RNA , Receptor de Interferon alfa e beta/deficiência , Infecções por Respirovirus/virologia , Análise de Sobrevida , Carga Viral
6.
J Virol ; 88(18): 10303-11, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24991014

RESUMO

UNLABELLED: The interferon system provides the first line of host defense against virus infection. Mouse pathogenesis studies have revealed the importance of specific interferon-induced proteins in providing protection against specific viruses. We have previously reported that one such protein, Ifit2, protects neurons of the central nervous system from intranasal infection by the neurotropic rhabdovirus, vesicular stomatitis virus (VSV). Here, we demonstrate that Ifit2 protects the peripheral nervous system from VSV infection as well. In Ifit2(-/-) mice, VSV, injected subcutaneously into the footpad, entered the proximal lymph node, where it replicated and infected the nodal nerve endings. The infection spread to the sciatic nerve, the spinal cord, and the brain, causing paralysis. In contrast, in the wild-type mice, although VSV replicated equally well in the lymph node, infection of the sciatic nerve and the rest of the nervous system was impaired, thus preventing paralysis. Ifit2 protected only the nervous system from VSV infection; other tissues were well protected even in Ifit2(-/-) mice. These results indicate that Ifit2 is the interferon-induced protein that prevents VSV infection of neurons of both the peripheral and the central nervous systems, thus inhibiting the consequent neuropathy, but it is dispensable for protecting the cells of other tissues from VSV infection. IMPORTANCE: Although viral infection is quite common, the immune system effectively protects us from viral diseases. A major part of this protection is mediated by interferon, the antiviral cytokine secreted by virus-infected cells. To empower the neighboring uninfected cells in combating the oncoming infection, interferon induces the synthesis of more than 200 new proteins, many of which have antiviral activities. The virus studied here, vesicular stomatitis virus (VSV), like its relative, rabies virus, can cause neuropathy in mice if it enters the peripheral nervous system through skin lesions; however, interferon can protect neurons from VSV infection. We have identified a specific interferon-induced protein, Ifit2, as the protein that protects neurons from VSV infection. Surprisingly, Ifit2 was not needed to protect other cell types from VSV. Our results indicate that the effector antiviral proteins of the interferon system have highly specialized functions.


Assuntos
Sistema Nervoso Periférico/virologia , Proteínas/imunologia , Doenças dos Roedores/prevenção & controle , Estomatite Vesicular/prevenção & controle , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Encéfalo/imunologia , Encéfalo/virologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/imunologia , Neurônios/virologia , Sistema Nervoso Periférico/imunologia , Proteínas/genética , Proteínas de Ligação a RNA , Doenças dos Roedores/genética , Doenças dos Roedores/imunologia , Doenças dos Roedores/virologia , Estomatite Vesicular/genética , Estomatite Vesicular/imunologia , Estomatite Vesicular/virologia
7.
J Virol ; 88(2): 1051-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24198415

RESUMO

Type I interferons (IFN-α/ß) limit viral dissemination prior to the emergence of adaptive immune responses through the concerted action of interferon-stimulated genes (ISGs). Although IFN-α/ß induction by coronaviruses is modest, it effectively limits viral spread within the central nervous system (CNS) and protects against mortality. The protective roles of specific ISGs against the mouse hepatitis virus (MHV) members of the coronaviruses are largely unknown. This study demonstrates a protective role of the ISG Ifit2 in encephalitis induced by the dual hepato- and neurotropic MHV-A59. Contrasting the mild encephalitis and 100% survival of MHV-A59-infected wild-type (wt) mice, nearly 60% of infected Ifit2(-/-) mice exhibited severe encephalitis and succumbed between 6 and 8 days postinfection. Increased clinical disease in Ifit2(-/-) mice coincided with higher viral loads and enhanced viral spread throughout the CNS parenchyma. Ifit2(-/-) mice also expressed significantly reduced IFN-α/ß and downstream ISG mRNAs Ifit1, Isg15, and Pkr, while expression of proinflammatory cytokines and chemokines was only modestly affected in the CNS. Impaired IFN-α/ß induction in the absence of Ifit2 was confirmed by ex vivo mRNA analysis of microglia and macrophages, the prominent cell types producing IFN-α/ß following MHV CNS infection. Furthermore, both IFN-α/ß mRNA and protein production were significantly reduced in MHV-infected Ifit2(-/-) relative to wt bone marrow-derived macrophages. Collectively, the data implicate Ifit2 as a positive regulator of IFN-α/ß expression, rather than direct antiviral mediator, during MHV-induced encephalitis.


Assuntos
Sistema Nervoso Central/virologia , Encefalite/veterinária , Interferon-alfa/genética , Interferon beta/genética , Macrófagos/imunologia , Vírus da Hepatite Murina/fisiologia , Proteínas/imunologia , Doenças dos Roedores/imunologia , Animais , Proteínas Reguladoras de Apoptose , Sistema Nervoso Central/imunologia , Encefalite/genética , Encefalite/imunologia , Encefalite/virologia , Feminino , Interferon-alfa/imunologia , Interferon beta/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/genética , Proteínas/genética , Proteínas de Ligação a RNA , Doenças dos Roedores/genética , Doenças dos Roedores/virologia , Tropismo Viral
8.
mBio ; 4(2)2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23532979

RESUMO

Interferon (IFN) is required for protecting mice from viral pathogenesis; reciprocally, it mediates the deleterious septic shock response to bacterial infection. The critical transcription factor for IFN induction, in both cases, is IRF-3, which is activated by TLR3 or RIG-I signaling in response to virus infection and TLR4 signaling in response to bacterial infection. Here, we report that IRF-3's transcriptional activity required its coactivators, ß-catenin and CBP, to be modified by HDAC6-mediated deacetylation and protein kinase C isozyme ß (PKC-ß)-mediated phosphorylation, respectively, so that activated nuclear IRF-3 could form a stable transcription initiation complex at the target gene promoters. ß-Catenin bridges IRF-3 and CBP, and the modifications were required specifically for the interaction between ß-catenin and CBP but not ß-catenin and IRF-3. Consequently, like IRF-3(-/-) mice, HDAC6(-/-) mice were resistant to bacterial lipopolysaccharide-induced septic shock. Conversely, they were highly susceptible to pathogenesis caused by Sendai virus infection. Thus, HDAC6 is an essential component of the innate immune response to microbial infection.


Assuntos
Infecções Bacterianas/imunologia , Proteína de Ligação a CREB/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Vírus Sendai/imunologia , Choque Séptico , beta Catenina/metabolismo , Acetilação , Animais , Desacetilase 6 de Histona , Histona Desacetilases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Processamento de Proteína Pós-Traducional
9.
J Virol ; 87(1): 16-24, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23077293

RESUMO

Infection of cultured cells by paramyxoviruses causes cell death, mediated by a newly discovered apoptotic pathway activated by virus infection. The key proapoptotic protein in this pathway is interferon regulatory factor 3 (IRF-3), which upon activation by virus infection binds BAX, translocates it to mitochondria, and triggers apoptosis. When IRF-3-knockdown cells were infected with Sendai virus (SeV), persistent infection (PI) was established. The PI cells produced infectious SeV continuously and constitutively expressed many innate immune genes. Interferon signaling was blocked in these cells. The elevated levels of IRF-3-driven genes in the PI cells indicated that the amount of residual IRF-3 activated by endogenous SeV was high enough to drive the transcriptional effects of IRF-3 but too low to trigger its apoptotic activity. We confirmed this IRF-3 threshold idea by generating a tetracycline (Tet)-inducible cell line for IRF-3 expression, which enabled us to express various levels of IRF-3. PI could be established in the Tet-off cell line, and as expected, when doxycycline was withdrawn, the cells underwent apoptosis. Finally, we tested for PI establishment in 12 mouse embryo fibroblasts by natural selection. Eleven lines became persistently infected; although seven out of them had low IRF-3 levels, four did not. When one of the latter four was further analyzed, we observed that it expressed a very low level of caspase 3, the final executor protease of the apoptotic pathway. These results demonstrated that SeV PI can arise from infection of normal wild-type cells, but only if they can find a way to impair the IRF-3-dependent apoptotic pathway.


Assuntos
Apoptose , Interações Hospedeiro-Patógeno , Fator Regulador 3 de Interferon/metabolismo , Vírus Sendai/patogenicidade , Animais , Linhagem Celular , Fibroblastos/virologia , Humanos , Camundongos , Replicação Viral
10.
Cell Res ; 22(10): 1407-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22964712

RESUMO

Interferon carries out its cellular effects, including its antiviral effects, by inducing the synthesis of many new proteins, amongst which is the IFIT (ISG56) family of proteins. The first crystal structure of an IFIT, reported by Yang et al., revealed several functional properties of the protein that may help us to better understand the biological functions of these proteins.


Assuntos
Proteínas/química , Proteínas/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Cristalografia por Raios X , Humanos , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína , RNA/metabolismo , Proteínas de Ligação a RNA , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
11.
Sci Signal ; 5(233): ra50, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22810896

RESUMO

Toll-like receptors (TLRs) recognize specific microbial products and elicit innate immune signals to activate specific transcription factors that induce protective proteins, such as interferon. TLR3 is localized to endosomes and recognizes double-stranded RNA (dsRNA), which is generated by virally infected or apoptotic cells. TLR3 has been genetically linked to several human diseases, including some without viral etiology. Unlike other TLRs, TLR3 requires phosphorylation of two specific tyrosine residues in its cytoplasmic domain to recruit the adaptor protein TRIF (Toll-interleukin-1 receptor domain-containing adaptor protein inducing interferon-ß) and initiate the antiviral response. We showed that two protein tyrosine kinases, the epidermal growth factor receptor (EGFR) ErbB1 and Src, bound sequentially to dsRNA-activated TLR3 and phosphorylated the two tyrosine residues. In cells lacking EGFR or treated with an inhibitor of EGFR, viral replication was enhanced and induction of antiviral genes was impaired. Thus, these results reveal a connection between antiviral innate immunity and cell growth regulators.


Assuntos
Receptores ErbB/metabolismo , Imunidade Inata/fisiologia , Transdução de Sinais/fisiologia , Receptor 3 Toll-Like/metabolismo , Viroses/imunologia , Quinases da Família src/metabolismo , Animais , Western Blotting , Linhagem Celular , Primers do DNA/genética , Endossomos/metabolismo , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/genética , Vetores Genéticos , Humanos , Imunidade Inata/genética , Imunoprecipitação , Camundongos , Camundongos Knockout , Análise em Microsséries , Microscopia Confocal , Fosforilação , Plasmídeos/genética , Reação em Cadeia da Polimerase , Interferência de RNA , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Transdução de Sinais/genética , Receptor 3 Toll-Like/genética
12.
PLoS Pathog ; 8(5): e1002712, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615570

RESUMO

Interferon protects mice from vesicular stomatitis virus (VSV) infection and pathogenesis; however, it is not known which of the numerous interferon-stimulated genes (ISG) mediate the antiviral effect. A prominent family of ISGs is the interferon-induced with tetratricopeptide repeats (Ifit) genes comprising three members in mice, Ifit1/ISG56, Ifit2/ISG54 and Ifit3/ISG49. Intranasal infection with a low dose of VSV is not lethal to wild-type mice and all three Ifit genes are induced in the central nervous system of the infected mice. We tested their potential contributions to the observed protection of wild-type mice from VSV pathogenesis, by taking advantage of the newly generated knockout mice lacking either Ifit2 or Ifit1. We observed that in Ifit2 knockout (Ifit2(-/-)) mice, intranasal VSV infection was uniformly lethal and death was preceded by neurological signs, such as ataxia and hind limb paralysis. In contrast, wild-type and Ifit1(-/-) mice were highly protected and survived without developing such disease. However, when VSV was injected intracranially, virus replication and survival were not significantly different between wild-type and Ifit2(-/-) mice. When administered intranasally, VSV entered the central nervous system through the olfactory bulbs, where it replicated equivalently in wild-type and Ifit2(-/-) mice and induced interferon-ß. However, as the infection spread to other regions of the brain, VSV titers rose several hundred folds higher in Ifit2(-/-) mice as compared to wild-type mice. This was not caused by a broadened cell tropism in the brains of Ifit2(-/-) mice, where VSV still replicated selectively in neurons. Surprisingly, this advantage for VSV replication in the brains of Ifit2(-/-) mice was not observed in other organs, such as lung and liver. Pathogenesis by another neurotropic RNA virus, encephalomyocarditis virus, was not enhanced in the brains of Ifit2(-/-) mice. Our study provides a clear demonstration of tissue-, virus- and ISG-specific antiviral action of interferon.


Assuntos
Encéfalo/virologia , Proteínas/metabolismo , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Vírus da Encefalomiocardite/imunologia , Vírus da Encefalomiocardite/patogenicidade , Feminino , Interferon beta/metabolismo , Fígado/virologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas/genética , Proteínas de Ligação a RNA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Estomatite Vesicular/patologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia , Replicação Viral
13.
J Immunol ; 188(6): 2825-33, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22323545

RESUMO

dsRNA is a common pathogen-associated molecular pattern that is recognized by cellular TLR3 and used by virus-infected cells to activate specific transcription factors and trigger induction of antiviral genes. In this article, we report a new branch of TLR3 signaling that does not lead to gene induction but affects many cellular properties, such as cell migration, adhesion, and proliferation. We demonstrated that the migration of multiple cell lineages was affected by dsRNA treatment or influenza virus infection in a TLR3-dependent fashion. Surprisingly, for this effect of TLR3 signaling, the adaptor proteins, TRIF and MyD88, were not required. The effects of the new pathway were mediated by the proto-oncoprotein c-Src, which bound to TLR3 after dsRNA stimulation of cells. The response was biphasic: upon dsRNA treatment, we observed an immediate increase in cell motility followed by its strong inhibition. Our results indicate that the first phase was mediated by dsRNA-induced phosphorylation and activation of Src, whereas the second phase resulted from the sequestration of activated Src in lipid rafts, thus decreasing its active cytoplasmic pool. As expected, two other functions of Src, its effect on cell adhesion and cell proliferation, were also inhibited by dsRNA treatment. These results demonstrate that activated TLR3 can engage Src to trigger multiple cellular effects and reveal a possible link between innate immune response and cell growth regulation. This study also provides a rare example of TLR-mediated cellular effects that do not require gene induction and the first example, to our knowledge, of an adaptor-independent effect of any TLR.


Assuntos
Transdução de Sinais/imunologia , Receptor 3 Toll-Like/metabolismo , Viroses/imunologia , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Adesão Celular/imunologia , Movimento Celular/imunologia , Proliferação de Células , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/metabolismo , Camundongos , RNA de Cadeia Dupla/imunologia , RNA Viral/imunologia , Receptor 3 Toll-Like/imunologia
14.
J Interferon Cytokine Res ; 31(1): 71-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20950130

RESUMO

The ISG56/IFIT1 family of genes is clustered on human chromosome 10 and is comprised of 4 members, ISG56/IFIT1, ISG54/IFIT2, ISG60/IFIT3, and ISG58/IFIT5, whose homologs are evolutionarily conserved from mammals to amphibians. While these genes are normally silent in most cell types, their transcription is strongly induced by interferons, virus infection, and molecular patterns such as double-stranded RNA or lipopolysaccharides. The encoded P56 family proteins are characterized by multiple repeats of tetratricopeptide repeat helix-turn-helix motifs mediating a variety of protein-protein interactions, which result in a multitude of effects on cellular and viral functions, such as translation initiation, virus replication, double-stranded RNA signaling, cell migration, and proliferation.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , Interferons/metabolismo , Família Multigênica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Humanos , Imunidade Inata , Domínios e Motivos de Interação entre Proteínas , Proteínas de Ligação a RNA , Ativação Transcricional
15.
Nature ; 468(7322): 452-6, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21085181

RESUMO

Cellular messenger RNA (mRNA) of higher eukaryotes and many viral RNAs are methylated at the N-7 and 2'-O positions of the 5' guanosine cap by specific nuclear and cytoplasmic methyltransferases (MTases), respectively. Whereas N-7 methylation is essential for RNA translation and stability, the function of 2'-O methylation has remained uncertain since its discovery 35 years ago. Here we show that a West Nile virus (WNV) mutant (E218A) that lacks 2'-O MTase activity was attenuated in wild-type primary cells and mice but was pathogenic in the absence of type I interferon (IFN) signalling. 2'-O methylation of viral RNA did not affect IFN induction in WNV-infected fibroblasts but instead modulated the antiviral effects of IFN-induced proteins with tetratricopeptide repeats (IFIT), which are interferon-stimulated genes (ISGs) implicated in regulation of protein translation. Poxvirus and coronavirus mutants that lacked 2'-O MTase activity similarly showed enhanced sensitivity to the antiviral actions of IFN and, specifically, IFIT proteins. Our results demonstrate that the 2'-O methylation of the 5' cap of viral RNA functions to subvert innate host antiviral responses through escape of IFIT-mediated suppression, and suggest an evolutionary explanation for 2'-O methylation of cellular mRNA: to distinguish self from non-self RNA. Differential methylation of cytoplasmic RNA probably serves as an example for pattern recognition and restriction of propagation of foreign viral RNA in host cells.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/imunologia , Imunidade Inata/imunologia , Interferons/imunologia , Proteínas/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Células Cultivadas , Coronavirus/enzimologia , Coronavirus/genética , Coronavirus/imunologia , Coronavirus/fisiologia , Fibroblastos , Regulação da Expressão Gênica/genética , Humanos , Imunidade Inata/genética , Interferons/deficiência , Interferons/genética , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Modelos Imunológicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Poxviridae/enzimologia , Poxviridae/genética , Poxviridae/imunologia , Poxviridae/fisiologia , Biossíntese de Proteínas/imunologia , Proteínas/genética , Capuzes de RNA/genética , Capuzes de RNA/imunologia , RNA Viral/genética , RNA Viral/imunologia , Proteínas de Ligação a RNA , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Taxa de Sobrevida , Replicação Viral , Vírus do Nilo Ocidental/enzimologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia
16.
J Virol ; 84(24): 13036-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926571

RESUMO

The interferon (IFN)-induced protein P56 inhibits human papillomavirus (HPV) DNA replication by binding to HPV E1, which has several distinct functions in initiating viral DNA replication. Here, we determined that P56 inhibited HPV type 18 (HPV18) E1's DNA helicase activity, E2 binding, and HPV Ori sequence-specific DNA binding but not nonspecific DNA binding. We observed that deletion of a single amino acid, F399, produced an E1 mutant that could not bind P56. This E1 mutant retained its ability to support Ori DNA replication, but this activity was not inhibited by IFN, demonstrating that P56 is the principal executor of the anti-HPV action of IFN.


Assuntos
Antivirais/farmacologia , DNA Helicases/antagonistas & inibidores , Replicação do DNA/efeitos dos fármacos , Interferons/farmacologia , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , DNA Helicases/metabolismo , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Oncogênicas Virais/antagonistas & inibidores , Proteínas Oncogênicas Virais/genética , Papillomaviridae/fisiologia , Conformação Proteica , Proteínas de Ligação a RNA , Origem de Replicação/efeitos dos fármacos , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/genética
17.
Biofactors ; 35(1): 14-20, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19319841

RESUMO

Interferons represent a family of cytokines, which is of central importance in the innate immune response to virus infections. All interferons act as secreted ligands of specific cell surface receptors, eliciting the transcription of hundreds of interferon-stimulated genes whose protein products have antiviral activity, as well as antimicrobial, antiproliferative/antitumor, and immunomodulatory effects. Expression of type I and III interferons is induced in virtually all cell types upon recognition of viral molecular patterns, especially nucleic acids, by cytoplasmic and endosomal receptors, whereas type II interferon is induced by cytokines such as IL-12, and its expression is restricted to immune cells such as T cells and NK cells. The effectiveness of the interferon system in counteracting viral infections is reflected by the multitude of inhibitors of interferon induction or interferon action that are encoded by many viruses, preventing their eradication and resulting in the continued coexistence of viruses and vertebrates. The unique biological functions of interferons have led to their therapeutic use in the treatment of diseases such as hepatitis, multiple sclerosis, and certain leukemias.


Assuntos
Interferons/metabolismo , Viroses/metabolismo , Animais , Humanos , Modelos Biológicos , Transdução de Sinais
18.
J Virol ; 82(22): 11045-53, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18768971

RESUMO

The interferon-stimulated gene 56 (ISG56) family is induced strongly in response to virus infection, interferons (IFNs) and double-stranded RNA (dsRNA). In the mouse, this family comprises three members, ISG56, ISG54, and ISG49, which are clustered on chromosome 19 and encode the corresponding proteins p56, p54, and p49. Here, we report differential properties of these proteins and their distinct induction patterns in different cell types. All three murine proteins bound to the c-subunit of the translation initiation factor eIF3, but unlike the other members, p49 did not inhibit protein synthesis. Using a newly raised antibody, we demonstrated that both in vitro and in vivo, p49 expression was strongly induced by IFN, dsRNA, and Sendai virus. However, in kidney mesangial cells, as opposed to podocytes, encephalomyocarditis virus, vesicular stomatitis virus, or extracellular dsRNA did not induce any of the p56 family proteins, although they were robustly expressed after Sendai virus infection or dsRNA transfection. Furthermore, protein-specific differences in the regulation of p56 family members became evident in various leukocyte types: all three proteins were induced by IFN in T cells, but in B cells p56 and ISG56 mRNA could not be detected. Similarly, p56 was selectively uninducible in plasmacytoid dendritic cells, whereas in myeloid dendritic cells, all three family members were expressed. These results revealed novel cell type-, inducer-, and gene-specific regulation of the ISG56 family of genes.


Assuntos
Fator de Iniciação 3 em Eucariotos/antagonistas & inibidores , Biossíntese de Proteínas , Fatores de Transcrição/biossíntese , Fatores de Transcrição/fisiologia , Animais , Linfócitos B/metabolismo , Linhagem Celular , Células Cultivadas , Células Dendríticas/metabolismo , Vírus da Encefalomiocardite/fisiologia , Fator de Iniciação 3 em Eucariotos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Interferons/metabolismo , Células Mesangiais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Podócitos/virologia , Ligação Proteica , RNA de Cadeia Dupla/metabolismo , Vírus Sendai/fisiologia , Linfócitos T/metabolismo , Vírus da Estomatite Vesicular Indiana/fisiologia
19.
J Virol ; 79(17): 10968-77, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16103148

RESUMO

Hepatitis A virus (HAV) antagonizes the innate immune response by inhibition of double-stranded RNA (dsRNA)-induced beta interferon (IFN-beta) gene expression. In this report, we show that this is due to an interaction of HAV with the intracellular dsRNA-induced retinoic acid-inducible gene I (RIG-I)-mediated signaling pathway upstream of the kinases responsible for interferon regulatory factor 3 (IRF-3) phosphorylation (TBK1 and IKKepsilon). In consequence, IRF-3 is not activated for nuclear translocation and gene induction. In addition, we found that HAV reduces TRIF (TIR domain-containing adaptor inducing IFN-beta)-mediated IRF-3 activation, which is part of the Toll-like receptor 3 signaling pathway. As IRF-3 is necessary for IFN-beta transcription, inhibition of this factor results in efficient suppression of IFN-beta synthesis. This ability of HAV seems to be of considerable importance for HAV replication, as HAV is not resistant to IFN-beta, and it may allow the virus to establish infection and preserve the sites of virus production in later stages of the infection.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Vírus da Hepatite A/patogenicidade , Hepatite A/imunologia , Interferon beta/antagonistas & inibidores , RNA Helicases/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Proteína DEAD-box 58 , RNA Helicases DEAD-box , Hepatite A/metabolismo , Vírus da Hepatite A/crescimento & desenvolvimento , Humanos , Quinase I-kappa B , Fator Regulador 3 de Interferon , Interferon beta/genética , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos , Receptor 3 Toll-Like , Receptores Toll-Like , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA