Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Virol ; 97(10): e0095923, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37772825

RESUMO

IMPORTANCE: Viral encephalomyelitis outcome is dependent on host responses to neuronal infection. Interferon (IFN) is an important component of the innate response, and IFN regulatory factor (IRF) 7 is an inducible transcription factor for the synthesis of IFN-α. IRF7-deficient mice develop fatal paralysis after CNS infection with Sindbis virus, while wild-type mice recover. Irf7 -/- mice produce low levels of IFN-α but high levels of IFN-ß with induction of IFN-stimulated genes, so the reason for this difference is not understood. The current study shows that Irf7 -/- mice developed inflammation earlier but failed to clear virus from motor neuron-rich regions of the brainstem and spinal cord. Levels of IFN-γ and virus-specific antibody were comparable, indicating that IRF7 deficiency does not impair expression of these known viral clearance factors. Therefore, IRF7 is either necessary for the neuronal response to currently identified mediators of clearance or enables the production of additional antiviral factor(s) needed for clearance.


Assuntos
Infecções por Alphavirus , Encefalomielite , Fator Regulador 7 de Interferon , Sindbis virus , Animais , Camundongos , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Tronco Encefálico/virologia , Encefalomielite/imunologia , Encefalomielite/virologia , Inflamação/virologia , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/imunologia , Interferon beta/metabolismo , Neurônios Motores/virologia , Sindbis virus/imunologia , Medula Espinal/virologia
2.
J Gen Virol ; 102(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34661517

RESUMO

Rabies is a zoonotic disease caused by the rabies virus (RABV). RABV can lead to fatal encephalitis and is still a serious threat in most parts of the world. Interferon regulatory factor 7 (IRF7) is the main transcriptional regulator of type I IFN, and it is crucial for the induction of IFNα/ß and the type I IFN-dependent immune response. In this study, we focused on the role of IRF7 in the pathogenicity and immunogenicity of RABV using an IRF7-/- mouse model. The results showed that the absence of IRF7 made mice more susceptible to RABV, because IRF7 restricted the replication of RABV in the early stage of infection. IRF7 deficiency affected the recruitment of plasmacytoid dendritic cells to the draining lymph nodes (dLNs), reduced the production of type I IFN and expression of IFN-stimulated genes. Furthermore, we found that the ability to produce specific RABV-neutralizing antibody was impaired in IRF7-/- mice. Consistently, IRF7 deficiency affected the recruitment of germinal-centre B cells to dLNs, and the generation of plasma cells and RABV-specific antibody secreting cells. Moreover, the absence of IRF7 downregulated the induction of IFN-γ and reduced type 1 T helper cell (Th1)-dependent antibody production. Collectively, our findings demonstrate that IRF7 promotes humoral immune responses and compromises the pathogenicity of RABV in a mouse model.


Assuntos
Fator Regulador 7 de Interferon/fisiologia , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Raiva/imunologia , Raiva/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Imunidade Humoral , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Interferons/análise , Linfonodos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vacina Antirrábica/imunologia , Células Th1/imunologia , Carga Viral
3.
Science ; 370(6515)2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-32972995

RESUMO

Clinical outcome upon infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ranges from silent infection to lethal coronavirus disease 2019 (COVID-19). We have found an enrichment in rare variants predicted to be loss-of-function (LOF) at the 13 human loci known to govern Toll-like receptor 3 (TLR3)- and interferon regulatory factor 7 (IRF7)-dependent type I interferon (IFN) immunity to influenza virus in 659 patients with life-threatening COVID-19 pneumonia relative to 534 subjects with asymptomatic or benign infection. By testing these and other rare variants at these 13 loci, we experimentally defined LOF variants underlying autosomal-recessive or autosomal-dominant deficiencies in 23 patients (3.5%) 17 to 77 years of age. We show that human fibroblasts with mutations affecting this circuit are vulnerable to SARS-CoV-2. Inborn errors of TLR3- and IRF7-dependent type I IFN immunity can underlie life-threatening COVID-19 pneumonia in patients with no prior severe infection.


Assuntos
Infecções por Coronavirus/genética , Infecções por Coronavirus/imunologia , Interferon Tipo I/imunologia , Mutação com Perda de Função , Pneumonia Viral/genética , Pneumonia Viral/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Infecções Assintomáticas , Betacoronavirus , COVID-19 , Criança , Pré-Escolar , Feminino , Loci Gênicos , Predisposição Genética para Doença , Humanos , Lactente , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Masculino , Pessoa de Meia-Idade , Pandemias , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , SARS-CoV-2 , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética , Adulto Jovem
4.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31619554

RESUMO

Type I interferons (IFNs) are key mediators of the innate immune response. Although members of this family of cytokines signal through a single shared receptor, biochemical and functional variation exists in response to different IFN subtypes. While previous work has demonstrated that type I IFNs are essential to control infection by chikungunya virus (CHIKV), a globally emerging alphavirus, the contributions of individual IFN subtypes remain undefined. To address this question, we evaluated CHIKV pathogenesis in mice lacking IFN-ß (IFN-ß knockout [IFN-ß-KO] mice or mice treated with an IFN-ß-blocking antibody) or IFN-α (IFN regulatory factor 7 knockout [IRF7-KO] mice or mice treated with a pan-IFN-α-blocking antibody). Mice lacking either IFN-α or IFN-ß developed severe clinical disease following infection with CHIKV, with a marked increase in foot swelling compared to wild-type mice. Virological analysis revealed that mice lacking IFN-α sustained elevated infection in the infected ankle and in distant tissues. In contrast, IFN-ß-KO mice displayed minimal differences in viral burdens within the ankle or at distal sites and instead had an altered cellular immune response. Mice lacking IFN-ß had increased neutrophil infiltration into musculoskeletal tissues, and depletion of neutrophils in IFN-ß-KO but not IRF7-KO mice mitigated musculoskeletal disease caused by CHIKV. Our findings suggest disparate roles for the IFN subtypes during CHIKV infection, with IFN-α limiting early viral replication and dissemination and IFN-ß modulating neutrophil-mediated inflammation.IMPORTANCE Type I interferons (IFNs) possess a range of biological activity and protect against a number of viruses, including alphaviruses. Despite signaling through a shared receptor, there are established biochemical and functional differences among the IFN subtypes. The significance of our research is in demonstrating that IFN-α and IFN-ß both have protective roles during acute chikungunya virus (CHIKV) infection but do so by distinct mechanisms. IFN-α limits CHIKV replication and dissemination, whereas IFN-ß protects from CHIKV pathogenesis by limiting inflammation mediated by neutrophils. Our findings support the premise that the IFN subtypes have distinct biological activities in the antiviral response.


Assuntos
Febre de Chikungunya/genética , Vírus Chikungunya/patogenicidade , Fator Regulador 7 de Interferon/genética , Interferon-alfa/genética , Interferon beta/genética , Neutrófilos/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Osso e Ossos/imunologia , Osso e Ossos/patologia , Osso e Ossos/virologia , Febre de Chikungunya/imunologia , Febre de Chikungunya/patologia , Febre de Chikungunya/virologia , Vírus Chikungunya/imunologia , Feminino , Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Inflamação , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/imunologia , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/deficiência , Interferon-alfa/imunologia , Interferon beta/antagonistas & inibidores , Interferon beta/deficiência , Interferon beta/imunologia , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Músculo Esquelético/virologia , Infiltração de Neutrófilos , Neutrófilos/patologia , Neutrófilos/virologia , Tarso Animal/imunologia , Tarso Animal/patologia , Tarso Animal/virologia , Replicação Viral
5.
J Gen Virol ; 100(1): 46-62, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30451651

RESUMO

Interferon (IFN) regulatory factors (IRFs) are important determinants of the innate response to infection. We evaluated the role(s) of combined and individual IRF deficiencies in the outcome of infection of C57BL/6 mice with Sindbis virus, an alphavirus that infects neurons and causes encephalomyelitis. The brain and spinal cord levels of Irf7, but not Irf3 mRNAs, were increased after infection. IRF3/5/7-/- and IRF3/7-/- mice died within 3-4 days with uncontrolled virus replication, similar to IFNα receptor-deficient mice, while all wild-type (WT) mice recovered. IRF3-/- and IRF7-/- mice had brain levels of IFNα that were lower, but brain and spinal cord levels of IFNß and IFN-stimulated gene mRNAs that were similar to or higher than WT mice without detectable serum IFN or increases in Ifna or Ifnb mRNAs in the lymph nodes, indicating that the differences in outcome were not due to deficiencies in the central nervous system (CNS) type I IFN response. IRF3-/- mice developed persistent neurological deficits and had more spinal cord inflammation and higher CNS levels of Il1b and Ifnγ mRNAs than WT mice, but all mice survived. IRF7-/- mice died 5-8 days after infection with rapidly progressive paralysis and differed from both WT and IRF3-/- mice in the induction of higher CNS levels of IFNß, tumour necrosis factor (TNF) α and Cxcl13 mRNA, delayed virus clearance and more extensive cell death. Therefore, fatal disease in IRF7-/- mice is likely due to immune-mediated neurotoxicity associated with failure to regulate the production of inflammatory cytokines such as TNFα in the CNS.


Assuntos
Infecções por Alphavirus/fisiopatologia , Encefalomielite/fisiopatologia , Interações Hospedeiro-Patógeno , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Sindbis virus/crescimento & desenvolvimento , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/deficiência , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medula Espinal/patologia , Análise de Sobrevida
6.
Elife ; 72018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29914621

RESUMO

Type I interferon (IFN-I) responses are critical for the control of RNA virus infections, however, many viruses, including Dengue (DENV) and Chikungunya (CHIKV) virus, do not directly activate plasmacytoid dendritic cells (pDCs), robust IFN-I producing cells. Herein, we demonstrated that DENV and CHIKV infected cells are sensed by pDCs, indirectly, resulting in selective IRF7 activation and IFN-I production, in the absence of other inflammatory cytokine responses. To elucidate pDC immunomodulatory functions, we developed a mouse model in which IRF7 signaling is restricted to pDC. Despite undetectable levels of IFN-I protein, pDC-restricted IRF7 signaling controlled both viruses and was sufficient to protect mice from lethal CHIKV infection. Early pDC IRF7-signaling resulted in amplification of downstream antiviral responses, including an accelerated natural killer (NK) cell-mediated type II IFN response. These studies revealed the dominant, yet indirect role of pDC IRF7-signaling in directing both type I and II IFN responses during arbovirus infections.


Assuntos
Febre de Chikungunya/imunologia , Dengue/imunologia , Interações Hospedeiro-Patógeno/imunologia , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/imunologia , Animais , Febre de Chikungunya/genética , Febre de Chikungunya/mortalidade , Febre de Chikungunya/patologia , Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Células Dendríticas/imunologia , Células Dendríticas/virologia , Dengue/genética , Dengue/mortalidade , Dengue/patologia , Vírus da Dengue/crescimento & desenvolvimento , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/genética , Interferon gama/genética , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Viral/antagonistas & inibidores , RNA Viral/genética , RNA Viral/imunologia , Transdução de Sinais , Baço/imunologia , Baço/virologia , Análise de Sobrevida
7.
PLoS Pathog ; 13(12): e1006748, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29244871

RESUMO

Chikungunya virus (CHIKV) and Ross River virus (RRV) are mosquito-transmitted alphaviruses that cause debilitating acute and chronic musculoskeletal disease. Monocytes are implicated in the pathogenesis of these infections; however, their specific roles are not well defined. To investigate the role of inflammatory Ly6ChiCCR2+ monocytes in alphavirus pathogenesis, we used CCR2-DTR transgenic mice, enabling depletion of these cells by administration of diptheria toxin (DT). DT-treated CCR2-DTR mice displayed more severe disease following CHIKV and RRV infection and had fewer Ly6Chi monocytes and NK cells in circulation and muscle tissue compared with DT-treated WT mice. Furthermore, depletion of CCR2+ or Gr1+ cells, but not NK cells or neutrophils alone, restored virulence and increased viral loads in mice infected with an RRV strain encoding attenuating mutations in nsP1 to levels detected in monocyte-depleted mice infected with fully virulent RRV. Disease severity and viral loads also were increased in DT-treated CCR2-DTR+;Rag1-/- mice infected with the nsP1 mutant virus, confirming that these effects are independent of adaptive immunity. Monocytes and macrophages sorted from muscle tissue of RRV-infected mice were viral RNA positive and had elevated expression of Irf7, and co-culture of Ly6Chi monocytes with RRV-infected cells resulted in induction of type I IFN gene expression in monocytes that was Irf3;Irf7 and Mavs-dependent. Consistent with these data, viral loads of the attenuated nsP1 mutant virus were equivalent to those of WT RRV in Mavs-/- mice. Finally, reconstitution of Irf3-/-;Irf7-/- mice with CCR2-DTR bone marrow rescued mice from severe infection, and this effect was reversed by depletion of CCR2+ cells, indicating that CCR2+ hematopoietic cells are capable of inducing an antiviral response. Collectively, these data suggest that MAVS-dependent production of type I IFN by monocytes is critical for control of acute alphavirus infection and that determinants in nsP1, the viral RNA capping protein, counteract this response.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Monócitos/imunologia , Monócitos/virologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Antígenos Ly/metabolismo , Vírus Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Toxina Diftérica/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/imunologia , Humanos , Inflamação/virologia , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , Receptores CCR2/genética , Receptores CCR2/metabolismo , Ross River virus/genética , Ross River virus/imunologia , Ross River virus/patogenicidade , Carga Viral , Virulência/genética , Virulência/imunologia
8.
PLoS One ; 11(5): e0155243, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27182740

RESUMO

Chikungunya virus is an arbovirus spread predominantly by Aedes aegypti and Ae. albopictus mosquitoes, and causes debilitating arthralgia and arthritis. While these are common manifestations during acute infection and it has been suggested they can recur in patients chronically, gaps in knowledge regarding the pathogenesis still exist. Two established mouse models were utilized (adult IRF 3/7 -/- -/- and wild-type C57BL/6J mice) to evaluate disease manifestations in bones and joints at various timepoints. Novel lesions in C57BL/6J mice consisted of periostitis (91%) and foci of cartilage of necrosis (50% of mice at 21 DPI). Additionally, at 21 DPI, 50% and 75% of mice exhibited periosteal bone proliferation affecting the metatarsal bones, apparent via histology and µCT, respectively. µCT analysis did not reveal any alterations in trabecular bone volume measurements in C57BL/6J mice. Novel lesions demonstrated in IRF 3/7 -/- -/- mice at 5 DPI included focal regions of cartilage necrosis (20%), periosteal necrosis (66%), and multifocal ischemic bone marrow necrosis (100%). Contralateral feet in 100% of mice of both strains had similar, though milder lesions. Additionally, comparison of control IRF 3/7 -/- -/- and wild-type C57BL/6J mice demonstrated differences in cortical bone. These experiments demonstrate novel manifestations of disease similar to those occurring in humans, adding insight into disease pathogenesis, and representing new potential targets for therapeutic interventions. Additionally, results demonstrate the utility of µCT in studies of bone and joint pathology and illustrate differences in bone dynamics between mouse strains.


Assuntos
Doenças Ósseas/etiologia , Doenças Ósseas/patologia , Febre de Chikungunya/complicações , Febre de Chikungunya/virologia , Vírus Chikungunya , Artropatias/etiologia , Artropatias/patologia , Animais , Biópsia , Doenças Ósseas/diagnóstico , Modelos Animais de Doenças , Progressão da Doença , Feminino , Inflamação/etiologia , Inflamação/patologia , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/deficiência , Artropatias/diagnóstico , Masculino , Camundongos , Camundongos Knockout , Necrose/etiologia , Necrose/patologia , Fenótipo , Microtomografia por Raio-X
9.
Curr Opin Immunol ; 38: 109-20, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26761402

RESUMO

Influenza viruses cause mild to moderate respiratory illness in most people, and only rarely devastating or fatal infections. The virulence factors encoded by viral genes can explain seasonal or geographic differences at the population level but are unlikely to account for inter-individual clinical variability. Inherited or acquired immunodeficiencies may thus underlie severe cases of influenza. The crucial role of host genes was first demonstrated by forward genetics in inbred mice, with the identification of interferon (IFN)-α/ß-inducible Mx1 as a canonical influenza susceptibility gene. Reverse genetics has subsequently characterized the in vivo role of other mouse genes involved in IFN-α/ß and -λ immunity. A series of in vitro studies with mouse and human cells have also refined the cell-intrinsic mechanisms of protection against influenza viruses. Population-based human genetic studies have not yet uncovered variants with a significant impact. Interestingly, human primary immunodeficiencies affecting T and B cells were also not found to predispose to severe influenza. Recently however, human IRF7 was shown to be essential for IFN-α/ß- and IFN-λ-dependent protective immunity against primary influenza in vivo, as inferred from a patient with life-threatening influenza revealed to be IRF7-deficient by whole exome sequencing. Next generation sequencing of human exomes and genomes will facilitate the analysis of the human genetic determinism of severe influenza.


Assuntos
Imunidade Inata , Hospedeiro Imunocomprometido , Influenza Humana/imunologia , Fator Regulador 7 de Interferon/imunologia , Proteínas de Resistência a Myxovirus/imunologia , Orthomyxoviridae/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Humanos , Influenza Humana/genética , Influenza Humana/patologia , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Camundongos , Camundongos Transgênicos , Proteínas de Resistência a Myxovirus/deficiência , Proteínas de Resistência a Myxovirus/genética , Orthomyxoviridae/patogenicidade , Transdução de Sinais
10.
PLoS One ; 10(10): e0139481, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26447467

RESUMO

Chikungunya virus (CHIKV) is a reemerging, ordinarily mosquito-transmitted, alphavirus that occasionally produces hemorrhagic manifestations, such as nose bleed and bleeding gums, in human patients. Interferon response factor 3 and 7 deficient (IRF3/7-/-) mice, which are deficient for interferon α/ß responses, reliably develop hemorrhagic manifestations after CHIKV infection. Here we show that infectious virus was present in the oral cavity of CHIKV infected IRF3/7-/- mice, likely due to hemorrhagic lesions in the olfactory epithelium that allow egress of infected blood into the nasal, and subsequently, oral cavities. In addition, IRF3/7-/- mice were more susceptible to infection with CHIKV via intranasal and oral routes, with IRF3/7-/- mice also able to transmit virus mouse-to-mouse without an arthropod vector. Cynomolgus macaques often show bleeding gums after CHIKV infection, and analysis of saliva from several infected monkeys also revealed the presence of viral RNA and infectious virus. Furthermore, saliva samples collected from several acute CHIKV patients with hemorrhagic manifestations were found to contain viral RNA and infectious virus. Oral fluids can therefore be infectious during acute CHIKV infections, likely due to hemorrhagic manifestations in the oral/nasal cavities.


Assuntos
Febre de Chikungunya/patologia , Vírus Chikungunya/genética , Saliva/virologia , Adolescente , Animais , Febre de Chikungunya/transmissão , Febre de Chikungunya/virologia , Vírus Chikungunya/isolamento & purificação , Criança , Modelos Animais de Doenças , Feminino , Haplorrinos , Humanos , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa Olfatória/patologia , Mucosa Olfatória/virologia , RNA Viral/metabolismo , Carga Viral
11.
Glia ; 63(4): 595-610, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25422089

RESUMO

Microglia are generally considered the immune cells of the central nervous system. Recent studies have demonstrated that under specific polarization conditions, microglia develop into two different phenotypes, termed M1-like and M2-like microglia. However, the phenotypic characteristics of M1-like- and M2-like-polarized microglia and the mechanisms that regulate polarization are largely unknown. In this study, we characterized lipopolysaccharide-treated M1-like and IL-4-treated M2-like microglia and investigated the mechanisms that regulate phenotypic switching. The addition of M2-like microglial conditioned medium (CM) to primary neurons resulted in an increase in neurite length when compared with neurons treated with M1-like microglial CM, possibly because of the enhanced secretion of neurotrophic factors by M2-like microglia. M1-like microglia were morphologically characterized by larger soma, whereas M2-like microglia were characterized by long processes. M2-like microglia exhibited greater phagocytic capacity than M1-like microglia. These features switched in response to polarization cues. We found that expression of interferon regulatory factor 7 (IRF7) increased during the M2-like to M1-like switch in microglia in vitro and in vivo. Knockdown of IRF7 using siRNA suppressed the expression of M1 marker mRNA and reduced phosphorylation of STAT1. Our findings suggest that IRF7 signaling may play an important role in microglial polarization switching.


Assuntos
Polaridade Celular/fisiologia , Fator Regulador 7 de Interferon/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Animais , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Fator Regulador 7 de Interferon/deficiência , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/efeitos dos fármacos , Fatores de Crescimento Neural/metabolismo , Neuritos/efeitos dos fármacos , Fenótipo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno , Fator de Transcrição STAT1/genética , Transdução de Sinais/efeitos dos fármacos
12.
PLoS Negl Trop Dis ; 8(6): e2933, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24901990

RESUMO

The new world arenavirus Junín virus (JUNV) is the causative agent of Argentine hemorrhagic fever, a lethal human infectious disease. Adult laboratory mice are generally resistant to peripheral infection by JUNV. The mechanism underlying the mouse resistance to JUNV infection is largely unknown. We have reported that interferon receptor knockout mice succumb to JUNV infection, indicating the critical role of interferon in restricting JUNV infection in mice. Here we report that the pathogenic and vaccine strains of JUNV were highly sensitive to interferon in murine primary cells. Treatment with low concentrations of interferon abrogated viral NP protein expression in murine cells. The replication of both JUNVs was enhanced in IRF3/IRF7 deficient cells. In addition, the vaccine strain of JUNV displayed impaired growth in primary murine cells. Our data suggested a direct and potent role of host interferon response in restricting JUNV replication in mice. The defect in viral growth for vaccine JUNV might also partially explain its attenuation in mice.


Assuntos
Antivirais/farmacologia , Interferons/imunologia , Interferons/farmacologia , Vírus Junin/efeitos dos fármacos , Vírus Junin/imunologia , Animais , Células Cultivadas , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/deficiência , Interferons/deficiência , Vírus Junin/crescimento & desenvolvimento , Vírus Junin/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos
13.
Hypertension ; 63(4): 713-22, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24396025

RESUMO

Cardiac hypertrophy is a complex pathological process that involves multiple factors including inflammation and apoptosis. Interferon regulatory factor 7 (IRF7) is a multifunctional regulator that participates in immune regulation, cell differentiation, apoptosis, and oncogenesis. However, the role of IRF7 in cardiac hypertrophy remains unclear. We performed aortic banding in cardiac-specific IRF7 transgenic mice, IRF7 knockout mice, and the wild-type littermates of these mice. Our results demonstrated that IRF7 was downregulated in aortic banding-induced animal hearts and cardiomyocytes that had been treated with angiotensin II or phenylephrine for 48 hours. Accordingly, heart-specific overexpression of IRF7 significantly attenuated pressure overload-induced cardiac hypertrophy, fibrosis, and dysfunction, whereas loss of IRF7 led to opposite effects. Moreover, IRF7 protected against angiotensin II-induced cardiomyocyte hypertrophy in vitro. Mechanistically, we identified that IRF7-dependent cardioprotection was mediated through IRF7 binding to inhibitor of κB kinase-ß, and subsequent nuclear factor-κB inactivation. In fact, blocking nuclear factor-κB signaling with cardiac-specific inhibitors of κBα(S32A/S36A) super-repressor transgene counteracted the adverse effect of IRF7 deficiency. Conversely, activation of nuclear factor-κB signaling via a cardiac-specific conditional inhibitor of κB kinase-ß(S177E/S181E) (constitutively active) transgene negated the antihypertrophic effect of IRF7 overexpression. Our data demonstrate that IRF7 acts as a novel negative regulator of pathological cardiac hypertrophy by inhibiting nuclear factor-κB signaling and may constitute a potential therapeutic target for pathological cardiac hypertrophy.


Assuntos
Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Fator Regulador 7 de Interferon/fisiologia , Transdução de Sinais/fisiologia , Angiotensina II/farmacologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Hipertrofia/induzido quimicamente , Hipertrofia/patologia , Hipertrofia/fisiopatologia , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/fisiologia , Técnicas In Vitro , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia
14.
Mol Immunol ; 57(2): 100-10, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24096085

RESUMO

Measles virus (MV) infects CD150Tg/Ifnar (IFN alpha receptor)(-/-) mice but not CD150 (a human MV receptor)-transgenic (Tg) mice. We have shown that bone marrow-derived dendritic cells (BMDCs) from CD150Tg/Ifnar(-/-) mice are permissive to MV in contrast to those from simple CD150Tg mice, which reveals a crucial role of type I interferon (IFN) in natural tropism against MV. Yet, the mechanism whereby BMDCs produce initial type I IFN has not been elucidated in MV infection. RNA virus infection usually allows cells to generate double-stranded RNA and induce activation of IFN regulatory factor (IRF) 3/7 transcription factors, leading to the production of type I IFN through the retinoic acid-inducible gene I (RIG-I)/melanoma differentiation-associated gene 5 (MDA5)-mitochondrial antiviral signaling protein (MAVS) pathway. In mouse experimental BMDCs models, we found CD150Tg/Mavs(-/-)BMDCs, but not CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs, permissive to MV. IFN-α/ß were not induced in MV-infected CD150Tg/Mavs(-/-)BMDCs, while IFN-ß was subtly induced in CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs. In vivo systemic infection was therefore established by transfer of MV-infected CD150Tg/Mavs(-/-) BMDCs to CD150Tg/Ifnar(-/-) mice. These data indicate that MAVS-dependent, IRF3/7-independent IFN-ß induction triggers the activation of the IFNAR pathway so as to restrict the spread of MV by infected BMDCs. Hence, MAVS participates in the initial induction of type I IFN in BMDCs and IFNAR protects against MV spreading. We also showed the importance of IL-10-producing CD4(+) T cells induced by MV-infected BMDCs in vitro, which may account for immune modulation due to the functional aberration of DCs.


Assuntos
Células Dendríticas/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/genética , Interferon beta/metabolismo , Sarampo/imunologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Antígenos CD/metabolismo , Células da Medula Óssea/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Chlorocebus aethiops , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Células Dendríticas/citologia , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Interferon beta/biossíntese , Interleucina-10/metabolismo , Vírus do Sarampo/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/imunologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Células Vero
15.
J Immunol ; 191(8): 4194-201, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24043884

RESUMO

We investigated the roles of IFN regulatory factor (IRF)-3 and IRF-7 in innate antiviral immunity against dengue virus (DENV). Double-deficient Irf-3(-/-)7(-/-) mice infected with the DENV2 strain S221 possessed 1,000-150,000 fold higher levels of viral RNA than wild-type and single-deficient mice 24 h postinfection (hpi); however, they remained resistant to lethal infection. IFN-α/ß was induced similarly in wild-type and Irf-3(-/-) mice post-DENV infection, whereas in the Irf-7(-/-) and Irf-3(-/-)7(-/-) mice, significantly low levels of IFN-α/ß expression was observed within 24 hpi. IFN-stimulated gene induction was also delayed in Irf-3(-/-)7(-/-) mice relative to wild-type and single-deficient mice. In particular, Cxcl10 and Ifnα2 were rapidly induced independently of both IRF-3 and IRF-7 in the Irf-3(-/-)7(-/-) mice with DENV infection. Higher levels of serum IFN-γ, IL-6, CXCL10, IL-8, IL-12 p70, and TNF were also observed in Irf-3(-/-)7(-/-) mice 24 hpi, at which time point viral titers peaked and started to be cleared. Ab-mediated blockade experiments revealed that IFN-γ, CXCL10, and CXCR3 function to restrict DENV replication in Irf-3(-/-)7(-/-) mice. Additionally, the IFN-stimulated genes Cxcl10, Ifit1, Ifit3, and Mx2 can be induced via an IRF-3- and IRF-7-independent pathway that does not involve IFN-γ signaling for protection against DENV. Collectively, these results demonstrate that IRF-3 and IRF-7 are redundant, albeit IRF-7 plays a more important role than IRF-3 in inducing the initial IFN-α/ß response; only the combined actions of IRF-3 and IRF-7 are necessary for efficient control of early DENV infection; and the late, IRF-3- and IRF-7-independent pathway contributes to anti-DENV immunity.


Assuntos
Vírus da Dengue/imunologia , Dengue/imunologia , Imunidade Inata , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon-alfa/sangue , Interferon beta/sangue , Proteínas Adaptadoras de Transdução de Sinal , Aedes , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/sangue , Quimiocina CXCL10/imunologia , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Interferon gama/sangue , Interferon gama/imunologia , Interleucina-12/sangue , Interleucina-6/sangue , Interleucina-8/sangue , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Resistência a Myxovirus/metabolismo , Proteínas/metabolismo , RNA Viral/sangue , Proteínas de Ligação a RNA , Receptores CXCR3/imunologia , Transdução de Sinais , Fatores de Necrose Tumoral/sangue , Carga Viral , Replicação Viral/imunologia
16.
Nature ; 490(7420): 421-5, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22982991

RESUMO

Antiviral responses must be tightly regulated to defend rapidly against infection while minimizing inflammatory damage. Type 1 interferons (IFN-I) are crucial mediators of antiviral responses and their transcription is regulated by a variety of transcription factors; principal among these is the family of interferon regulatory factors (IRFs). The IRF gene regulatory networks are complex and contain multiple feedback loops. The tools of systems biology are well suited to elucidate the complex interactions that give rise to precise coordination of the interferon response. Here we have used an unbiased systems approach to predict that a member of the forkhead family of transcription factors, FOXO3, is a negative regulator of a subset of antiviral genes. This prediction was validated using macrophages isolated from Foxo3-null mice. Genome-wide location analysis combined with gene deletion studies identified the Irf7 gene as a critical target of FOXO3. FOXO3 was identified as a negative regulator of Irf7 transcription and we have further demonstrated that FOXO3, IRF7 and IFN-I form a coherent feed-forward regulatory circuit. Our data suggest that the FOXO3-IRF7 regulatory circuit represents a novel mechanism for establishing the requisite set points in the interferon pathway that balances the beneficial effects and deleterious sequelae of the antiviral response.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/imunologia , Inflamação/imunologia , Inflamação/patologia , Fator Regulador 7 de Interferon/metabolismo , Vesiculovirus/imunologia , Animais , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Deleção de Genes , Inflamação/genética , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Reprodutibilidade dos Testes
17.
J Immunol ; 189(6): 2860-8, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22896628

RESUMO

Type I IFNs are induced by pathogens to protect the host from infection and boost the immune response. We have recently demonstrated that this IFN response is not restricted to pathogens, as the Gram-positive bacterium Lactobacillus acidophilus, a natural inhabitant of the intestine, induces high levels of IFN-ß in dendritic cells. In the current study, we investigate the intracellular pathways involved in IFN-ß upon stimulation of dendritic cells with L. acidophilus and reveal that this IFN-ß induction requires phagosomal uptake and processing but bypasses the endosomal receptors TLR7 and TLR9. The IFN-ß production is fully dependent on the TIR adapter molecule MyD88, partly dependent on IFN regulatory factor (IRF)1, but independent of the TIR domain-containing adapter inducing IFN-ß MyD88 adapter-like, IRF and IRF7. However, our results suggest that IRF3 and IRF7 have complementary roles in IFN-ß signaling. The IFN-ß production is strongly impaired by inhibitors of spleen tyrosine kinase (Syk) and PI3K. Our results indicate that L. acidophilus induces IFN-ß independently of the receptors typically used by bacteria, as it requires MyD88, Syk, and PI3K signaling and phagosomal processing to activate IRF1 and IRF3/IRF7 and thereby the release of IFN-ß.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Fator Regulador 1 de Interferon/fisiologia , Fator Regulador 3 de Interferon/fisiologia , Fator Regulador 7 de Interferon/fisiologia , Interferon beta/metabolismo , Lactobacillus acidophilus/imunologia , Fator 88 de Diferenciação Mieloide/fisiologia , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Endossomos/imunologia , Endossomos/metabolismo , Endossomos/microbiologia , Fator Regulador 1 de Interferon/deficiência , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fagossomos/imunologia , Fagossomos/metabolismo , Processamento de Proteína Pós-Traducional/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
18.
J Virol ; 86(18): 9888-98, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761364

RESUMO

Chikungunya virus (CHIKV) infections can produce severe disease and mortality. Here we show that CHIKV infection of adult mice deficient in interferon response factors 3 and 7 (IRF3/7(-/-)) is lethal. Mortality was associated with undetectable levels of alpha/beta interferon (IFN-α/ß) in serum, ∼50- and ∼10-fold increases in levels of IFN-γ and tumor necrosis factor (TNF), respectively, increased virus replication, edema, vasculitis, hemorrhage, fever followed by hypothermia, oliguria, thrombocytopenia, and raised hematocrits. These features are consistent with hemorrhagic shock and were also evident in infected IFN-α/ß receptor-deficient mice. In situ hybridization suggested CHIKV infection of endothelium, fibroblasts, skeletal muscle, mononuclear cells, chondrocytes, and keratinocytes in IRF3/7(-/-) mice; all but the latter two stained positive in wild-type mice. Vaccination protected IRF3/7(-/-) mice, suggesting that defective antibody responses were not responsible for mortality. IPS-1- and TRIF-dependent pathways were primarily responsible for IFN-α/ß induction, with IRF7 being upregulated >100-fold in infected wild-type mice. These studies suggest that inadequate IFN-α/ß responses following virus infection can be sufficient to induce hemorrhagic fever and shock, a finding with implications for understanding severe CHIKV disease and dengue hemorrhagic fever/dengue shock syndrome.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/prevenção & controle , Vírus Chikungunya/patogenicidade , Fator Regulador 3 de Interferon/fisiologia , Fator Regulador 7 de Interferon/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Infecções por Alphavirus/patologia , Animais , Febre de Chikungunya , Vírus Chikungunya/imunologia , Vírus Chikungunya/fisiologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Interferon-alfa/biossíntese , Interferon-alfa/farmacologia , Interferon beta/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/fisiologia , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/fisiologia , Choque Hemorrágico/imunologia , Choque Hemorrágico/prevenção & controle , Replicação Viral/efeitos dos fármacos
19.
J Virol ; 86(13): 7384-92, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22514347

RESUMO

Interferon (IFN) regulatory factors (IRFs) are a family of transcription factors involved in regulating type I IFN genes and other genes participating in the early antiviral host response. To better understand the mechanisms involved in virus-induced central nervous system (CNS) inflammation, we studied the influence of IRF1, -3, -7, and -9 on the transcriptional activity of key genes encoding antiviral host factors in the CNS of mice infected with lymphocytic choriomeningitis virus (LCMV). A key finding is that neither IRF3 nor IRF7 is absolutely required for induction of a type I IFN response in the LCMV-infected CNS, whereas concurrent elimination of both factors markedly reduces the virus-induced host response. This is unlike the situation in the periphery, where deficiency of IRF7 almost eliminates the LCMV-induced production of the type I IFNs. This difference is seemingly related to the local environment, as peripheral production of type I IFNs is severely reduced in intracerebrally (i.c.) infected IRF7-deficient mice, which undergo a combined infection of the CNS and peripheral organs, such as spleen and lymph nodes. Interestingly, despite the redundancy of IRF7 in initiating the type I IFN response in the CNS, the response is not abolished in IFN-ß-deficient mice, as might have been expected. Collectively, these data demonstrate that the early type I IFN response to LCMV infection in the CNS is controlled by a concerted action of IRF3 and -7. Consequently this work provides strong evidence for differential regulation of the type I IFN response in the CNS versus the periphery during viral infection.


Assuntos
Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/biossíntese , Vírus da Coriomeningite Linfocítica/imunologia , Sistema Nervoso/imunologia , Sistema Nervoso/virologia , Animais , Feminino , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
20.
Stroke ; 43(5): 1383-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22403050

RESUMO

BACKGROUND AND PURPOSE: Systemic administration of Toll-like receptor (TLR) 4 and TLR9 agonists before cerebral ischemia have been shown to reduce ischemic injury by reprogramming the response of the brain to stroke. Our goal was to explore the mechanism of TLR-induced neuroprotection by determining whether a TLR7 agonist also protects against stroke injury. METHODS: C57Bl/6, TNF(-/-), interferon (IFN) regulatory factor 7(-/-), or type I IFN receptor (IFNAR)(-/-) mice were subcutaneously administered the TLR7 agonist Gardiquimod (GDQ) 72 hours before middle cerebral artery occlusion. Infarct volume and functional outcome were determined after reperfusion. Plasma cytokine responses and induction of mRNA for IFN-related genes in the brain were measured. IFNAR(-/-) mice also were treated with the TLR4 agonist (lipopolysaccharide) or the TLR9 agonist before middle cerebral artery occlusion and infarct volumes measured. RESULTS: The results show that GDQ reduces infarct volume as well as functional deficits in mice. GDQ pretreatment provided robust neuroprotection in TNF(-/-) mice, indicating that TNF was not essential. GDQ induced a significant increase in plasma IFNα levels and both IRF7(-/-) and IFNAR(-/-) mice failed to be protected, implicating a role for IFN signaling in TLR7-mediated protection. CONCLUSIONS: Our studies provide the first evidence that TLR7 preconditioning can mediate neuroprotection against ischemic injury. Moreover, we show that the mechanism of protection is unique from other TLR preconditioning ligands in that it is independent of TNF and dependent on IFNAR.


Assuntos
Aminoquinolinas/uso terapêutico , Encéfalo/irrigação sanguínea , Imidazóis/uso terapêutico , Precondicionamento Isquêmico/métodos , Glicoproteínas de Membrana/agonistas , Fármacos Neuroprotetores/uso terapêutico , Receptor de Interferon alfa e beta/fisiologia , Acidente Vascular Cerebral/prevenção & controle , Receptor 7 Toll-Like/agonistas , Animais , Infarto Encefálico/patologia , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA